Functional Neuropeptidomics in the Decapod Crustacean: Method
Transcription
Functional Neuropeptidomics in the Decapod Crustacean: Method
Functional Neuropeptidomics in the Decapod Crustacean: Method Development and Application to Behavioral Neuroscience Research By Claire Margaret Schmerberg A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy (Pharmaceutical Sciences) at the UNIVERSITY OF WISCONSIN-MADISON 2012 Date of final oral examination: 12/06/12 The dissertation is approved by the following members of the Final Oral Committee: Lingjun Li, Professor, Pharmaceutical Sciences and Chemistry Ronald R. Burnette, Professor and Division Chair, Pharmaceutical Sciences Ei Terasawa, Professor, Pediatrics Craig S. Atwood, Associate Professor, Medicine Arash Bashirullah, Assistant Professor, Pharmaceutical Sciences ©Copyright by Claire M. Schmerberg 2012 All Rights Reserved i Acknowledgements I would first like to acknowledge my advisor Lingjun Li, without whom this work would not be possible. Beyond the initial concepts underlying this work, she has given me the freedom and support to become an independent scientist. Dr. Li is an outstanding scientist and mentor who has developed a world-class research program. The creativity and excellence of work in her lab is without comparison. In the tradition of legendary scientists, Dr. Li allows curiosity to direct her research program and displays an incredible tenacity to overcome obstacles in research. This dissertation work directly reflects these qualities: several of the techniques are challenging to implement to the point that no others have tried them before, and I was able to investigate fundamental neurobiological concepts in an animal model that is not typically considered to display such complex behaviors. Dr. Li gave me the freedom to ask the scientific questions I found interesting, and the support to persevere in challenging situations. She also has always been supportive in endeavors to improve my professional skills and academic profile. It has been an honor to work with Dr. Li and I will forever be thankful for this opportunity. The members of my thesis committee also provided invaluable support and insights throughout the dissertation preparation process. Their patience and constructive criticism have raised the academic caliber of this work substantially. I am also very grateful for their support and regard for my future academic career. I am very thankful to Dr. Ei Terasawa, Dr. Arash Bashirullah, Dr. Ron Burnette, and Dr. Craig Atwood. Several other people have contributed directly to this work. This includes my labmates Zhidan Liang and Nicole Woodards; former labmate Dr. Ruibing Chen; several undergraduates who have worked with me: Andrew Kozicki, Lauren Putterman, and Kevin Hayes; and Alex Laztka of the Vander Zanden Lab. I also received helpful input on these projects from Dr. ii Brendan Walker of Washington State University, Kirk J. Grubbs of the Currie Lab, my labmate Dr. Chenxi Jia, and former labmates Dr. Xiaoyue Jiang, Dr. Robert Cunningham, and Dr. Heidi Behrens. The UW-Madison Writing Center Mellon-Wisconsin Dissertation Camp was instrumental in preparing this document and in my professional development. Projects related to this work but not included in it were aided by a former labmate, Dr. Weifeng Cao, and undergraduate students: Katherine Zimny and Gajan Muthuvel. The Trout Lake Research Station from the Department of Limnology, UW-Madison, and Lindsay Sargent of the University of Notre Dame Environmental Research Center were of great assistance. The Analytical Instrumentation Center of the School of Pharmacy and the Biotechnology Center provided equipment. Constructive criticism and support on this work was given by all members of the Li lab during my tenure here, including several already mentioned and Hui “Vivian” Ye, Dr. Di Ma, Dr. Robert Sturm, and Zichuan Zhang. Other people in the department who have provided criticism and support include Gary Girdaukas, Dr. Richard Peterson, and Dr. Steve Oakes. I am also indebted to the people who first encouraged me to pursue a career in science. My first research advisors, Dr. So-Hye Cho in the lab of Dr. SonBinh Nguyen and Dr. Bradley Cooke in the lab of Dr. Catherine Woolley at Northwestern University provided great support to me in my undergraduate research. Throughout my graduate career I was supported financially by a variety of mechanisms for which I am incredibly grateful. These include the School of Pharmacy Kenneth Connors Wisconsin Distinguished Graduate Fellowship, the Biotechnology Training Program at UWMadison (NIH training grant 5 T32GM08349), the Vilas Conference Presentation Fund, and the American Chemical Society Women Chemists’ Committee Eli Lilly Travel Award. I have also iii been supported on the following grants to Lingjun Li: NIH grant R01DK071801, and NSF grant CHE-0967784. I have been incredibly lucky to have great emotional support, which is mentioned last but may be the most important factor in completing this dissertation. My boyfriend, Kirk Grubbs, and my parents, Fred and Nancy Schmerberg, have sustained me in the most challenging times during my graduate career. At times I was convinced I could not complete this; but Kirk’s and my parents’ belief that I could kept me from quitting. I also owe a debt of gratitude to other friends and family members, including my brother Luke and my friends Nicole, Alissa, Charlie, and Garret. Needed breaks from the mental strain were provided by the Dane County Humane Society, Eagle Heights Community Gardens, and my pets. I am also grateful to my grandmother, Betty Robinson Gingras Knapp, who supported me unconditionally and created a family culture that places a great value on higher education. She was a woman of outstanding courage, integrity, and compassion. She passed away shortly before I was able to complete this work, but I know that she would be proud of my achievements. iv Table of Contents Page Part I. Introductory Information Acknowledgements Table of Contents Abstract Chapter 1. Introduction and Overall Summary i iv v 1 Part II. Background Information Chapter 2: Neuropeptide Discovery in the Decapod Crustacean Chapter 3: Methods for Function-Driven Discovery and Functional Assessment of Neuropeptides 8 123 Part III. Development of Tools and Methodology for Functional Neuropeptidomics Studies Chapter 4. Affinity-Enhanced Microdialysis for Increased Neuropeptide Recovery Chapter 5. Mapping the Neuropeptidome of the Species Orconectes rusticus Chapter 6. Development of Methods for Neuropeptide Quantitation with DataIndependent MS/MS 175 207 250 Part IV. Application of Tools and Methodology in Behavioral Experiments Chapter 7. Application of Microdialysis and Data-Independent MS/MS Quantitation for Identification of Neuropeptides Involved in Feeding Behavior Chapter 8. Quantification of Neuropeptides Altered in Acute and Long-Term Ethanol Exposure 272 317 Part V. Conclusions and Supplemental Information Chapter 9. Conclusions and Future Directions 339 Appendices A. Protocols for Animal Care, Sample Collection and Preparation, Instrumental Analysis, and Results Interpretation B. Crustacean Neuropeptide Database C. Neuropeptides and Proteins Identified in Orconectes rusticus 344 387 419 v Functional Neuropeptidomics in the Decapod Crustacean: Method Development and Application to Behavioral Neuroscience Research Claire Margaret Schmerberg Under the supervision of Professor Lingjun Li At the University of Wisconsin-Madison Abstract Neuropeptides (NPs) represent an important class of signaling molecules whose identities and functions are not yet fully understood, due mostly to difficulties in studying them using traditional biochemical methods. These molecules have been implicated in a variety of processes, including the generation of feeding-related and motivated behaviors, as well as adaptation to environmental changes. In this work, analytical chemistry methods were developed and applied to the decapod crustacean (DC), a simple model animal, toward the end of studying timeresolved changes in NPs during behavior. First, new tools and methods for function-driven neuropeptidomics—an approach to discover NPs while concurrently gaining information about their function—were developed. The neuropeptidome of Orconectes rusticus, a DC used in behavioral neuroscience and a potential model animal for studying environment-neuroendocrine interactions, was described. Affinityenhanced microdialysis was explored to improve sampling of NPs concurrent with behavior. A new strategy for mass spectrometry data collection that enables simultaneous quantification and identification (MSE) was developed and applied for the first time to NPs. Proof-of-principle experiments validated this technique for quantifying NPs that changed after feeding in tissue extracts of the blue crab, Callinectes sapidus. These tools were then applied to study dynamic changes in NPs in the Jonah crab, Cancer borealis, under conditions related to feeding and in the vi rock crab, Cancer irroratus, related to motivated behavior. Time-resolved changes in NPs during feeding in C. borealis were characterized using microdialysis and non-targeted quantitative MSE. This permitted a close correlation between behavioral and neurochemical changes, allowing putative assignment of roles for several NPs in food-related reward and ingestive behavior. Changes in NPs in C. irroratus as a result of acute and repeated exposure to a human drug of abuse, ethanol, were also characterized. This provides insight into neurochemical changes after long-term drug exposure, an important factor in the pathology of drug addiction, which is thought to be a disorder of the natural reward system. This work not only improves analytical chemistry tools for research into NP identification and functional determination, but also provides evidence for the potential roles of several NPs in feeding and motivation-related behaviors. Tools developed in this research will be useful for behavioral neuroscience and studies of neuroendocrine regulation of environmental adaptation. This work represents an important step in understanding the basic neurochemistry of reward and feeding and thus has relevance to basic science as well as human disease. 1 Chapter 1: Introduction and Overall Summary 1.1 Introduction The overall focus of the work presented in this dissertation is the development of cuttingedge analytical chemistry tools for use in neuroscience research, and the application of some of these tools to study dynamic neuroendocrine processes. It encompasses several areas of research, both in development and application. Physical experimental tools, a database, and a novel process for data analysis are developed. For application, feeding behavior and adaptation to repeated ethanol exposure are studied. The document is divided into five main sections: introductory information, background information, method development projects, application projects, and conclusions and supplemental information. In this work, I have striven to improve upon the promising tools offered by advanced analytical chemistry techniques in order to make them useful for relevant research into the neuroendocrine correlates of behavior, and have demonstrated the application of these tools in this field. The introductory information (Table of Contents, Abstract, and Chapter 1) serves as a general introduction to this dissertation work and briefly describes each project. 1.2. Background Information A main focus of this project is the study of neuropeptides (NPs), which are an important class of neural signaling molecules. Chapter 2 describes these molecules and some methods for their analysis, and catalogs the known crustacean neuropeptide families with putative functions. These compounds have many important functions and can act both within the brain and without; serving as a way for the brain and body to communicate information and coordinate activity. Although much is known about NPs in mammals, this work focuses on their function in decapod crustaceans. The simplicity of the crustacean nervous system, both in terms of the number and 2 types of compounds present, and in terms of the number and connectivity of the neurons present, is the main factor in choosing this model organism [1, 2]. The mammalian brain is more chemically complex due to the presence of additional proteins and peptide fragments of proteins that occur in vivo and immediately post-mortem [3]. The neuronal substrate is also much simpler and more extensively studied. Compared to the mammalian brain, which has on the order of 109 neurons, the crustacean nervous system has only thousands, and many in the famous centralpattern generating networks of several ganglia are identifiable and constant in properties and location from one individual to another [4, 5]. An extensive suite of useful analytical tools based on mass spectrometry (MS) and tandem MS (MS/MS) techniques has been developed for analysis of NPs in crustaceans [6], and many NPs have been studied, both for sequence information and functional information [7]. MS is a highly useful technique for studying NPs due to its high specificity and ability to rapidly characterize peptide sequences from small amounts of sample [6]. Chapter 2 enumerates many of the techniques that have been developed to this end and provides a list of known NPs and their functions. A new approach for discovery of new NPs, termed function-driven discovery, is discussed in Chapter 3. This tactic calls for experimental manipulation to elicit changes in NPs so that functional information is gleaned along with identity information [8]. Function-driven discovery relies heavily upon quantitative mass spectrometry methods, and a number of these are described in detail [9]. It also relies upon novel sampling methods, including microdialysis, which is capable of collecting samples from an animal concurrent with it performing a behavior. This permits the highest degree of correlation possible between changes in neurochemistry and changes in behavior short of NP administration [6, 10]. Additional methods for determining NP function, including those that do not inherently rely upon MS, are also described. This chapter 3 describes methods and approaches for using MS to obtain biologically relevant information along with identification of NPs. 1.3. Development of Tools and Methodology for Functional Neuropeptidomics Studies In this section, novel tools for NP analysis with functional relevance are developed. A method to increase the amount of NPs collected with microdialysis, termed affinity-enhanced microdialysis (AE-MD) is described in Chapter 4. This technique solves some of the challenges outlined in Ch. 3 in terms of MS quantification sensitivity to enable MD to sample NPs on a neurobiologically relevant time scale. Microdialysis relies upon diffusion of analytes into a probe that is surgically implanted into the animal. In the probe, a dialysis membrane is in contact with the extracellular space, and liquid flowing through the probe carries molecules that have diffused through that membrane out a length of tubing to where they can be collected and analyzed. AEMD relies upon increasing the rate of diffusion into the probe by adding solid affinity agents that act as “sinks” for analyte that has diffused into the probe. Thus, the driving force is increased and additional analyte can be collected [11]. This technique makes quantification of NPs possible with an experimental setup and condition that was not previously capable of doing so. Another experimental tool is developed in Chapter 5, a list of the known neuropeptides in a decapod species of particular interest, the crayfish Orconectes rusticus. This species is used extensively in behavioral neuroscience and ecology research. It is a model species for studying aggressive behavior [12-14], motivation-related drug-conditioned place preference behavior [1519], and learning and memory [20-24]. In ecology, it is useful for understanding the properties that make a species a successful invader [25-29]. Two methods for semi-automated analysis of MS/MS data to identify NPs are compared, with implications for other neuropeptidomics studies. 4 Characterizing the neuropeptidome of this species will enable further research using this useful model animal. In Chapter 6, a data analysis approach for obtaining quantitative and identifying information simultaneously based on a recent advance in MS/MS technology is developed. This technique uses data-independent acquisition (DIA) MS/MS, which has previously been demonstrated for quantitation and identification in proteomics studies, where multiple peptides per protein are obtained [30-34]. This technique is employed with an open-source, vendorneutral software typically used for multiple reaction monitoring (MRM) MS/MS quantification called Skyline [35]. Skyline is used to generate pseudo-MRM analysis of potentially all analytes in the sample. The linearity of this quantitation technique is demonstrated and it is applied to a real sample for comparison to results previously obtained with a different method. This chapter draws heavily on the concepts of Ch. 3, as DIA MS/MS quantitation is a highly useful tool for function-driven discovery. In this and other method development chapters, techniques for NP analysis, ranging from physical tools to data processing methods, are developed for use in behavioral neuroscience studies. 1.4. Application of Tools and Methodology in Behavioral Experiments Following development of MS/MS-based tools for NP analysis, these tools are applied to study two conditions of interest in behavioral neuroscience. In Chapter 7, microdialysis (MD) is used to sample NPs from the hemolymph of Jonah crabs, Cancer borealis, during feeding behavior. This process is known to be regulated heavily by NPs in mammals [36], but studying it in the crustacean may allow for dissection of multiple neural and chemical pathways that overlap heavily in mammals. This chapter also uses the quantitation technique developed in Ch. 6 for simultaneous quantification and sequence verification. A total of 28 NPs were quantified, and 5 5 were observed to have statistically significant changes throughout the course of feeding. This chapter demonstrates the applicability of MD and DIA MS/MS quantitation in a behavioral neuroscience experimental context. A final application is conducted in Chapter 8, in which NPs in the rock crab, Cancer irroratus, that change after acute and repeated exposure to ethanol, a common human drug of abuse, are characterized. It is expected that repeated exposure will lead to adaptations in the nervous system and its neurochemistry that can be hallmarks of the pathology of substance dependence or other disorders involving motivation. This chapter takes an untargeted approach using DIA MS/MS quantitation as developed in Ch. 6, and identifies 22 NPs that may be potential markers of changes in the neuroendocrine system that are related to long-term drug exposure. A novel assay for alcohol content in crab hemolymph is also developed. 1.5. Conclusions and Supplemental Information The final section of this dissertation contains conclusions and potential future directions that related work could take. It also contains several appendices, including Protocols, a database of crab NPs developed in Ch. 5 that is applicable for future NP discovery experiments, a comprehensive list of results from that chapter. These sections are intended to aid future work in this area, both by listing the procedures developed and improved in this thesis and by tabulating data that can be used for data processing. The work presented here will provide useful methods and experimental models for future research into NP identity and function. 1.6. Works Cited 1. Li, L.; Sweedler, J. V. Peptides in the brain: mass spectrometry-based measurement approaches and challenges. Annu Rev Anal Chem (Palo Alto Calif). 2008, 1, 451-483. 2. Chen, R.; Li, L. Mass spectral imaging and profiling of neuropeptides at the organ and cellular domains. Anal Bioanal Chem. 2010, 397, 3185-3193. 3. Dowell, J. A.; Heyden, W. V.; Li, L. Rat neuropeptidomics by LC-MS/MS and MALDI-FTMS: Enhanced dissection and extraction techniques coupled with 2D RP-RP HPLC. J Proteome Res. 2006, 5, 3368-3375. 4. Hooper, S. L.; DiCaprio, R. A. Crustacean Motor Pattern Generator Networks. Neurosignals. 2004, 13, 50-69. 6 5. Marder, E.; Bucher, D. Understanding Circuit Dynamics Using the Stomatogastric Nervous System of Lobsters and Crabs. Annual Review of Physiology. 2007, 69, 291-316. 6. Li, L. J.; Sweedler, J. V. Peptides in the Brain: Mass Spectrometry-Based Measurement Approaches and Challenges. Annu Rev Anal Chem. 2008, 1, 451-483. 7. Christie, A. E.; Stemmler, E. A.; Dickinson, P. S. Crustacean neuropeptides. Cell. Mol. Life Sci. 2010, 67, 41354169. 8. Schmerberg, C. M.; Li, L. Function-Driven Discovery of Neuropeptides with Mass Spectrometry-Based Tools. Protein Pept Lett. 2012. 9. Bantscheff, M.; Lemeer, S.; Savitski, M. M.; Kuster, B. Quantitative mass spectrometry in proteomics: critical review update from 2007 to the present. Anal. Bioanal. Chem. 2012, 404, 939-965. 10. Lee, G. J.; Park, J. H.; Park, H. K. Microdialysis applications in neuroscience. Neurol Res. 2008, 30, 661-668. 11. Duo, J.; Fletcher, H.; Stenken, J. A. Natural and synthetic affinity agents as microdialysis sampling mass transport enhancers: current progress and future perspectives. Biosens Bioelectron. 2006, 22, 449-457. 12. Tierney, A. J.; Greenlaw, M. A.; Dams-O'Connor, K.; Aig, S. D.; Perna, A. M. Behavioral effects of serotonin and serotonin agonists in two crayfish species, Procambarus clarkii and Orconectes rusticus. Comp Biochem Physiol A Mol Integr Physiol. 2004, 139, 495-502. 13. Tierney, A. J.; Kim, T.; Abrams, R. Dopamine in crayfish and other crustaceans: distribution in the central nervous system and physiological functions. Microsc Res Tech. 2003, 60, 325-335. 14. Panksepp, J. B.; Huber, R. Chronic alterations in serotonin function: dynamic neurochemical properties in agonistic behavior of the crayfish, Orconectes rusticus. J Neurobiol. 2002, 50, 276-290. 15. Dziopa, L.; Imeh-Nathaniel, A.; Baier, D.; Kiel, M.; Sameera, S.; Brager, A.; Beatriz, V.; Nathaniel, T. I. Morphine-conditioned cue alters c-Fos protein expression in the brain of crayfish. Brain Research Bulletin. 2011, 85, 385-395. 16. Huber, R. Amines and motivated behaviors: a simpler systems approach to complex behavioral phenomena. Journal of Comparative Physiology a-Neuroethology Sensory Neural and Behavioral Physiology. 2005, 191, 231-239. 17. Nathaniel, T. I.; Huber, R.; Panksepp, J. Repeated cocaine treatments induce distinct locomotor effects in Crayfish. Brain Research Bulletin. 2012, 87, 328-333. 18. Nathaniel, T. I.; Panksepp, J.; Huber, R. Drug-seeking behavior in an invertebrate system: Evidence of morphine-induced reward, extinction and reinstatement in crayfish. Behavioural Brain Research. 2009, 197, 331-338. 19. Panksepp, J. B.; Huber, R. Ethological analyses of crayfish behavior: a new invertebrate system for measuring the rewarding properties of psychostimulants. Behavioural Brain Research. 2004, 153, 171-180. 20. Acquistapace, P.; Hazlett, B. A.; Gherardi, F. Unsuccessful predation and learning of predator cues by crayfish. J Crustacean Biol. 2003, 23, 364-370. 21. Hazlett, B. A. Conditioned reinforcement in the crayfish Orconectes rusticus. Behaviour. 2007, 144, 847-859. 22. Tierney, A. J.; Lee, J. Spatial Learning in a T-Maze by the Crayfish Orconectes rusticus. J Comp Psychol. 2011, 125, 31-39. 23. Weisbord, C. D.; Callaghan, D. T.; Pyle, G. G. Associative learning in male rusty crayfish (Orconectes rusticus): conditioned behavioural response to an egg cue from walleye (Sander vitreus). Can J Zool. 2012, 90, 8592. 24. Tierney, A. J.; Andrews, K. Spatial behavior in male and female crayfish (Orconectes rusticus): learning strategies and memory duration. Anim Cogn. 2012. 25. Klocker, C. A.; Strayer, D. L. Interactions among an invasive crayfish (Orconectes rusticus), a native crayfish (Orconectes limosus), and native bivalves (Sphaeriidae and Unionidae). Northeast Nat. 2004, 11, 167-178. 26. Martin, A. L.; Moore, P. A. Field observations of agonism in the crayfish, Orconectes rusticus: shelter use in a natural environment. Ethology. 2007, 113, 1192-1201. 27. Martin, A. L.; Moore, P. A. The influence of dominance on shelter preference and eviction rates in the crayfish, Orconectes rusticus. Ethology. 2008, 114, 351-360. 28. Pintor, L. M.; Sih, A. Differences in growth and foraging behavior of native and introduced populations of an invasive crayfish. Biol Invasions. 2009, 11, 1895-1902. 29. Olden, J. D.; Vander Zanden, M. J.; Johnson, P. T. J. Assessing ecosystem vulnerability to invasive rusty crayfish (Orconectes rusticus). Ecol Appl. 2011, 21, 2587-2599. 30. Baggerman, G.; Boonen, K.; Verleyen, P.; Loof, A. D.; Schoofs, L. Peptidomic analysis of the larval Drosophila melanogaster central nervous system by two-dimensional capillary liquid chromatography quadrupole time-of-flight mass spectrometry. Journal of Mass Spectrometry. 2005, 40, 250-260. 7 31. Finamore, F.; Pieroni, L.; Ronci, M.; Marzano, V.; Mortera, S. L.; Romano, M.; Cortese, C.; Federici, G.; Urbani, A. Proteomics investigation of human platelets by shotgun nUPLC-MSE and 2DE experimental strategies: a comparative study. Blood Transfus. 2010, 8 Suppl 3, s140-148. 32. Levin, Y.; Hradetzky, E.; Bahn, S. Quantification of proteins using data-independent analysis (MSE) in simple andcomplex samples: a systematic evaluation. Proteomics. 2011, 11, 3273-3287. 33. Pieroni, L.; Finamore, F.; Ronci, M.; Mattoscio, D.; Marzano, V.; Mortera, S. L.; Quattrucci, S.; Federici, G.; Romano, M.; Urbani, A. Proteomics investigation of human platelets in healthy donors and cystic fibrosis patients by shotgun nUPLC-MSE and 2DE: a comparative study. Mol Biosyst. 2011, 7, 630-639. 34. Silva, J. C.; Gorenstein, M. V.; Li, G. Z.; Vissers, J. P.; Geromanos, S. J. Absolute quantification of proteins by LCMSE: a virtue of parallel MS acquisition. Mol Cell Proteomics. 2006, 5, 144-156. 35. MacLean, B.; Tomazela, D. M.; Shulman, N.; Chambers, M.; Finney, G. L.; Frewen, B.; Kern, R.; Tabb, D. L.; Liebler, D. C.; MacCoss, M. J. Skyline: an open source document editor for creating and analyzing targeted proteomics experiments. Bioinformatics. 2010, 26, 966-968. 36. Moran, T. H.; Dailey, M. J. Intestinal feedback signaling and satiety. Physiol Behav. 2011, 105, 77-81. 8 Chapter 2: Neuropeptide Discovery in the Decapod Crustacean Adapted from Schmerberg, C. M., Chen, R., and Li, L. Prog. Neurobiol. 2012 [in preparation]. 2.1. Abstract Invertebrates have played an important role in the discovery and characterization of neuropeptides (NPs). NPs are a distinct class of neurotransmitters (NTs) that are composed of short amino acid chains, and they can act both synaptically like classical NTs and systemically in a manner similar to neurohormones. Discovery and characterization of the identities and functions of NPs have made great strides in invertebrate model animals, particularly in decapod crustaceans, due to a number of factors. In this review, advances in techniques for NP identification and quantitation are discussed, with a focus on mass spectrometry (MS)-based tools. Assays for determining NP function are also described. Finally, the NP families known thus far in crustaceans are described in detail. As this article contains both methodology information and the current state of knowledge with regards to crustacean NPs, it will provide useful insight to researchers studying NPs in vertebrates as well as in invertebrates. 2.2. Introduction Neuropeptides (NPs) are a class of neural signaling molecules that are defined by their composition—short chains of amino acids—and activity—neurochemical signaling or modulation of signaling. These compounds are important in neural signaling throughout the animal kingdom and have been implicated in a wide variety of disorders in humans. However, they have not been studied as intensely as the classical small molecule neurotransmitters (NTs) for a variety of reasons. The chemical complexity of NPs poses a great challenge to their study. These molecules are much larger than NTs, ranging from 500-8,000 Da, and have multiple functional groups 9 within their structures. Due to their intermediate size, larger than small molecule neurotransmitters (NTs) but smaller than proteins, NPs can be more difficult to separate from other components of the cell. Methods for isolating either small molecules or proteins are quite mature due to their longer history of study. In addition, the multiple functional groups on the variety of amino acid side chains of NPs are all potentially reactive. Small molecule NTs typically contain only a few reactive groups, and many such side chains on proteins are hidden from the environment by complex tertiary structures. A wider variety of NPs than small molecule NTs is also possible mathematically, because the molecules can contain 4-20+ amino acids, each of which has more than 20 possible identities. The size, complexity, and reactivity of NPs present challenges for their study. In addition, the genomic information of an organism can be of limited use for NP study because a number of important processing steps occur between translation and release of a NP at the synapse or into the circulation (see Fig. 2.1). Preprohormone genes may carry multiple copies of an NP, or multiple isoforms of NPs with similar sequence motifs (said to be in the same family). The cleavage of preprohormones into active NPs is also not always predictable. Although there are several known prohormone processing proteins with known cleavage sites (such as dibasic AAs), some NPs result from unknown cleavage mechanisms In addition, many types of posttranslational modifications may affect the activities of NPs, and these modifications cannot be determined by genetic techniques from gene sequences (reviewed in [1]). Finally, even in this “post-genomic era”, not all species of interest have sequenced genomes, including all crustacean species with the exception of the water flea Daphnia pulex [2]. Due to the challenges of studying NPs, many researchers have turned to simpler model organisms to facilitate their study through other means. The study of NPs in invertebrates has been instrumental in our 10 understanding of these molecules. The simplicity of the blood-brain barrier in many invertebrates and the presence of well-defined, simple neural circuits have been important factors for using these species in NP research. The presence of a mature field investigating the effects of neuromodulators on neural circuits, using one of these well-defined circuits—the stomatogastric ganglion (STG)—and computational neuroscience methods, is another reason for studing NPs in crustaceans [3]. Several reviews on crustacean neuropeptides have been published, but most have focused on a single NP family, or have not discussed the use of cutting-edge analytical chemistry tools, such as mass spectrometry, in the study of NPs in detail [4, 5]. The following review will discuss past achievements and recent advances in the discovery and functional analysis of neuropeptides in invertebrates, focusing on practical aspects, as well as contain a summary of the known NPs in crustacean species. 2.3. Analysis Methods for Invertebrate Neuropeptide Discovery Analysis of NPs must take into account the challenges in their analysis mentioned above. Recent advances in mass spectrometry (MS) have greatly expanded the possible applications of this technique, and have made it ideal for NP study, particularly in invertebrates. This review will mention classical biochemistry techniques for NP study, but will focus on MS techniques. Techniques that have been replaced largely by MS, such as Edman degradation, will not be discussed in detail. 2.3.1. Immunochemical Techniques For many years, NPs have been detected with antibodies. Antibodies are used ubiquitously throughout molecular biology due to their supposed specificity and ease of visualization. Primary or secondary antibodies can be conjugated with a fluorescent dye, horseradish peroxidase (HRP), radioactive isotopes, metal (for electron microscopy), or other 11 label for visualization or quantification. HRP and fluorescence are most commonly used to determine the location where the NP is found. Radioimmunoassay (RIA) and enzyme-linked immuno-sorbent assay (ELISA) are just two of the quantitative techniques that can be conducted. Although highly specific, high-affinity antibodies can be created, the quality of the antibody (particularly of polyclonals) will vary from batch to batch, and high specificity is rarely found in NP antibodies. Often, the portion of the NP sequence that the antibody recognizes is found on many different NPs, due largely to the presence of common sequence motifs in NPs of the same family. For instance, many invertebrate FMRFamide-like peptides typically end in the sequence LRFamide. It has been the authors’ experience that polyclonal antibodies raised against NPs will have affinity for multiple NPs of the same family, along with other unrelated NPs that may share some similar sequence motifs, such as an amidated N-terminus. Therefore, it is impossible to distinguish between the members of a single NP family, all of which may have distinct functions, by using immunochemical techniques. 2.3.2. Mass Spectrometric Techniques Mass spectrometry (MS) is a technique that measures the mass to charge ratio (m/z) of compounds in the gas phase. Taking molecules from solid or liquid preparations and putting them in the gas phase with charge on them is a major concern in MS, and it is referred to as ionization. Typically, the process of ionization is harsh to the molecule and induces structural and chemical changes. In early mass spectrometry of small chemical compounds, a fraction of the analyte would remain unfragmented upon ionization due to the strength of the chemical bonds, and thus fragment and precursor masses would be obtained from a single mass spectrum. The fragment masses could be reassembled into the intact mass to determine the molecule’s structure. This was not the case, however, with large biomolecules, which are present at lower 12 concentrations and have more labile chemical bonds; only fragment ions were observed with early ionization techniques, and it was nearly impossible to reassemble these fragments into the original molecule. With the advent of soft ionization techniques in the late 20th century, it became possible to conduct mass spectrometric analysis of intact biomolecules with multiple functional groups and modifications intact. This opened up a new field of biological MS, and NPs are ideally suited for this analysis. Several strategies for NP analysis by MS-based techniques are illustrated in Fig. 2.2. Soft ionization techniques include electrospray ionization (ESI) and matrix-assisted laser desorption ionization (MALDI), among others. In ESI, analytes in a liquid stream are ionized by spraying the liquid through a small pore with a large electrical charge applied to it. This causes the molecules to enter the gas phase and to gain electrical charge(s) in droplets while the liquid solvent evaporates, leaving just charged analytes. MALDI employs a laser to irradiate a co-crystallized mixture of the analyte and a laser-absorbing chemical called the matrix. One conceptualization of the mechanism by which MALDI works is that irradiation of the matrix removes the analytes and matrix molecules from the crystallized surface in a gas-phase plume, at which time a charge is also imparted to the molecules. Unlike earlier methods for ionization used in mass spectrometry, these techniques can impart charge to analytes without fragmenting them, and therefore determination of the m/z of an intact biomolecule is possible. Adding additional steps of MS analysis permits fragmentation of the intact molecule and determination of the m/z’s of its fragments, which can then be reassembled to determine the structure of the original molecule. This is typically done in two steps as tandem MS (MS/MS or MS2), although more steps of MS analysis can be conducted to gain further information (n steps of MS, called MSn). For peptides, including NPs, fragmentation in MS/MS typically occurs along the peptide backbone, and most of the 20 common amino acids 13 (AAs) have unique masses. Thus, the m/z change between peaks can be calculated and correlated with an AA, and the sequence of the peptide can be determined from the MS/MS spectrum. As MS identifies the m/z of analytes, it has greater specificity than antibody-based methods, the pitfalls of which have been discussed above. The sensitivity and quantitative abilities of MSbased techniques have also improved in recent years. Soft ionization MS and MS/MS are the most specific methods for NP identification. The application of these powerful techniques to NP study will be discussed in several following sections. A workflow illustrating some of these techniques in application to studying crustacean NPs is illustrated in Fig. 2.2. 2.3.3. Sampling Although seemingly simple, the question of how to obtain NP samples from an invertebrate can be quite complex. The choice of whether to obtain samples from tissues or physiological fluids (hemolymph in arthropods) has profound implications on the physiological relevance of the NPs collected and the type of information one can obtain about these NPs. Samples from tissues typically contain all NPs, including those stored at synaptic terminals, preprohormones recently translated in the endoplasmic reticulum (ER), intermediate processing products in vesicles undergoing transport to the axon terminal, and extracellularly released NPs. In contrast, samples from physiological fluids contain only released NPs. Fig. 2.1 shows the process by which NPs are synthesized, stored, and released, and demonstrates some of the possible steps along a NP’s processing pathway. Spatial distribution of NPs is completely lost when studying samples taken from circulating fluids, unlike tissue samples for which NP distribution can be mapped with histology or MS. The application of tissue-based and fluidbased techniques, with a focus on preparation for mass spectrometric analysis, will be discussed in the following section. A schematic showing many of these techniques is found in Fig. 2.2. 14 2.3.3.1. Sampling from tissues For all tissue-based techniques, the organ of interest is dissected from the animal prior to being subjected to NP analysis via imaging or survey techniques. Thus, studies in which time or experimental-related changes are of interest must use different animals for each condition, and inter-animal variability is a concern. Imaging techniques differ from survey techniques by showing the spatial distribution of NPs throughout a tissue, whereas survey techniques yield a single NP profile for the entire tissue. 2.3.3.1.1. Tissue imaging The development of mass spectrometry imaging (MSI) in recent decades has greatly advanced the study of neuropeptides. MSI typically relies upon MALDI ionization, which has a high tolerance for salt and impurities in the sample. This facilitates the detection of bioactive compounds directly from complex samples and even biological tissues. Using this mass spectrometric technique, a number of tissue-based neuropeptide discovery methods such as single cell analysis, direct tissue analysis, and MS imaging (MSI) have been developed. These techniques combine molecular identification with localization within the animal or even within a cell. A number of biological tissues have been studied, including single neurons, cell clusters, ganglia, and single vesicles. Combined with accurate mass measurement or de novo sequencing, the neuropeptide content in the target specimen can be readily identified without the extensive sample extraction and purification steps that are required by solution based techniques. A number of useful reviews of MSI have recently been published [6-12], and this work will serve as a summary and include perspectives from the authors’ own experience. In the first section, MSI of large tissues will be discussed. 15 2.3.3.1.1.1. MSI of NPs—Technical Aspects In MSI, a Cartesian grid is superimposed upon the tissue, and multiple mass spectra are acquired, at least one for each spot on the tissue. An image can then be generated by processing this set of MS data with each pixel in the image corresponding to the spectra acquired at that point. The density of ions at each point can be mapped across the tissue using different colors or intensities of color to indicate the ion intensity. Following a single acquisition of mass spectra across the tissue in this manner, ion density maps can be rapidly generated for the entire tissue for any m/z within the range of MS acquisition, and thus MSI allows for analysis of far more analytes in a single experiment than histochemical techniques. MSI is particularly attractive in neuroscience because the location of biologically active molecules can be directly related to their functions, and MSI is also helpful for investigation of expression changes that occur as a result of experimental perturbation or animal development. The spatial distribution of target molecules obtained by MSI can be lost when tissues are homogenized for some other types of MS analysis techniques. A recent paper employing MSI is of particular interest [13]. Two types of technologies are typically used in MSI: matrix-assisted laser desorption/ionization (MALDI) and secondary ion mass spectrometry (SIMS). The choice of technique is dependent on the spatial resolution desired, the chemical properties of analytes of interest, and the instrumentation available. For optimal spatial resolution, SIMS imaging has submicron precision. However, its method of ionization relies upon a high energy (5-25 keV) primary ion beam (Ca+, In+, Cs+, Aun+ etc.) to bombard the surface of the sample and produce ions for MS. Not only does this require specialized equipment, the high energy of the ion beam can break down the target molecules, particularly large biological compounds, into smaller fragments. Fragmentation is less common in MALDI MS due to ionization by laser irradiation, 16 and thus larger intact molecules can be detected, with the tradeoff of reduced spatial resolution (~50 µm). For imaging techniques, depending on the thickness of the specimen, tissue can be placed directly on the sample plate, or frozen and sliced by a cryostat into 5 to 15 µm sections prior to transferring it to the plate. This is the full extent of sample preparation for SIMS. In MALDI-MS, a matrix must be applied to the sample prior to instrumental analysis. This is a crucial aspect of sample preparation, because deposition of MALDI matrix in must be homogeneous to avoid significant migration of analytes on the surface of the tissue or the formation of “hot spots” due to more or less matrix present in a single location. Some methods of matrix application that limit migration and uneven coating include robotic micro-spotters or modified printers creating dense arrays of droplets [14], sublimation of matrix onto tissue, and airbrushes for matrix application. The sample is then ready for data acquisition. Data is acquired in a grid pattern with a predetermined step size (e.g. 100µm), depending on the sample size and required resolution. Most often, specialized software is used to acquire and process data. Notable software for MSI includes BioMap (developed by Novartis and available free of charge at www.maldi-msi.org) and vendor-specific software. The processing software can generate heat map ion images of target m/z values by plotting the intensity of a single compound in each mass spectrum against the x, y coordinates of each pixel. Over the last decade, numerous developments have been achieved to improve the quality of MALDI imaging results, both in sample preparation methodologies and instrumentation. For sample preparation, a number of approaches have been tried, including altering the tissue itself or the matrix, to improve sensitivity and/or spatial resolution. Ethanol has been commonly used for tissue fixation [15]. By rinsing with serial ethanol-containing solutions, physiological salts and impurities can be removed from the tissue to improve spectral quality. Additionally, ethanol can 17 fix the proteins in the tissue sample in place to eliminate redistribution of these proteins. A final benefit of ethanol fixation is greater stability over time. However, based on the observations in our laboratory, rinsing with ethanol may cause migration of hydrophilic NPs [13]. Glycerol stabilization has also been employed to preserve cell morphology and prevent migration of NPs [16]. In addition to treatment of tissue, sample thickness has also been investigated as a factor in the sensitivity of MSI for biomolecules. Sugiura et al. systematically investigated serial mouse brain sections with a range of thicknesses, and concluded that thinner tissue sections (<10 µm) were optimal for MSI [17]. Matrix-related improvements to MSI have also been explored. For example, depositing gold on the tissue surface increased the signal intensities in the peptide mass range [18]. Resolution is another important aspect that is usually limited by matrix crystal size and the dimension of the ionizing laser beam. A plethora of methods and devices have been used to reduce matrix crystal size, and only a few examples will be discussed here. One matrix application method reported by Sugiura and colleagues combined spraying and dispensing droplets. This led to more homogeneous crystals and increased mass spectral peak intensity [19]. Sold ionic matricies have been created from the mixture of a conventional MALDI matrix with an organic base (e.g. aniline). These matricies improved spectral quality, crystal homogeneity, and resistance to laser irradiation (i.e. they were not ablated rapidly) [20]. A large amount of effort has gone into improving sensitivity and spatial resolution for MSI focusing on the sample and matrix. Improvements to the spatial resolution of MALDI-MSI data by alteration of laser properties are also of note. The diameter of a laser beam is usually 50 or 100 µm for commercial instruments equipped with Nd:YAG or N2 lasers. The oversampling technique can be used to 18 improve spatial resolution to a size smaller than the laser diameter by altering the way in which images are acquired. First, the matrix in one location is completely ablated. The sample plate is then moved at a step size that is smaller than the laser diameter, and spectra are acquired again until matrix is ablated, and so on. Thus the spectra acquired are from an area with size equal to the small step size the plate was moved, which is much smaller than the laser size [21]. This technique is valuable for studying small neural structures. New laser developments are also capable of improving spatial resolution. Compared to classical Nd:YAG or N2 laser, the Smartbeam laser developed by Bruker Daltonics has an adjustable focus size ranging from 10 to 80µm, which allows a smaller step size to be used during experiments. A combination of improvements in sample preparation, matrix deposition, and lasers have improved the sensitivity and spatial resolution of MSI over the past several years, but these remain as major challenges to consider in MSI experiments. 2.3.3.1.1.2. MSI of NPs—Recent Advances and Comparison to Traditional Techniques MALDI-IMS has been used to map a variety of compounds in tissue, including cholesterol and phospholipids [22], proteins [23], and pharmaceuticals [24, 25]. More recently, the use of IMS to map the distribution of neuropeptides, particularly in mammalian neural tissues, has gained attention. In one study, SIMS and MALDI imaging were both employed to map the distribution of NPs, lipids, and small molecules in rat spinal cord. This study determined that several NPs—including substance P and somatostatin-14—were colocalized [26]. MSI has been used to profile the NPs in several invertebrate species, notably in Aplysia. Its millimetersized ganglia have been profiled, and the peptide contents of cell bodies and neuronal processes (neurites) in single isolated Aplysia neurons have been compared [16, 27]. Crustacean species have also been subjected to MSI. Multiple neuropeptide families in the brain and pericardial 19 organ of the crab Cancer borealis have been mapped in detail [13]. Three-dimensional MALDI imaging of the brain of C. borealis was also recently presented by Chen and coworkers. The simple scheme employed was able to generate a thorough description of neuropeptide localization in the crab brain. In the image obtained, a great degree of correlation is seen between NPs and the identified structures of the crab brain [28]. MSI has had recent success in imaging the distribution of NPs, particularly in invertebrate tissues. Traditionally the spatial localization and relative amounts of bioactive peptides are visualized with immunostaining or in situ hybridization. Immunostaining uses fluorescent, enzyme-linked, or radiolabeled antibody to probe the location of target proteins or peptides in the tissue. It has the advantage that the precise peptide sequence is not required, and its high resolution enables visualization of subcellular distribution. However, one main drawback exists to this method—the specificity of antibodies, which has disadvantages described previously. Frequently, multiple neuropeptides (typically from the same NP family) react with a single antibody raised against a single NP. Thus, extensive rounds of antibody development may be required to obtain an antibody that is specific for a single NP, and in many cases it may be impossible to create such an antibody. This type of experiment is also unsuitable for discovering novel NPs because the compound must be identified before an antibody can be made for it. For in situ hybridization, a nucleotide probe binds to the target gene transcript and is visualized, frequently through fluorescent tagging of the probe. Thus, this technique also requires prior knowledge of the target nucleotide sequence, which is again unsuitable for novel NP discovery and difficult to carry out in organisms without known genomes. MSI is more flexible than immunostaining or in situ hybridization because the peptides of interest do not need to be preselected. The sample preparation of MSI is also relatively simple without the need for 20 extensive washing and fixation. Furthermore, numerous target molecules with diverse chemical structures can be analyzed simultaneously, and their colocalization can be visualized simply to achieve valuable information regarding interactions between different neuropeptides. 2.3.3.1.2. Tissue Survey Survey techniques include homogenization followed by extraction, single-cell analysis of large molluscan neurons, and direct tissue profiling of small organs in the decapod crustacean nervous system. These are illustrated in Fig. 2.2. Of these three, homogenization followed by extraction is the most common, and can be used in conjunction with both MS and immunochemical detection. One or more of the desired organ or organs are collected and the tissues are homogenized. NPs are then extracted from the homogenate with a solvent that selectively solubilizes NPs over lipids and proteins, such as acidified methanol. This sample is then subjected to a number of clean-up steps (delipidation, desalting, ultracentrifugation) and analytical separations (LC, electrophoresis) to reduce the sample complexity prior to NP analysis, which is conducted by MS or immunochemistry. Fig. 2.3 illustrates some of the sample preparation techniques that can be used on liquid samples. 2.3.3.1.3. Direct Tissue, Single-Cell, and Single Vesicle Analysis Direct tissue profiling and single neuron analysis—techniques based on MALDI-MS— are useful in obtaining a “snapshot” of the NP content of a small organ or single neuron. In these methods, the tissue or single neuron to be characterized is carefully dissected, then washed, placed on a MALDI plate, and coated with the desired matrix. The strength of this method lies in the ability to determine which NPs are present in a small organ or large moluscan neuron with minimal sample preparation steps. This increases the sensitivity and speed of analysis over extraction techniques. 21 2.3.3.1.3.1. Detailed Information on Sample Preparation for Direct Tissue and Single Cell Analysis As the first step of these studies, tissue dissection is crucial to the quality of the acquired mass spectra. Molluscan neurons (50-500 µm) have been demonstrated to be suitable for singlecell analysis. For smaller cells, micromanipulation techniques using micro glass capillary or small diameter glass micropipettes are required to isolate the cells and transfer them to the sample plate [29]. When the cells are too small or difficult to dissect, a small piece of tissue or neuronal cluster can be analyzed directly using similar methods to MSI, in which the small tissue is subjected directly to laser irradiation for ionization. Sample preparation also plays a pivotal role in this type of analysis. The physiological salt concentrations associated with neurons from marine specimens or the saline solution used for dissection greatly interferes with MALDI analysis. These concentrations are about four times higher in marine crustaceans compared to mammals. Several simple rinsing procedures have been developed to enable direct assay for peptides with minimal sample handling. Garden et al. employed on-target water washing of Aplysia neurons prior to MALDI in order to reduce the high levels of salts found in this marine mollusk [30]. Extracellular salts and physiological saline can also be removed in a stepwise fashion by spot-to-spot cell transfers to a fresh matrix drop on the sample plate. Li and colleagues employed concentrated 2, 5-dihydroxybenzoic acid (DHB) matrix solution in 4:1 acetone/water mixture for a method of this fashion, termed on-target micro-extraction [31]. An alternative simplified preparation method involves replacing the physiological saline with the DHB matrix solution during dissection, so that the salt removal occurs simultaneously with neuronal isolation. For direct tissue analysis, rinsing samples first in acidified methanol followed by dilute DHB (10mg/ml) aqueous solution has been shown to 22 efficiently remove physiological saline and extract the neuropeptides from the cell for sensitive MS detection [32]. Sample preparation for small tissue analysis mostly involves methods to remove salts present in the animal under normal conditions, as these physiological concentrations of salt are more than sufficient to inhibit ionization in MALDI. Once the sample is dissected and prepared, the following procedure is relatively simple. For matrix deposition, the dried-droplet method can usually be used with success. One of many widely used MALDI matrices can be chosen, such as 2, 5-dihydroxybenzoic acid (DHB) or alpha-cyano-4-hydroxycinnamic acid (CHCA) for peptide analysis, and sinipinic acid (SA) for protein analysis. In the authors’ experience, DHB is preferred for tissue-based analysis for several reasons. These include stabilization of cell membranes for improved microdissection, reducing proteolytic enzyme activity, and replacement of salt ions present in the tissue [30]. Storage in a DHB solution is also able to preserve the tissue for up to several years without obvious degradation of the NP content [33]. MALDI matricies can be used during or after tissue dissection to improve spectral quality for small tissue analysis with a number of advantages. However, sensitivity is a crucial concern for analyzing direct tissue and single cell samples, due to the miniscule sample amount. Sample dilution induced by matrix spreading can reduce signal intensity and cause poor spectral quality. It is reported that deposition of matrix in nanoliter volumes over a period of 10 s can be used to eliminate spreading [34]. Wang et al. modified the MALDI plate with a parafilm coating to improve the sensitivity of detection by concentrating the samples on target via the solvent-repellent effect of the parafilm surface [35]. In addition, several novel small-scale sample preparation techniques have been developed recently to ease the complicated dissection procedure for single cell analysis while increasing the detection sensitivity. For instance, neuropeptide content in neurons and ganglia can be easily 23 extracted by incubating them in an aqueous solution of DHB. The resulting signaling moleculecontaining solution can be directly examined by MALDI MS [33]. A DHB solution-based capillary electrophoresis separation method was also developed to couple with MALDI analysis of liquid incubated with small neuronal tissues or single cells [36]. This separation prior to analysis will improve the dynamic range of detection and increase the number of analytes detected by reducing sample complexity. In an additional innovation to assay large numbers of individual cells for comprehensive profiling of tissue slices or organs, a massively parallel preparation of single-cell-sized samples was developed by placing a thin tissue section on an array of glass beads attached to parafilm. When the parafilm is stretched, the tissue is divided into thousands of small pieces, which can be analyzed separately [37]. This mechanical separation allows greater spatial resolution in imaging of the tissue than would previously have been possible. Although most sample preparation techniques for small tissue analysis can induce analyte dilution, specialized matrix deposition techniques and separation, both chemical and mechanical, can increase sensitivity. 2.3.3.1.3.2. Recent Developments in Direct Tissue and Single Cell Analysis The study of individual identified cells is extremely valuable for neuropeptide discovery, since fewer analytes are present in a single cell compared to tissue extracts. Furthermore, it is also important to investigate chemically heterogeneous nervous systems in which cells specialize in the synthesis of functionally and/or chemically distinct signaling molecules. Thus knowledge of neuropeptide complements on a cellular basis is essential for understanding their bioactivities. The first single-cell MALDI-MS study was performed using identified neurons from the Lymnaea stagnalis brain [38]. The mollusk is a good choice for improving sample preparation techniques for single cell analysis, since the relatively large size of their neurons and greater 24 simplicity of their nervous system makes dissection and related sample handling less challenging than in other species. Since then, various freshwater and marine mollusks have been studied, including Helix aspersa, Aplysia californica, Aplysia vaccaria, Phyllaphlysia taylori, and additional work in Lymnaea stagnalis [39-42]. In these studies, a large range of NPs have been identified, such as insulin-related peptides, FMRFamide-related peptides and small cardioactive peptides. For further review, see recent papers by Li and Hummon [43, 44]. Single-cell analysis using molluscan neurons has made significant advances in our ability both to analyze small, complex samples and in our understanding of their neuronal NP content. With improvements in sample preparation methods and increases in performance of MS instruments, many more organisms have been studied in recent years. Ma and coworkers reported the first single cell study of individual insect neurons by analyzing pheromone biosynthesis activating neuropeptide (PBAN) in subesophageal ganglion neurons (diameter around 20 µm) from the corn earworm moth Helicoverpa zea [45]. Recently Neupert et al. performed dissection and MS analysis of identified neurons and neurosecretory cells from the American cockroach Periplaneta americana. In these studies, a large number of FMRFamiderelated peptides (FaRPs) [46] and allatotropin-related peptides (ATRP) [47] were detected. Neupert and coworkers also reported the first detection of tick periviscerokinin from single cells in Ixodes ricinus and Boophilus microplus [48]. On an even smaller scale, single vesicles from the atrial gland of Aplysia californica have been analyzed by Rubakhin and coworkers. A wide range of bioactive peptides within individual vesicles (1-2 µm) were identified, including products from several genes colocalized within the same vesicle [49], which runs counter to the canonical model of how NPs are loaded into vesicles. Other species have been successfully 25 subjected to single-cell MALDI analysis for NP content, including the nematode Ascaris suum [50] and the rat Rattus norwegicus [51]. In addition to studies using single cells or organelles, various reports have been published analyzing neuron clusters or endocrine nerve tissues. For these samples, direct tissue analysis is widely used, especially for smaller cells that are difficult to isolate. This method is a simple and fast way to achieve a snapshot of the neuropeptide content in a target organ. For example, multiple putative tackykinin-related peptides in Drosophila were identified by directly analyzing the anterior-ventral brain using MALDI MS [52]. Direct tissue analysis (analysis of discrete points, as opposed to IMS of all possible points) is also extremely useful to acquire a complete mapping of neuropeptide distribution throughout the whole animal. For example, Yew et al. detected numerous neuropeptide families in several major neurohemal structures in nematodes, and a body-region-specific distribution of these neuropeptides was observed [53]. Larger tissues can be analyzed using direct tissue profiling to rapidly obtain profiles of the NP content of an entiretissue or of discrete spots within the tissue. This method is also extensively used for neuropeptide discovery in crustaceans. The neuroendocrine tissues, pericardial organ (PO) and sinus gland (SG), from several different crustacean species were analyzed by Li et al. using MALDI-TOF, and found to contain a large number of putative neuropeptides [54, 55]. In more recent studies, neuroendocrine organs isolated from the crab Cancer borealis were analyzed by a MALDI Fourier transform mass spectrometry (FTMS) instrument using an in-cell accumulation technique. This technique improves the detection sensitivity by accumulating ions from many subsequent laser shots within the cell prior to analysis, which is particularly helpful for analytes from a complex tissue sample [32]. This same MALDI-FTMS platform was also used for analysis of large peptides, such as 26 crustacean hyperglycemic hormones (CHH), in studies that incorporated an in-cell cleanup step to remove matrix adducts, which facilitated detection of these large molecules [56]. Direct tissue analysis has been used, along with a wide variety of other sample preparation techniques, to detect and characterize hundreds of neuropeptides in the crustacean [57-59]. Several putative tachykinin-related peptides have also recently been characterized in midgut epithelial tissues and several neural organs in multiple Cancer species, and the gut-brain distribution of these peptides appears to be conserved [60]. Direct tissue MALDI analysis has been an instrumental technique for crustacean NP discovery and localization. 2.3.3.1.4. Comparison of Tissue-Based Techniques All three types of tissue-based sampling techniques for mass spectrometry analysis have unique advantages. Imaging is the only method to obtain a detailed map of the distribution of one or more NPs throughout an organ. Direct tissue and single neuron analyses yield a quick snapshot of the NP content of the sample, and can be conducted with less material, which in turn leads to increased detection sensitivity. NP distribution information obtained from imaging, direct tissue, and single neuron analysis can be integrated with functional information already known about specific ganglia, sub-structures of these ganglia, or identified neurons. For instance, an NP found solely in the olfactory bulb of the crab brain, either by MSI or direct tissue analysis, may have a functional role in the chemical senses and/or how they affect other neuronal processes. Thus, direct tissue, imaging, and single cell analysis techniques allow for preliminary assignment of NP function based on the known roles of certain subsets of cells to which the NPs are localized. In contrast, homogenization followed by extraction reduces the functional information content of the sample by combining NPs from many cells that may have individual functions into a single sample—all spatial distribution information is lost. Solid-phase ionization 27 techniques are the only option for intact tissue analyses, however, and these techniques often suffer from run-to-run irreproducibility and poorer quality MS/MS spectra due to singly charged ions. Homogenization and extraction of neural tissues has advantages over intact tissue-based techniques in the variety of treatments that the samples can undergo to improve the quality and sensitivity of analysis. Chief among these are liquid-phase reactions and analytical separations, illustrated in Fig. 2.3. For higher-mass NPs, an enzymatic digestion step is valuable to generate fragments with masses within the range where modern mass spectrometers have high sensitivity and resolution. Enzymatic reactions can also be carried out to characterize certain posttranslational modifications. Other reactions that are most efficiently conducted on samples in the liquid phase include labeling reactions to improve MS/MS fragmentation, identify functional groups, and incorporate isotopes for more reliable MS and MS/MS quantitation. Although reactions can be conducted on the surface of solid samples, this is less efficient and can lead to diffusion of NPs away from their original location in the tissue. Enzymatic reactions conducted on tissue samples for histochemistry are less common. A variety of techniques to reduce sample complexity, such as immunoprecipitation (IP), delipidation, desalting, ultracentrifugation, ultrafiltration, and dialysis are also most amenable to liquid samples (illustrated in Fig. 2.3). These techniques are capable of enriching neuropeptides by antibody-antigen interactions, hydrophobicity, and molecular weight. Additional liquid-based separation techniques, such as LC (from preparative to UPLC scale) and electrophoresis (capillary or gel), are compatible with homogenized and extracted NPs, but not with intact tissues. Removing salts, lipids, and high molecular weight compounds allows for better ionization of samples in MS. IP, LC, and electrophoresis separate analytes to decrease the 28 complexity and increase the sensitivity of analysis. Extracted samples from multiple animals can also be pooled to increase sensitivity and allow detection of low-abundance analytes. When using MS, liquid-based ionization methods can be used to introduce extracted NPs into the mass spectrometer. Chief among these is electrospray ionization (ESI), which has the advantage of adding multiple charges to a single NP. This increases the quality of fragment ions observed in a subsequent MS/MS spectrum and facilitates de novo sequencing. ESI also is more reproducible from run-to-run than solid-phase ionization techniques, which improves quantitative analysis. 2.3.3.2. Sampling from Biological Fluids in Invertebrates Liquid-based sampling relies upon withdrawing a biological fluid from the experimental animal and analyzing this substance for the presence of NPs. Fig. 2.3 illustrates some methods of obtaining liquid samples from invertebrates. For invertebrates, this usually takes the form of hemolymph sampling. This substance is analogous to blood in invertebrates, and bathes all organs due to the open circulatory system of invertebrates. Extremely small samples of hemolymph can be used, such as the hemolymph from a single fruit fly [61]. All of the above solution-based sample cleanup, labeling, and separations techniques can be conducted on this fluid, depending on the volume obtained, which can then be analyzed by MS or immunochemical techniques. However, hemolymph withdrawal can be challenging and can expose the animal to a great deal of stress. Many small invertebrates can only be used for a single hemolymph sample due to the limited volume. Aquatic invertebrates must be removed from the water for sampling, which exposes them to a great deal of stress and potentially hypoxia. Dilution of NPs in hemolymph and the complexity of hemolymph itself also decrease the sensitivity of detection. NPs released into this fluid may be present at a concentration that is 29 orders of magnitude less than the concentration at the site of release or action. For instance, many NPs are postulated to act in the micro- to nanomolar range at their receptors. However, diffusion from the micro-environment of immediately adjacent to the cell into the hemolymph as a whole dilutes these compounds into a much larger total volume. For this reason, detection of NPs in hemolymph with high-specificity tools such as mass spectrometry is challenging. As an example, hundreds of NPs have been detected in tissue extracts from Cancer borealis [35, 56, 59, 62-66], but studies where sampling has been conducted from the hemolymph have only identified tens of these NPs [67-69] (summarized in Table 2.1). The hemolymph also contains many high-abundance proteins and protein fragments in vivo, which can obscure the detection of low-abundance NPs. In the same hemolymph studies mentioned above [67-69], only 11 NPs could be detected in hemolymph collected directly from the crab via needle, and fragments of two high-abundance proteins (cryptocyanin and actin) dominated the mass spectra obtained. With the sample collection technique of microdialysis (MD), the number of NPs detected increased to 35 with fragments of only one high-abundance protein collected. This technique allows sampling from the hemolymph by utilizing a MD probe, which is implanted into the animal with the tip of the probe in contact with the hemolymph. At the tip of this probe is a semipermeable dialysis membrane, through which molecules below a certain molecular weight (the molecular weight cutoff or MWCO) can diffuse into the probe. Liquid is constantly pumped through the probe at a low flow rate, and this liquid carries the molecules that have diffused through the probe tip through the outlet tubing. The liquid that flows out can then be collected for analysis. In this situation, the main advantage of MD is the exclusion of high molecular weight compounds from the sample, which leads to a less complex sample with fewer proteins that can degrade and interfere with the detection of low-abundance 30 NPs. However, high-abundance protein fragments are still detected in MD samples, indicating that these fragments must be present in the hemolymph in vivo. 2.3.3.3. Comparison of tissue-based to liquid-based techniques 2.3.3.3.1. Advantages of liquid-based techniques The two advantages of liquid-based sampling are the ability to sample only NPs released into the extracellular space, and the potential to sample multiple times from the same animal (for larger invertebrates). Sampling only NPs in the extracellular environment yields more biologically relevant information about these compounds, including the final bioactive form released by the cell for signaling. Tissue-based sampling, on the other hand, utilizes homogenization, a laser beam, or detergents to disrupt cell membranes, and thus the NPs collected represent not only those released in response to a particular stimulus, but those that are being translated, processed, or stored inside the cell. These compounds’ sequences may not represent the final bioactive form, but some prehormone along the post-translational processing pathway. Tissue-based sampling techniques sample inactive prepro- and pro-peptides, stored peptides, and released peptides, whereas liquid-based techniques only sample those NPs released into the extracellular space. In addition to revealing the active form of the NPs, sampling from the extracellular environment also provides more insight into the regulation of NP release by a certain physiological process. Tissue-based techniques can only detect net changes in the NP content of the cell, which are due to the sum of NP release and changes in the amount of pre-prohormone translation. It is thus difficult to tell from tissue sampling by what mechanism the NP is regulated in response to a physiological change. In contrast, the only factors that influence NP concentration in the extracellular fluid are release from cells and degradation over time, which is 31 usually a zero-order process not tied into the animal’s response to stimuli. By sampling from the hemolymph, we are better able to determine if a NP is released by cells in response to a certain change. Not only does fluid sampling improve the quality of data obtained about NP release, but it permits multiple samples to be taken from a single animal. This decreases problems with interanimal variability in the study of dynamic physiological processes. Neuropeptides are known to be involved in both rapid synaptic communication and long-term physiological modulation, and thus the temporal course of their release is of great importance when attempting to determine their function. A great deal of inter-animal variability is observed in baseline NP concentrations, both in tissue extracts and liquid samples. Comparing the net change in NP concentration in a single animal provides more relevant information about NP function. Baseline concentrations of NPs can change several-fold between individual invertebrates. The change in NP concentration in response to a stimulus is often 1-2 fold, and thus lies within the inter-animal variability if different animals are used for each time point around the stimulus. Sampling from the same animal multiple times makes it possible to see NP concentration changes that would otherwise be lost in the inter-animal variation at each time point. Liquid-based sampling has the distinct advantage of allowing for repeated sampling from the same animal to observe changes in NPs that are obscured due to high baseline variability in NP concentration between individuals. For these reasons, liquid-based sampling is advantageous when studying rapid changes in NP concentration associated with function. 2.3.3.3.2. Advantages of tissue-based techniques Tissue-based techniques have the advantage, however, when distribution of an NP is of interest. Due to the open circulatory system of most invertebrates, NPs that are present there are 32 not localized to any one region of the body. Any association with a particular organ or subsection of an organ is lost when sampling from the hemolymph. With tissue-based techniques, the localization of NPs throughout the nervous system can be determined. This can be important in well-defined nervous systems, such as ganglia with a known function (i.e. the pericardial organ as a neurosecretory organ), subsections of ganglia associated by connectivity with a certain function (i.e. the antennal lobe in the crustacean brain), or in a ganglion for which the connectivity of each neuron in a circuit is known (i.e. the stomatogastric ganglion of the crustacean). In addition, the local neurotransmitter roles of some NPs will not be reflected in hemolymph concentrations of these compounds, as they will be present only at synapses in the tissue. Any NP released at a synapse is not likely to diffuse into the hemolymph in amounts that can be detected with precision. When the localization of an NP throughout the body of an invertebrate is important, tissue-based techniques are preferred over fluid-based sampling. 2.4. Bioinformatics Tools for Discovery of Invertebrate Neuropeptides 2.4.1. Peptide Mass Matching for NP Discovery Once spectra have been acquired, they must be interpreted to determine the identity of the compounds present. The simplest way to identify neuropeptides from MS spectra is by searching experimental masses against a known neuropeptide database. This can be achieved by comparing with a list of known neuropeptide masses via manual comparison or by automated search. Public neuropeptide information databases are currently of great use, such as SwePep (http://www.swepep.org/) [70] and the Endogenous Regulatory OligoPeptides (EROP) database (http:// erop.inbi.ras.ru./). SwePep is designed for identification of mouse endogenous peptides by their mass based on Uniprot annotation, although it is no longer updated regularly. EROP database contains the known bioactive peptides from information of the literature. This type of 33 analysis is analagous to peptide mass fingerprinting of a digested protein, a commonly used approach in proteomics. As mentioned previously, neuropeptide prohormones are processed into multiple gene products by the actions of a number of enzymes. Bioinformatics tools have been developed to aid in predicting the final peptide forms from a prohormone; these tools use binary logistic regression models trained on known neuropeptides to identify cleaved basic sites among the many possible cleavage sites that exist in a prohormone [71, 72]. Thus, one can make wellinformed decisions on the expected neuropeptides from a novel prohormone. These prediction tools are accessible via http://neuroproteomics.scs.uiuc.edu/neuropred.html [73], which also contains lists of prohormones and their associated peptides for a variety of common neuronal models. As described above, there are two common methods of performing MS analysis of brain tissues — direct profiling via MALDI and ESI-MS/MS of extracts. The direct profiling of a cell or a tissue has an advantage in that many of the peptides from a specific prohormone are detected at the same time. When multiple peptides are detected using accurate mass, even if MS/MS cannot be obtained, the ability to confidently assign the peaks is enhanced. Single cell MS is an extreme example where often a significant fraction of a prohormone sequence can be covered with accurate mass MALDI MS measurements. 2.4.2. Strategies for MS/MS Sequencing of NPs For extracts or larger brain regions, one gains the ability to work with more complex samples but loses the advantage of prohormone coverage. In such cases, the ability to sequence novel neuropeptides can be critical. Neuropeptide sequencing requires an instrument capable of producing fragment ions (via MS/MS). Depending on instrument availability and the species of 34 interest, a number of different data analysis strategies can be employed for MS/MS neuropeptide identification. First of all, if the species of interest has a sequenced genome, a standard protein database search can be performed using proteomic-based search engines, such as Mascot (Matrix Science, http://www.matrixscience.com/) or SEQUEST (Thermo Corp., http://www.thermo.com/) [74]. Although these search engines were designed for proteomics applications, they are easily adapted to neuropeptidomics. The proteins in the database are virtually digested into peptides. Then the fragmentation spectrum is compared with these virtual peptides. The software can also take into account PTMs and selects the most likely peptide using a scoring function. The problem with this method is the lack of a virtual enzyme that has exactly the same cleavage pattern as the protein convertases that cleave neuropeptide precursors. Considering all possible cleavage sites and PTMs is thus required, although it increases the false positive rate, prolongs the search time, and reduces the identification confidence. If the species of interest does not have a sequenced genome, de novo sequencing of the neuropeptide is usually required. In contrast to database searching, de novo peptide sequencing uses no a priori information, and instead relies upon direct ‘reading’ of the amino acid sequence from the MS/MS spectrum. With a great deal of experience, de novo sequencing can be done manually. However, this is extremely labor intensive and time consuming. Fortunately, several software packages exist which perform de novo sequencing directly on MS/MS data, including LuteFisk (http://www.hairyfatguy.com/lutefisk/), PepSeq (packaged with Waters’ MassLynx software, http://www.waters.com/), Mascot Distiller (Matrix Science), PEAKS (Bioinformatics Solutions, http://www.bioinformaticssolutions.com/), (http://proteomics.ucsd.edu/Software/PepNovo.html), and others [75]. PepNovo A number of these 35 programs, such as PEAKS and Mascot Distiller, include automated or batch de novo processing to enhance sequencing speed and flexibility. Another way to identify neuropeptides employing a database is by sequence tag searching. Finding the complete sequence from fragmentation spectra is only possible when spectra of high quality have been obtained. In contrast to Mascot and SEQUEST, which base identifications on parent masses and MS/MS fragmentation patterns, sequence tag searches are performed using small contiguous strings of identified amino acids (sequence tags) compared against sequences in the database [76]. Even if the exact protein is not present in the database, there is a high likelihood that homologues of the protein can be found. Some sequence tagging programs can handle homologue mutations or possible sequencing errors. Since this technique does not rely on parent mass identification, it is a powerful technique for identifying NPs containing PTMs. For example, MS-Seq (Protein Prospector, http://prospector.ucsf.edu/) and Mascot offer sequence tag searching for single tag queries. Both of these interfaces are flexible and powerful but can be time consuming if analyzing a large volume of data. 2.4.3. Verification of MS/MS Sequencing for NPs Basic Local Alignment Search Tool (BLAST) homology search (http://www.ncbi.nlm.nih.gov/BLAST/) can be also performed to confirm the identification of neuropeptides from de novo sequencing. A BLAST search compares a partial sequence against the database of a closely related species. While a homology search rarely yields a complete sequence, it can provide useful information about evolutionary origins and potential function of the partially sequenced SP. More recently, the use of pattern finding software, such as SPIDER (Bioinformatics Solutions) and MEME (http://meme.sdsc.edu/meme/) has improved partial sequence homology searches [77]. These programs are optimized for MS/MS derived data and 36 are more tolerant to sequencing errors than BLAST searches. We certainly expect that bioinformatics tools tailored to the unique features of NPs will continue to expand. Every sequencing method can introduce faults. It is argued that the combination of multiple search methods can be used to reduce the false positives and increase the coverage and confidence for peptide identification, since different search engines have different criteria of interpretation, and if two or more independent methods produce the same result, then it is more likely the result is accurate. The combination of Mascot and SEQUEST has been demonstrated to improve the sensitivity and accuracy of protein identification in shotgun proteomics experiments [78]. Indeed, more and more software programs are beginning to incorporate this consensus functionality. For example, the most recent PEAKS software package incorporates five different search engines, including Mascot, SEQUEST, OMSSA, X!Tandem, and PEAKS Protein Identification. The software can combine the results to generate a unique report, which provides better coverage and accuracy for peptide and protein identification. 2.4.4. Genome Mining for NP Discovery in Crustaceans Recently, a number of cDNA libraries have been constructed for crustacean species with the goal of allowing for gene-based studies of their physiology. For many of these libraries, expressed sequence tags (ESTs) have been generated and submitted to publicly accessible databases [79, 80], creating a rich resource from which to mine potential proteins of interest. In silico analysis methods have been established to discover novel neuropeptides from these databases. These methods use known prepro-hormones or known peptide sequences as queries to search for transcripts putatively encoding neuropeptide precursor proteins using the BLAST program tblastn (search of translated nucleotide database using a protein query). The transcripts thus obtained are subjected to processing via several on-line protein programs to predict the 37 mature forms of the peptides contained within them, which are then verified via homology to known peptide isoforms. This strategy has been used successfully for peptide discovery in insects [81-83], and has been used to study neuropeptides in crustaceans. For example, in a recent study, 12 unannotated ESTs encoding putative neuropeptide prepro-hormones were identified from six different species [84]. Predicted processing of the encoded precursor proteins revealed over 60 putative neuropeptides. The use of in silico analysis can also help to resolve the ambiguity of MS/MS data interpretation for de novo sequencing. For example, it permits assignment of leucine and isoleucine in amino acid sequences, which cannot be conducted with MS/MS due to the fact that these two residues are isobaric, and it also can confirm previously discovered neuropeptides. With the continued addition of new ESTs to publicly accessible databases, undoubtedly other neuropeptide precursors will be included among them, and as additional in silico searches are conducted, more neuropeptides will be discovered, providing a foundation for future molecular, mass spectral and physiological investigations. Recently, the first crustacean genome was published for the Cladoceran Daphnia pulex [2]. A number of papers have used genome mining in combination with biochemical and mass spectrometric techniques to characterize NPs present in D. pulex. Neuropeptides from multiple families have been identified from genomics information [85-87]. In some cases, immunohistochemistry has been used to verify the presence of a certain NP in this animal [87]. Further studies have verified NPs predicted from genomic analyses by MS/MS [86]. Finally, the a putative red pigment concentrating hormone (RPCH) identified from the D. pulex genome has been synthesized and used in a pigment dispersion assay in another species to determine how its activity varies from forms of RPCH previously identified in other crustaceans [88]. Nearly every family of crustacean neuropeptide has been identified from genome mining in D. pulex. 38 However, some remain elusive. Further refinement of genome searching algorithms may provide additional information about those NPs not yet identified from the genomic information. In addition, the sequencing of additional crustacean genomes will be a great boon to NP discovery. Although Daphnia is an important crustacean genus in environmental assessment, it is of limited use in understanding neuropeptide function due to its small size and limited set of behaviors. Sequencing the genomes of decapod crustaceans, where more is already known about NP sequences, would be of great utility. 2.6. Known Crustacean Neuropeptides A large number of NPs have been discovered and characterized in many crustacean and other invertebrate species. The following section will discuss current knowledge about the identities of these NPs, their distribution throughout the body, physiological functions of these NPs, and molecular mechanisms by which they act. Due to the large number of NPs identified and the wide variety of decapod crustacean species studied, this section cannot contain all current knowledge, but an attempt has been made to include the most influential and/or recent findings. NPs are often classified into families of similar compounds based on shared amino acid sequence motifs, and each family will be discussed separately. 2.6.1. RFamide-Related Peptides The first RFamide peptide—and the first identified invertebrate neuropeptide, FMRFamide, was characterized from the mollusk Macrocallista nimbosa in the late 1970s by Price and Greenberg using multiple chromatographic separations followed by Edman degradation and various biochemical techniques [89]. It was initially recognized for its cardioexcitatory/myoactive activity, determined in functional assays where either natural or synthetic FMRFamide was applied to the isolated ventricle of Mercenaria mercenaria or on the 39 radular protractor muscle of Busycon contrarium [89]. In the following years, a large diversity of FMRFamide related peptides (FaRPs) possessing arginine (R) and amidated phenylalanine (F) as a C-terminal motif have been found throughout the invertebrate animal kingdom, observed in Nematoda, Annelida and Arthropoda, among others. RFamide peptides show a remarkable array of physiological functions, including as the well-known cardiovascular activity, along with modulation of sensory organs, motility, reproduction, and feeding. The diversity of possible functions is likely a consequence of the large diversity in their amino acid sequences. Since the first identification of FMRFamide in M. nimbosa, around 30 isoforms of FMRFamide related peptides (FaRPs) have been discovered in several major classes of mollusks using both traditional biochemical methods and mass spectrometric methods. A recent review includes most of the progress in FaRP characterization in the phylum Mollusca [90]. FaRPs have also been extensively studied in nematodes, especially Caenorhabditis elegans. The completion of the C. elegans genome sequencing project was achieved in 1998, and the complexity of NP signaling in this simple nematode is illustrated in studies mining this genome. Overall, 23 genes encoding FaRPs (as known as flp genes) were initially found in C. elegans [91, 92]; and a total of 59 distinct putative FaRPs can be predicted from processing of these 23 genes, as multiple mature FaRPs forms can be produced from each of the propeptide precursors. By systematic BLAST searching of the nematode EST database, conducted by McVeigh et al., the total number of identified nematode flp genes has increased to 31. The study by McVeigh and colleagues predicted an additional 25 novel distinct FaRPs from these findings [93]. A recent twodimensional LC–MS/MS study analyzed whole body extract from C. elegans of multiple developmental stages [94]. This work identified 25 putative FaRPs and characterized several common posttranslational modifications, including amidation, pyroglutamic acid C-termini, and 40 methionine oxidation [94]. Other animal models from the phylum Annelida have also been analysed for FaRP content, such as the earthworm [95] and leech [96]. 2.6.1.1. FMRFamide-Related Peptides in Crustaceans The FaRPs are also present in crustaceans as one of the families with the most diverse amino acid sequences. In the first study of these peptides in the crustacean, immunocytochemistry of eyestalk tissue from the prawn Palaemon serratus revealed that this tissue contains a compound that reacts with a polyclonal anti-FMRFamide antibody [97]. Through the following two decades, using similar techniques, FaRPs were detected in numerous crustacean species, including Cancer borealis, Panulirus interruptus, Homarus americanus, and Procambarus clarkii. FMRFamide-like immunoreactivity has been observed in all major neuronal organs in the crustacean, including the central nervous system (CNS), the STNS, and the neurosecretory organs: pericardial organ (PO) and sinus gland (SG). However, accurate characterization of FaRPs in the crustacean was hindered due to the difficulty of performing accurate and high throughput sequence analysis. At the beginning of the 21st century, only a few FaRP isoforms had been characterized using the microsequence analysis technique (Edman degradation) [98-100]. In recent years, with the development of MS techniques, a large number of FaRPs have been discovered, revealing the high complexity of this peptide family in crustaceans, similar to the observed complexity of the flp genes in C. elegans. For example, in a recent neuropeptidomic study of C. borealis, Ma and coworkers identified 41 FaRPs along with more than 100 neuropeptides from other families [59]. Based on several largescale mass spectrometric neuropeptide characterization studies, the number of FaRPs identified in the crustacean has greatly increased in recent years. [57, 59, 101-103]. These peptides can usually be divided into three different groups based on their C terminal motif, including – 41 FLRFamide, -YLRFamide, and –RLRFamide, with distinct localizations and physiological functions. FaRPs havealso been extensively studied in insects, including the cockroach Periplaneta americana [104], the grey flesh fly Neobellieria bullata [105], andthe fruit fly Drosophila melanogaster [106]. The division of insect FaRPs into subfamilies follows different rules than in the crustacean. FaRPs in insects are divided into four subfamilies, including -FMRFamide containing peptides, myosuppressins (XDVXHXFLRFamide), sulfakinins (-DYGHMRFamide, Y represents a sulfated tyrosine) and short neuropeptide F (SNPF, -RLRFamide). Some of those peptides are remarkably conserved between insects and crustaceans. For example, sulfakinins [107] and SNPFs have both been isolated and characterized in several crustacean species in addition to the insect species in which they were originally described. Furthermore, the – FLRFamide containing peptides in crustaceans show great structural similarity to insect myosuppressins. It is important to note, however, that peptides containing the exact sequence FMRFamide have not been observed in crustaceans as yet, despite the large number of FaRP family members described in these species. 2.6.1.2. Functions of FMRFamide-Related Peptides As described above, FaRPs are present in many different organisms with a large diversity of amino acid sequences. These peptides also exert a large array of physiological functions, from modulating synaptic transmission within neural circuits to acting as hormones in the circulating fluid to affect behaviors. Many studies have shown that FaRPs are vital in several major aspects of the animal’s life, such as reproduction and feeding, and these functions are often conserved between different organisms at different evolutionary levels. Several articles have reviewed their functions specific to certain phyla or organisms, such as mollusks [90], insects [108], nematodes 42 [109]and crustaceans [110]. A general and cross-species discussion will be given in this paper regarding the most recent progresses. The first isolation and characterization of FMRFamide arose from an investigation into its cardioactivity in mollusks, and thus members of this family are known primarily as cardioactive peptides. The regulatory functions of FaRPs on cardiac outputs have been well documented in many different organisms. They can also modulate the contraction of many other types of smooth muscle tissue tissue in diverse species. For instance, FaRPs have been shown to modulate the alimentary tract and hindgut ganglia of B. amphitrite [111], pharyngeal motility in H. medicinalis [112], and crop movements in D. melanogaster [113]. FaRPs are also known to positively and negatively regulate the reproductive axis in vertebrates by modulating the gonadotropin-releasing hormone (GnRH) neurons. Different FaRPs stimulate (kisspeptin) and inhibit (gonadotropin-inhibiting hormone (GnIH)) this axis [114]. A great deal of evidence has also demonstrated that FaRPs play a regulatory role in reproduction in invertebrates as well. In Octopus vulgaris, the short peptide FMRFamide was observed to modulate the effect of dopamine on photoreceptors in animals adapted to light or dark conditions. In both light- and dark- adapted animals, dopamine + FMRFamide caused the retinae to quickly adapt to light exposure, which has the overall effect of inhibiting sexual maturation. [115]. Several FaRPs, including FMRFa, FLRFa, FIRFa, and an extended FLRFa, have also been found to play a role in peptidergic control of egg-laying in the cephalopod Sepia offcinalis by regulating oocyte transport through the oviduct [116]. Recently, it was demonstrated that mutations in the G protein coupled receptors for certain FaRPs in C. elegans cause inhibition of egg-laying [117], again perhaps due to action on muscles in the oviduct, or more direct functions on the reproductive axis. 43 Another important role identified for FaRPs in several species is to regulate feeding. In crustaceans, –FLRFamides are able to modulate the rhythmic contractions of several parts in the digestive system. In 1990, a cholecystokinin (CCK)-like hormone (vertebrate homolog of sulfakinin) was found to activate the gastric mill rhythm in the lobster, levels of a peptide that reacted with anti-CCK increased in the hemolymph following feeding, and a CCK antagonist inhibited feeding-induced gastric mill rhythms [118]. Furthermore, Mercier and colleagues detected increased FaRP immunoreactivity in the hemolymph of crayfish 1 hour following feeding [110]. Additional support for the role of FaRPs in feeding regulation comes from evidence that FaRPs modulate the rhythmic outputs of several food intake and digestion-related central pattern generators in Aplysia [119]. Studies in decapod crustaceans and the mollusk Aplysia suggest a role for FaRPs in feeding. A number of studies in insects also support a role for FaRPs in feeding behaviors. Hill and Orchard reportedthat under different diet and feeding states the expression level of FaRPs in cockroach gut tissue changed [120]. In addition, different FaRP family peptides occasionally affect the visceral muscles in ways that are opposite but are necessary for overall food processing. For instance, insect sulfakinins stimulate the foregut muscles, and myosuppressins inhibit spontaneous contractions of the foregut. However, both sulfakinins and myosuppressins inhibited feeding in vivo [121-123]. The role of FMRFamide-related peptides in feeding is complex and may be due to their more well-known myoactive properties. It is clear, however, that FaRPs play some role in feeding in several invertebrate species, including insects, crustaceans, and Aplysia. Other putative functions for FaRPs include regulation of reproduction, which is more commonly seen in vertebrates but has also been observed in two cephalopod 44 species and one nematode. The canonical function of FaRPs is muscle stimulation, which has been demonstrated on the heart and several portions of the digestive tract. 2.6.1.3. Molecular Mechanism of FMRFamide-Related Peptide Action It is well known that the activity of most FaRPs is mediated mostly through G-protein coupled receptors (GPCRs). However, studies of neurons in mollusks revealed that FMRFamides cause a fast depolarizing response by activating a sodium channel on the cell membrane. The observation of FMRFamide-induced inward currents when the GPCR-blocking agent guanosine was present indicates that a FMRFamide-gated ion channel exists. First cloned from the snail Helix, this FMRFamide-gated sodium channel (FaNaCh) has also been observed in several other species reviewed in [124, 125]). So far, FaNaCh is the only known ionotropic receptor that is regulated directly by peptides in invertebrates. Interestingly, a vertebrate cationic channel gated by the FaRP kisspeptin has also been found, and it is a member of the transient receptor potential channel type C (TRPC) family [126-128]. Direct control of ion conductance by NPs is rare, and it may be significant that vertebrate and invetertebrate FaRPs both act via this mechanism. The physiological implications of FaNaCh are still elusive and further study will be necessary to determine the significance of this nonclassical mechanism of NP action. A crustacean GPCR for FaRPs has not yet been identified, and thus its molecular action through this receptor is also currently unknown. 2.6.2. Allatostatins 2.6.2.1. Types of Allatostatins Found in Crustaceans The allatostatin (AST) peptides are named for their first identified function. In insects, ASTs inhibit juvenile hormone synthesis in the corpora allata [129]. Many ASTs have since been identified in various invertebrates [101, 130, 131], and they are now identified as allatostatins by 45 sequence homology to the first described ASTs. Three main groups exist, based on sequence tags of peptides that modulate the corpora allata in different insect species. A-type allatostatins possess the common C-terminal pentapeptide motif Y/FXFGL-NH2 and were first identified in the cockroach [129, 132]. B-type ASTspossess the C-terminal sequence W(X)6Wamide, with X being variable amino acids and were first found in the cricket [133]. Finally, C-type ASTs possess a nonamidated, conserved C-terminus –PISCF and were first isolated from Manduca sexta and Lepidoptera [134]. Several reviews have presented the structures of these peptides, their allatostatic and other functions, and the organisms in which they have been found, especially focusing on insect species [135-138]. It was long believed that only A-type ASTs were present in crustaceans. The first crustacean A-type AST was found in the crab Carcinus maenas [139]. Recently, more than 100 crustacean A-type allatostatins have been reported using both biochemical methods and mass spectrometric techniques in multiple crustacean species [32, 57, 59, 101, 102, 130, 140, 141], showing a large degree of sequence diversity. Recently, a gene encoding this peptide family was cloned in the freshwater prawn, Macrobrachium rosenbergii. This gene encodes a massive polypeptide precursorcontaining 701 amino acid residues. This prepro-AST is capable of producing 35 potential mature A-type AST peptides [142]. In keeping with the A-type AST family sequence tag, these peptides contain YAFGL as the primary C terminus. Using state-ofthe-art mass spectrometry, the concept of only A-type ASTs being present in crustaceans was refuted, as a B-type AST was identified in Cancer productus POs [101]. Following this discovery, additional B-type AST isoforms were identified in numerous crustacean species [59, 143]. The existence of non-A-type ASTs in crustaceans was further supported by recent discoveries of two C-type AST isoforms [64, 144]. In contrast to the diversity of A- and B-type 46 AST isoforms, there are usually only one or two isoforms of C-type ASTs found in each organism, similar to insects. The variation in the primary sequences of ASTs may result in different physiological activities. Immunohistochemistry studies show that ASTs are widely distributed in both the CNS and stomatogastric nervous systems (STNS) of various crustacean species [130, 145, 146]. Additional research has mapped ASTs throughout several neuroendocrine structures, including the pericardial organ, sinus gland and perineural sheath of the thoracic ganglia [140, 146, 147]. Finally, ASTs have also been detected at relatively low concentrations in the hemolymph [67, 68, 148]. These observations suggest ASTs can function via both neural and humoral pathways, with both neurotransmitter and neurohormonal roles. 2.6.2.2. Allatostatin Function in Crustaceans The signature role of ASTs in insects is regulation of development and reproduction by inhibiting juvenile hormone (JH) synthesis in the corpora allata (CA). In crustaceans, the mandibular organs (MO), located at the bottom of the mandibular tendon, are homologous to CA. MO can produce the sesquiterpenoids methyl farnesoate (MF) and its immediate precursor, farnesoic acid (FA). MF is a precursor to insect JHs that may serve the same role in crustaceans as JH does in insects. It has been reported that treatment with Dippu-ASTs on isolated MO from adult crayfish, Procambarus clarkii, increased MF synthesis by MOs that were not actively producing large levels of MF [148]. This study goes on to show that ASTs do not affect FA levels, and thus the authors suggest that ASTs act on the enzyme that converts FA to MF. Observations in this study are contradictory to the canonical role of ASTs in inhibition of the CA in insects. However, non-classical roles for ASTs in insects are similar to the observed increase in MO activity seen in this study. As yet, there is no evidence that suggests ASTs act as inhibitors as well as stimulators of MF synthesis in crustaceans, however very few studies have 47 been performed and only A-type AST has been tested. Further investigation is required to fully understand the physiological functions of ASTs on regulation of sesquiterpenoid biosynthesis in crustacean and arthropods in general. The currently accepted nomenclature of allatostatin is based on the initial discovery of one function of these groups of peptides in insects–the inhibition of juvenile hormone biosynthesis by the corpora allata. However, ASTs also have numerous other functions, including activity modulation in the central pattern generator (CPG) networks of the crab nervous system. In an isolated STNS preparation, Diplotera allatostatins were able to reduce the pyloric rhythm frequency. This was dose-dependent and the amount of reduction was related to the initial frequency [145]. Although the C-terminal sequences of the three subtypes of ASTs vary greatly, they have been shown to act in similar ways on the STNS [64, 143]. These Cterminal motifs may also be the most functionally relevant parts of the ASTs, as the C-terminal pentapeptide of A-type ASTshas been shown to produce activity at its receptor [149]. The action of ASTs on electrically excitable cells has also been demonstrated using A-type ASTs on the neuromuscular junctions of several crustacean species. The application of these compounds led to reductions in the amplitude of signals transmitted and the movements thus elicited [150, 151]. A-AST has also been shown to induce action in the cardiac ganglion (CG) CPG, responsible for contraction of the heart. In isolated CGs from C. borealis, it strongly inhibits the motor output[152]. This finding demonstrates yet another CPG in which A-ASTs reduce activity, leading to the hypothesis that A-ASTs may be important chemical messengers for reduced activity in the heart, in other neural circuits, and in excitable cells in general. A contradictory result was obtained from application of C-type AST to H. americanus heart preparations. The amplitude of the heartbeat was instead increased in a state-dependent manner. This may indicate 48 that A-and C-typeASTs have opposite effects on heart function. A-type ASTs have also been shown to alter sensory neuron activity [153]. Finally, an inhibitory endocrine role for ASTs on the rhythmic hindgut contractions in many arthropod species has been described [154-156]. ASTs have also been detected in the gut tissue in crustaceans [142, 157], however their physiological activities have not been tested. In general, it appears that AST-type peptides inhibit excitable cells throughout the bodies of invertebrates. It should be noted, however, that most studies investigate A-type ASTs, and there is some evidence that different AST subfamilies may have opposing functions. 2.6.2.3 Molecular Mechanisms of Allatostatin Action Allatostatin receptors are GPCRs similar in structure to those of the mammalian somatostatin/galanin/opioid receptor family [158]. A unique receptor family has been identified for each of the A-, B- and C-type AST subfamilies in Drosophila [159]. The first identified AST receptors were found in Drosophila, which carries two genes that encode GPCRs for A-type AST peptides. Both of these A-AST receptors are members of the rhodopsin-like GPCR family, whose closest vertebrate homolog is the galanin receptor family [158, 160]. B-type AST receptors and vertebrate bombesin receptors appear to be most related [161]. Due to the presence of a disulfide bridge at the C-terminus of C-type ASTs, they are thought to be homologous to somatostatin. Additional support for this homology comes from the similarity between their receptors [162, 163]. Due to the specificity of different AST subtypes for different receptor families, functional similarities in AST actions are more likely to occur in downstream signaling molecules. Drosophila AST activates G protein-coupled inwardly rectifying potassium (GIRK) channels [158]. This decreases cellular excitability by hyperpolarizing neurons and reducing their input resistance [163, 164]. This reduced excitability is a possible explanation for the 49 inhibitory actions of A-, B- and C-type ASTs on rhythmic activity in the STG. Because GPCRs are commonly desensitized following prolonged activation, typically by phosphorylation or internalization [165], long-term exposure to a single type of AST might eventually lead to a loss of efficacy. Release of another form of AST could continue to propagate the signal, as it would act upon different receptors, but have similar downstream targets. This may be an explanation for the diversity of AST isoforms present and their similar release profiles. AST peptides appear to have distinct receptor types for each subfamily, but in general lead to reduced excitability. This divergence of receptors but convergence of effector proteins along with the diversity of ASTs present but similarity in their release and functional relevance may constitute a method to provide continuous signaling even in the presence of receptor desensitization. 2.6.3. Tachykinins and related peptides The tachykinin NP family was first identified in vertebrates. They can function as brain/gut peptides that affect both central and peripheral tissues. Some well-known mammalian tachykinins include substance P and neurokinins, which possess the C-terminal motif of FXGLMa. The first invertebrate tachykinin Lom-TK-I was characterized from brain of Locusta migratoria. By the early 2000’s, around forty invertebrate tachykinins (Inv-TKs) and tachykininrelated pepides (TKRPs) had been characterized from a wide range of invertebrates, including insects, crustaceans, worms and mollusks. Some earlier studies were reviewed in articles from Severini et al. and Satake et al. [166, 167]. More recent studies, particularly those in crustaceans, will be the focus of this review. 2.6.3.1. Tachykinins and Related Peptides in Invertebrates Most TKRPs contain the primary sequence motif –FXGYRa. Using mass spectrometric techniques, several novel TKRPs have been recently identified. In insects, TKRP is a large 50 family with numerous isoforms. A number of studies have identified these peptides in various Heteropteran species. In one such study, six different TKRPs were de novo sequenced from the antennal lobes of seven different species, five of the Pentatomoidea superfamily (shield and stinkbugs) and two others. The five Pentatomoidea species studied—Nezara viridula, Euschistus servus, Acrosternum hilare, Banasa dimiata, and Pentatoma rufipes—share the same TKRPs, whereas the other two more distantly related species—Oncopeltus fasciatus and Pyrrhocoris apterus—have distinct TKRPs [168]. Studies in two cockroach species identified two TKRP precursors that encode all of the nine previously reported LemTRPs in addition to four novel ones [169]. A study in the honey bee, Apis melifera, was able to definitively identify 4 different TKRPs by using a combination of genomic database mining and mass spectrometric analysis [170]. Typically, insect species have many different TRP isoforms. Crustacean tachykinin related peptides (CabTRPs) were first identified in the CNS of the crab C. borealis. In these seminal studies, two forms were identified: APSGFLGMRa (CabTRP 1a) and SGFLGMRa (CabTRP 1b) [171]. CabTRP 1b is the less potent of these two in a cockroach hindgut contraction bioassay. The 1b isoform does not alter the pyloric rhythm of the STG, whereas CabTRP 1a does. CabTRP 1a is released from the STG under normal physiological conditions in Cancer borealis [172]. It was also found in the anterior commissural organ in Cancer productus, using both immunohistochemical and mass spectrometric methods [173]. Using direct tissue analysis, several CabTRPs have been detected from midgut tissue in three closely related crustacean species, Cancer magister Dana, Cancer productus Randall, and Cancer borealis Stimpson. In the same study, the midgut epithelial endocrine cells were found to be a rich source of CabTRPs [174]. Recently a novel TKRP, TPSGFLGMRa, was identified inthe midgut and STNS of Cancer irroratus, C. borealis, C. magister, and C. productus. 51 Notably, it was not detected in Panulirus interruptus and Procambarus clarkii. The application of this peptide to the C. borealis STG elicited an identical pyloric motor pattern as CabTRP 1a [60]. This novel peptide was later detected in the American lobster Homarus americanus [175]. CabTRPs have also been detected in the hemolymph of C. borealis, which hints at a possible hormonal role for this peptide family [67, 174]. In a recent large-scale MS neuropeptidomics investigation of C. borealis, TPSGFLGMRa and its methionine oxidation-modified form were detected in the CNS and STNS [59]. Methionine oxidation is thought to be a modification that occurs during sample handling, not in vivo. Overall, only three CabTRPs and their posttranslationally modified peptides have been discovered in different crustacean species so far in contrast to the large numbers of isoforms observed in insect species, but they are found extensively in the CNS, STNS, midgut, and hemolymph; and at least two of these isoforms have identified functions. 2.6.3.2. Functions of Tachykinin and Related Peptides The mammalian tachykinin peptides, such as substance P and neuropeptide K and many others, are known to have a diverse range of biological functions, including but not limited to transmission of pain signals, various facets of reproduction, inflammation, immune system function, and food intake [176, 177]. In invertebrates, the TKRPs are found in both nervous tissue and digestive tissue, and many have been shown to modulate gut function, as described in several reviews focusing on insects [108, 178]. The stimulatory effects of TKRPs were first demonstrated in the hindgut of the cockroach, and TKRPs have since been shown to produce visceral muscle contraction in other insects. It has also been reported that Lom-TKs can cause contraction of the oviduct in L. migratoria [108]. Electrophysiological experiments on the crab STNS have found that application of CabTRP Ia elicits the pyloric rhythm or increases several 52 aspects of this rhythm in preparations where the rhythm is already active [171]. Therefore, tachykinin-related peptides may play an important role in feeding and/or digestion in the crustacean. A cardiac effect of TRPs is also suggested by evidence that CabTRP 1a increases the burst frequency and duty cycle in the cardiac ganglion [152]. Immunohistochemistry studies have documented the expression of TKRPs in the olfactory lobes of both insects and crustaceans in detail. Winther et al.reported that TKRPs affect odor perception and locomotor activity in Drosophila [179]. In this work, it was demonstrated that RNAi knockdown of the TKRP prehormone gene altered responses towards specific odorants and different odorant concentrations. Hyperactivity in locomotion assays was also observed in these knockdown flies. Quantitative MS analysis of A. melifera brains following foraging behavior demonstrated dramatic changes in the levels of TKRP expression. This may indicate a role for TKRPs in food collection in social insects, although the importance of olfaction in foraging and previous implication of TKRPs in olfaction may be an alternative explanation for these results [180]. A number of insect studies suggest that TKRPs stimulate food consumption when released from midgut into the hemolymph. This hormonal role for insect TKs has been suggested by the presence of TK immunoreactivity in the endocrine organs of the midgut in Leucophaea maderae [108]. In the cockroach, decreases in LemTRP-1 levels in the midgut were associated with higher levels of food intake. Additionally, starvation led to lower levels of LemTRP-1 in midgut but increased levels in hemolymph [108]. Furthermore, in studies where LemTRP-1 was injected into live cockroaches, orexigenic effectswere observed [181]. These findings suggest that midgut endocrine cells release LemTRP-1 into the hemolymph to increase feeding. 53 In addition to central and peripheral functions in adult animals, TKRPs are also involved in neural circuit development in both vertebrates and invertebrates [166, 182, 183]. As an animal matures, the output of its neural networks changes due to a number of factors. These include changes in the underlying networks, the modulatory inputs to these networks, and/or the effects of neuromodulators on the networks. In the lobster H. americanus, TKRPs are not observed in the STG of embryos and only begin to be observed in later stages, during which adult-like motor patterns also start to occur. TKRPs are expressed much later than other neuropeptide families [184]. A possible explanation for the reduced complexity of STNS output in embryonic lobsters is that not all neuromodulators, in particular TKRPs, are present at early developmental stages. Application of CabTRP 1a to isolated embryonic and adult STNS leads to changes in the STG motor patterns in both stages; however, this effect is different between the two stages. It is therefore likely that the receptor for CabTRP 1a is present in the embryonic STNS, despite the immaturity of the network in total [185]. Production of the NP itself is restricted to developmental stages in which the neuronal connectivity has matured. It is therefore possible that CabTRP 1a production is a signal of a mature neural network. 2.6.3.3. The Tachykinin Receptor The receptors for TKRPs belong to the GPCR superfamily, with the phospholipase C (PLC) -inositol triphosphate (InsP3)-calcium signal cascade as the intracellular mediator of the signal. So far five TKRP receptors (TKRPRs) have been identified in invertebrates: including DTKR (Drosophila melanogaster) [186], NKD (Drosophila melanogaster) [187, 188], STKR (Stomoxys calcitrans) [189], UTKR (Urechis unitinctus) [190], and oct-TKRPR (Octopus vulgaris) [191]. Studies of the structure and distribution of TKRPRs are essential to understand the physiological functions of these neuropeptides. In O. vulgaris the mRNA of oct-TKRPR was 54 detected in both the central nervous system and peripheral tissues, such as several smooth muscles including stomach, rectum, ovary, etc. This pattern is similar to the distribution of TKRPs in other invertebrate animals, and further implicates these peptides in a wide spectrum of physiological processes such as midgut contraction, feeding and reproduction [191]. Further research in crustaceans into the tachykinin receptor and its intracellular signaling pathways may provide additional insight into the importance of tachykinins in crustaceans. 2.6.4. Orcokinins The orcokinins were originally purified from the nervous system of the crayfish Orconectes limosus [192], and it is this species for which they are named. These NPs have been shown to modulate hindgut contraction with high potency, and have been detected by immunochemistry in various neural structures, including the STG, in several crustacean species [193]. The cDNA of two preproorcokinin genes has been isolated from the brain tissue of the red swamp crayfish, Procambarus clarkii. These genes encode 6 unique orcokinins, whose sequences have been verified via mass spectral techniques [194]. These peptides have since been found with a high degree of conservation in other crustacean species using both biochemical and mass spectrometric techniques [54, 57, 59, 102, 195-197]. A recent genomics study used the translated open reading frames of P. clarkii transcripts as queries in searching the available EST database of H. americanus. This led to the identification of five ESTs encoding H. americanus orcokinin orthologs. Further analysis led to the discovery of two distinct prepro-hormones, that, when processed, would generate 13 orcokinin or orcokinin related peptides, several of which were verified in vivo from MS analysis of tissues [198]. Many isoforms of orcokinins have been discovered or predicted in crustacean species. 55 It was not until recently that orcokinins were found in insects. The first insect orcokininlike peptide was found in brain extract from the cockroach Blattella germanica in 2004. However, it did not show activity in a gut tissue assay. Further genomic analysis by the same authors identified putative orcokinin-like peptides in several other invertebrates, including the mosquito Anopheles gambiae and the nematode Caenorhabditis elegans. Several other insect species in addition to A. gambiae were investigated but not found to have orcokinin-related sequences [199]. This finding provided the first evidence that the orcokinin peptide family is present across several phylla, although it does not appear to be universally present in arthropods. Immunocytochemical studies have shown that the orcokinin family is present extensively throughout the nervous system, neuroendocrine structures, and fibers throughout the gut [54, 200, 201]. Orcokinins have also been detected in the hemolymph at an approximate concentration of 10-11 M using an ELISA assay [193]. This finding suggests that orcokinins may be released into hemolymph and thus function as hormones in addition to their roles as locally acting neurotransmitters. Unlike most invertebrate NPs, the orcokinin peptides are neither N-terminally nor Cterminally blocked, with a few exceptions (C-terminal amidation) observed in crustaceans. This is atypical but not unique to the orcokinin NP family [202]. Bungart et al. studied the bioactivity of orcokinin analogues in the hindgut assay, and demonstrated that changes at the C-terminus have a lower impact than changes at the N-terminus in terms of activity [203].The uniqueness of a free N-terminus and the results of these analogue assays indicate that this portion of the orcokinin sequence is vital to its biological activity. Despite the extensive biochemical characterization of the orcokinin family, relatively little is known of their physiological roles. As mentioned previously, the bioactivity of the 56 orcokinins was first demonstrated in the hindgut of the crayfish [192, 200]. In addition, synthetic orcokinin has been applied to the STNS of Homarus americanus, with an alteration in the pyloric rhythm observed [54]. An additional putative role for orcokinin peptides is in circadian rhythms. In the cockroach Leucophaea maderae, the accessory medulla (AMe)—a small neuropil in the optic lobe believed to contain the master circadian clock—has orcokinin-like immunoreactivity [204]. Phase shifts in circadian locomotor activity were observed in the cockroach after injection of Asn(13)-orcokinin near the AMe at different points in the animal’s circadian cycle [205]. Functional roles for orcokinins include modulation of the central pattern generators of the crustacean STNS and regulation of the circadian clock in the cockroach. 2.6.5. Crustacean Cardioactive Peptide (CCAP) Crustacean cardioactive peptide (CCAP) was initially purified from the pericardial organs of the shore crab Carcinus maenas [206]. This peptide has 9 residues and an amidated Cterminus. It also has a disulfide bridge (PFcNAFTGc-NH2, with c indicating a Cys residue that participates in a disulfide bond). These structural characteristics, in particular the disulfide bond, are rarely found in crustacean NPs. After its original identification in C. maenas, CCAP was identified in several other crustaceans [59, 101] with a conserved sequence. CCAP immunoreactivity is present throughout the nervous system in crustaceans, but is found at a higher concentration in the pericardial organ [207-209]. 2.6.5.1. Functions of CCAP in the Crustacean CCAP modulates the heartbeat in a number of crustacean species [152, 206, 210, 211], likely by acting on the cardiac ganglion (CG). The CG is a central pattern generator (CPG) located within the single chamber heart. The CG is a well described CPG, containing five motor neurons, and four pacemaker interneurons. These motor neurons are electrically coupled, which 57 leads to synchronous firing to produce heart contraction [212, 213]. Investigation of exogenous CCAP on the intact working heart, on the semi-intact heart, and on the isolated CG shows that CCAP released from the POs as a neurohormone regulates the crab heart by acting on the heart muscle itself at lower hemolymph CCAP concentrations and on the neurons of the CG, most likely the premotor interneurons, at higher concentrations. Depending on the concentration (and thus the cell type affected), CCAP modulates contraction amplitude and frequency, as well as burst amplitude, duration, frequency, and number of spikes per burst [211]. Several other non-cardiac roles for CCAP have been discovered. In the horseshoe crab, Limulus polyphemus, CCAP was found to act on the midgut [208]. It also has activity in the eye, decreasing the electroretinogram amplitude, and thus the light sensitivity, in the isolated retina of the crayfish Orconectes limosus [214]. Several electrophysiological studies have also showed the effects of CCAP on the crab STNS [215, 216]. Work by Phlippen and colleagues used an immunoassay to determine the CCAP content of hemolymph in O. limosus and C. maenas undergoing ecdysis. CCAP levels were greatly elevated during the active phases of ecdysis— delineated as the stages in which the shell is actually shed, relying upon much movement, including rhythmic muscle movements—as opposed to the passive phases, which are characterized by increased water intake to swell the body to enable shell removal. They thus postulate that CCAP regulates several physiological changes observed in crustaceans around the time of molt, including heart rate and STNS activity changes, in addition to behavioral and muscle activity changes necessary for molting [217]. Although the primary described role of CCAP is on the heart itself, other functions have been observed. It is possible that CCAP coordinates these functions to occur at the same time as alterations in the heart rate as part of important processes in the animals’ life cycle, such as molting. 58 2.6.5.2. CCAP in Other Invertebrates In several insect species, CCAP or peptides with very similar sequences have been isolated and characterized [218, 219]. Many of these CCAP peptides have been discovered from genetic information. In Manduca sexta, the CCAP gene was isolated and determined to encode 125 amino acid residues in a prepropeptide that contains a single copy of CCAP. This CCAP gene is very similar to the CCAP gene found in Drosophila melanogaster [220]. As observed in crustaceans, CCAP causes contraction of the insect heart when applied in vitro [221-223]. Direct activity is also observed on a variety of muscles, and it follows the pattern of other neurotransmitters/neuromodulators. One such example is its activity on the leg muscle of M. sexta; CCAP excites rhythmic contractions [221]. It has also been observed to stimulate hindgut contraction in this species [219, 224]. Furthermore, CCAP can act upon the smooth muscle of the locust oviduct. A dose-dependent increase in contractions was observed, and oviducts of locusts at earlier developmental stages were found to be more sensitive to this effect. Spontaneous and neurally-evoked contractions were both effected [225, 226]. In addition, CCAP-like immunoreactivity has been detected in other smooth muscle neurons, including those in the abdominal ganglion that project to the spermatheca of adult female locusts. The study that identified this immunoreactivity also assessed the effect of CCAP on contractions of the spermatheca, and CCAP increased the basal tonus and frequency of spontaneous contractions, as well as the amplitude of neurally evoked contractions [227]. These data suggest that CCAP is likely to play an important role in reproduction via its activity on smooth muscles in reproductive organs. An additional role for CCAP has been identified in the cockroach, Periplaneta americana, for which the activity of digestive enzymes following food ingestion was increased by CCAP. The authors of that article postulate a system in which CCAP is released by gut 59 endocrine cells in response to food intake, and CCAP alters the efficacy of digestive enzymes [228]. This direct modulation of enzymatic activity is an uncommon mode of action for NPs. A putative CCAP GPCR—CG6111—has been identified in Drosophila melanogaster. This GPCR has sequence homology to the arginine vasopressin (AVP) receptor in vertebrates [229]. Although it is homologous to the AVP receptor, CG6111 is activated by adipokinetic hormone (AKH) and CCAP but not AVP. One CCAP GPCR was cloned from the mosquito Anopheles gambiae, and this receptor was found to be related to receptors for AKH and corazonin [230]. CCAP exists in insects as well as crustaceans, and a great deal of similarity in sequence, localization, function, and receptor type is observed between the two groups. 2.6.6. Corazonin 2.6.6.1. Insect Corazonins Corazonin was first identified from the nervous system and hemolymph of insects. A number of different isoforms of corazonin exist with slight sequence variations. Many of the isoforms appear to have species-specific activities. [Arg7]-Corazonin (pETFQYSRGWTNa) was first isolated from the corpora cardiaca (CC) of the cockroach Periplaneta americana, in which it is a potent cardiac stimulator [231]. The [Arg7] isoform has also been found in numerous other insect species. Another isoform—[His7]-corazonin (pETFQYSHGWNa)—was later isolated from the CC of the locust Schistocerca americana. This [His7]-corazonin also activated the Periplaneta heart muscle [232]. Significnatly, neither isoform altered the heart beat in the locust. The precursor of a third corazonin—[Thr4, His7]-corazonin (pQTFTYSHGWTNa)—was recently cloned from the honey bee Apis mellifera, and the final active form was verified by MS/MS [233]. Additional application of MS techniques to corazonin discovery in insects has yielded three additional isoforms—[Gln10]-corazonin (pQTFQYSRGWQNa), [Tyr3, Gln7, 60 Gln10]-corazonin (pQTYQYSQGWQNa), and [His4, Gln7]-corazonin (pQTFHYSQGWTNa)— from members of Mantophasmatodea and Hymenoptera [234]. [Arg7]-Corazonin was originally described from P. americana but is found in many other insect species. In contrast, the other isoforms are restricted to only a few insect groups or even single species. It is therefore postulated that corazonin modifications appeared late in the evolution of insects [234]. Several corazonin isoforms exist in insects, with some being more widespread than others. Like the presence of [Arg7]-corazinin, the presence of corazonin in the neurosecretory neurons of the CNS of insects seems to be highly conserved [235-240]. Other neurosecretory/neurohemal organs in P. americana contain corazonin, including the head neurohemal system in the retrocerebral complex, and in neurosecretory cells in the brain [34]. These neurosecretory cells project across the glandular part of the CC to reach their release sites. Corazonin has also been identified in the hemolymph of P. americana by immunological methods, which suggests it has a hormonal role [238]. Hormonal function is also supported by the long half-life of [His 7]-corazonin in hemolymph—determined to be 30 min by HPLC and MS analysis. This contrasts with the short half-lives of other neuropeptides, which usually degrade in a few minutes [241]. CCAP localization is highly conserved among insect species, and, along with its presence in the hemolymph and relatively long half-life, this is indicative of a hormonal role. 2.6.6.2. Crustacean Corazonin [Arg7]-Corazonin is the only corazonin isoform that has been identified in crustaceans. It was first found in the pericardial organ (PO) of the crab Cancer borealis using MALDI-TOF, and its sequence was confirmed by PSD fragmentation and by comparison with a synthesized corazonin standard [55]. It was later found in several other crustacean species with a conserved 61 primary sequence [57, 102]. In addition to the PO, it is also present in the CNS and STNS. Its release can be stimulated from the STG both by electrical stimulation of projection nerves and depolarization by K+ [172]. The isoform of corazonin found ubiquitously in insect species is also the isoform found in crustaceans. It is widely distributed throughout the nervous systems of crabs. 2.6.6.3. Functions of Corazonins Corazonin has a variety of functions, most of which have been studied in detail in insects. It can potently activate the isolated hyperneural muscle and the antennal heart of P. americana, but is not active on the oviduct or proctodeum [240]. These effects of corazonin on visceral muscles appear to occur only in Periplaneta species, as muscles in other cockroach species and the heart of Schistocecra americana (as mentioned above) showed little or no sensitivity to corazonin. In addition to its myotropic effects in certain insect species, [His7]-corazonin was found to induce the development of a black pattern in locusts in response to crowding [242]. A similar phenomenon was also observed in the grasshopper Oedipoda miniata, where injection of [His7]-corazonin caused darkening of the integument in a dose-dependent manner [243]. Corazonin’s effect on integument color has also been observed in the crayfish Procambarus clarkii. An interesting additional function for corazonin is revealed by experiments that demonstrated behavioral changes induced by corazonin in Locusta migratoria, Schistocerca gregaria, and Bombyx mori. In particular, it causes these insects to swarm and interact, as opposed to being solitary. The same researchers also found that corazonin changes thorax morphology in these species [244-248]. A final postulated function for corazonin is in the process of ecdysis in the hawk moth. Researchers have demonstrated that corazonin stimulate secdysis-related hormone release from the Inka cells in Manduca sexta [249]. Corazonin has an 62 important role in regulation of muscle excitability, particularly in the heart of Periplaneta cockroaches. It also alters the color of several insects and one crustacean species, although it is not classified with the canonical pigment-altering hormones. It seems to affect behavior and morphology in some insects, and promote ecdysis in one identified species. Many of corazonin’s functions are restricted to certain species, and a general role for corazonin in insects has yet to be determined. [250]. The function of corazonin in crustaceans remains largely a mystery. 2.6.6.4. The Corazonin Receptor Very little is known about the mechanism by which corazonin acts on target cells to produce the results described above. A corazonin-sensitive GPCR has been identified from the genome of Drosophila. It has a high degree of homology to the AKH receptor [229, 251]. A corazonin-sensitive GPCR has similarly been identified from A. gambiae. The Anopheles gambiae corazonin receptor is also related to its AKH receptor and its CCAP receptor [230]. Although very little is known about the molecular targets of corazonins, putative receptors have been identified from Drosophila and A. gambiae. 2.6.7. SIFamides The identification of the first SIFamide peptide was achieved in 1996 by extraction from 350,000 grey flesh flies (Neobellieria bullata) followed by extensive HPLC purification and a combination of ESI-QTOF MS and Edman degradation for amino acid analysis [252]. The Cterminal SIFamide sequence is the identifying characteristic for NPs of this family. Currently, six different isoforms are known from both insects and crustaceans, with high sequence conservation among these species. Like many NPs, the first function discovered for SIFamide was myoactivity. It was observed that SIFamide potently activated the L. migratoria oviduct, but it was found to have no 63 effect on the hindgut [252, 253]. Eventually, targeted cell ablation and RNAi revealed that SIFamide modulates sexual behavior in the fruit fly in a dramatic manner, with flies lacking SIFamide being highly promiscuous [254]. However, very few other studies have been performed to determine the functions of SIFamides in insects. Verleyen et al. recently reviewed several aspects of this peptide family, including progress in identification, localization, and assignment of physiological functions, as well as reviewing the discovery of the SIFamide GPCRs [255]. Originally, crustacean SIFamides were grouped with FMRFamide-related peptides. AntiFMRFamide antibody was observed to react with Penaeus monodon eyestalk extract, prompting further searches for FaRPs in this organ. The eyestalk extract was then analyzed by MALDITOF and Edman degradation to reveal the presence of a SIFamide with primary sequence of GYRKPPFNGSIFamide along with six FaRPs [99]. Later, another SIFamide was found in the lobster Homarus americanus with a sequence of VYRKPPFNGSIFamide [256]. This compound differs only on the first position of the N terminus, and is sometimes called [Val1]-SIFamide compared to [Gly1]-SIFamide. These two SIFamide sequences have since been observed in several other crustacean species, although [Val1]-SIFamide has only been discovered in Homarus spp. and C. maenas [102, 175]. An additional putative peptide with the sequence RKPPFNGSIFamide has been observed in MS analysis of neural tissue extract of several crustacean species [57, 59, 102]; however, it is not clear if this fragment exists in vivo or is present due to degradation. Its activity has also not been tested. Mining of the Daphnia pulex genome has yielded a putative SIFamide with sequence TRKLPFNGSIFamide. A peak with mass matching the theoretical mass for this compound was identified by MS but MS/MS sequence verification could not be conducted [86]. The nomenclature of this peptide is a bit 64 tricky, as it is shortened by one AA at the N-terminal end and has two substitutions—a Thr for a Tyr at the N-terminus and a Leu for the Pro at its fourth residue. MALDI imaging and immunohistochemistry studies have shown that SIFamide is widely distributed in the crustacean nervous system, including both the CNS and STNS [28, 256]. Christie et al. reported that [Gly1]-SIFamide is present in the midgut epithelial endocrine cells in several Cancer species,and is released upon exposure to high-potassium saline [257], which points at a possible hormonal role for SIFamide. Application of synthetic [Val1]SIFamide to the stomatogastric ganglion (STG) increases the amplitude and duration of bursts in the pyloric dilator (PD) neurons, causing an activation of the characteristic pyloric pattern [256]. Both [Val1]- and [Gly1]-SIFamides possess an internal dibasic site, [Arg3-Lys4], which could possibly undergo cleavage during prohormone processing to lead to the isoform PPFNGSIFamide. However, this octapeptide has not been detected via MS analysis of any of the investigated species, and did not produce significant activity changes when applied to the isolated STNS of H. americanus [175]. 2.6.8. RYamides RYamides were first found in the pericardial organ (PO) and PO releaseate (after high-K+ depolarization) from Cancer borealis [55]. Five different isoforms were sequenced in the original study, all possessing RYamide on the C terminus with higher variability at the N terminus. Most of these isoforms along with additional RYamides were later found in other crustacean species, such as Cancer productus [101] and Carcinus maenas [102]. Relatively little is known about the RYamides. Their localization has been mapped by MSI to the anterior bar region of the PO in C. borealis, a pattern not common to other NPs investigated in the study, including A-ASTs and RFamides [13]. In addition to the PO, some RYamide isoforms have also 65 been detected with lower abundance in the brain in several crab species [59]; however none of these peptides have been seen in the lobster H. americanus [57]. When applied to the isolated crab STG, RYamides did not produce overt physiological effects [55]. Therefore, the study of this peptide in other tissues will be required to understand the physiological functions of this peptide. 2.6.9. The CHH/MIH/GIH family The crustacean hyperglycemic hormone (CHH) superfamily is one of the largest groups of neuropeptides identified in crustaceans. Members of this family have been identified in more than thirty crustacean species (Table 2). Typically, these peptides are produced and released from the X organ- sinus gland (XO-SG) complex in the eyestalk, and are known to regulate numerous essential biological processes, including carbohydrate metabolism, reproduction, and molting. CHH-family peptides are also among the largest known crustacean neuropeptides, with mature peptides from this family having a molecular mass of 8–9 kDa. Another conserved feature is the presence of six cysteine residues at similar positions in their sequences, with the potential to form three disulfide bonds. The CHH family is divided into type I and type II peptides, based on their primary structures. The best studied peptides in this family are CHHs, which belong to the type I subfamily. Homology above 60% is observed in the primary sequences of type-I CHHs between different species. The type II subfamily contains NPs with different sequence characteristics from the type I peptides. These type II peptides have a variety of different functions, among them regulation of molting (molt-inhibiting hormone, MIH), reproduction (gonad-inhibiting hormone, GIH, also known as vitellogenesis-inhibiting hormone, VIH), and the varied functions of the mandibular organ (mandibular organ-inhibiting hormone, MOIH). Biochemical and genetic techniques have identified several precursor sequences for the 66 different CHH subtypes. These preprohormones of CHH-family peptides also contain CHH precursor-related peptides (CPRPs), which are located between the signal peptide and the actual hormone. The CPRPs have not yet been studied intensively. The CHH/MIH/GIH family is one of the most extensively studied neuropeptide families in crustaceans. Several review papers have been published discussing their structures, localizations and physiological functions [258-264]. Here the authors will update some more recent findings. 2.6.9.1. CHH in the Crustacean The CHH/MIH/GIH family is large and growing, having gained several new members in the last few years, due mostly to the expansion of genetic information available. These peptides are the largest known crustacean peptide hormones with molecular weights (MWs) between 8000 to 9000 Da, compared to 500-2000 Da for other neuropeptides. This larger size is a main reason that conventional characterization of CHH peptides relies on Edman degradation and/or cDNA cloning as opposed to MS-based techniques. Smaller crustacean NPs have MWs in the region for which mass spectrometers have had excellent performance over the last several decades, whereas only recently have high-resolution MS instruments been developed with the capability to adequately characterize compounds with MWs in the mass range of the CHHfamily peptides. Use of these high-resolution mass spectrometers in conjunction with a suite of tools typically used to study proteins has recently been reported by Ma et al. (2009). These researchers developed a MS technique combing both top-down (intact molecule fragmentation MS/MS) and bottom-up (enzymatic digestion of the peptide prior to MS analyses of each resulting fragment via MS/MS) strategies to characterize a novel CHH isoform from the crab C. borealis. No previous work had identified a CHH sequence solely with MS-based methods [56]. CHH-type I peptides have been more extensively studied since their first isolation and 67 identification from the XO-SG complex in 1979 [265]. This isoform was later found in other tissues by antibody-based techniques. For example, Keller et al. employed RIA to detect CHH at low concentrations in the pericardial organs of C. maenas [266]. Other immunochemical localization studies have identified CHH immunoreactivity in the second roots of the thoracic ganglia of H. americanus [267], the retina of P. clarkii [268], and the gut and abdominal peripheral neurons of C. maenas [269, 270]. The CHH family is one of the best-studied NP families in crustaceans, although MS-based study of it has been limited until recently. Its localization throughout several organs of the crustacean nervous system has been described using immunochemical techniques. 2.6.9.2. CHH Function in Crustaceans CHH is an important hormone that possesses multiple functions in crustaceans throughout all the life stages of the animal. As its name implies, CHH type I peptides cause hyperglycemia and hyperlipidemia in the hemolymph as part of the crustacean’s regulation of energy metabolism and storage [260]. Through binding with hemocyanin, CHH travels from neuroendocrine organs to the target tissues, such as midgut and musculature, through the hemolymph. It is thought that hemocyanin protects it from degradation by enzymes in the hemolymph, much as albumin does for peptide hormones in mammals. At the target tissues, CHH causes the release of free glucose from glycogen stores in a manner reminiscent of glucagon. CHH can also induce breakdown of lipid stores in the midgut, thus increasing hemolymph fatty acid levels [271, 272]. In addition to the hyperglycemic role of CHH, it is involved in other processes, such as osmoregulation [273], control of ecdysis [269, 274], inhibition of ovarian protein synthesis [275, 276], and inhibition of androgenic gland function [277]. It appears that the role of type I peptides is primarily to act on distant target tissue, 68 including endocrine glands, with a variety of end functions. Some of these functions overlap with those of the type II (GIH, MOIH, MIH) subfamily, such as action on ecdysis and reproductive organs. The adaptive character of CHHs has also been extensively studied. The level of hemolymph CHH increases in response to changes in environmental conditions such as oxygen, temperature, salinity, parasitism, heavy metals and exposure to lipopolysaccharide (causing immune system challenge) [278-283]. It should be noted that CHHs from the pericardial organ have different primary structures and possibly different functions compared to CHHs released from the SG. This is illustrated in research conducted by Chung and Zmora [278]. These authors looked into the expression and release of SG-CHH and PO-CHH in C. sapidus in response to stress induced by hypoxia, emersion and temperature change, concurrent with monitoring hemolymph glucose and lactate levels. Hemolymph glucose and lactate levels increased dramatically under all of these stresses. Glucose levels also significantly increased when SGCHH was injected into crabs, but PO-CHH did not have the same effect. In addition, hypoxia caused significant changes in the hemolymph concentrations of SG-CHH but not PO-CHH [278]. This suggests that SG-CHH carries out the more traditionally understood role for CHH, that of energy regulation, particularly in response to environmental stress. PO-CHH does not appear to be involved in these processes, and thus has a currently unknown function. Other functional studies of CHH in the crustacean are similar to this one in that they rely upon immunochemical methods for detection. They typically implicate CHH type I peptides in the role for which they were named, energy metabolism, in response to a variety of factors. 69 2.6.9.3. MIH in the Crustacean MIH is a member of the CHH/MIH/GIH type II subfamily, and it too has been found in many decapod species. In a recent review, MIH peptides were discussed in detail [262]. At the time of that review, 26 MIH or MIH-like sequences were known, coming from varied decapod species. The receptor for MIH has been identified, although it is not fully characterized. This receptoris localized primarily in the Y-organ. From in vitro studies, it has been determined that MIH delays molting by suppressing ecdysteroid synthesis by the Y-organs. A number of other features of MIH peptides are also discussed in detail in the review [262]. In summary, MIH peptides are type II peptides of the CHH family that are widely distributed among decapods. They inhibit molting, as their name implies, by acting directly on the Y-organ’s synthesis of ecdysteroids through a receptor about which little is known. 2.6.9.4. GIH in the Crustacean Compared with CHH and MIH, only a small number of gonad-inhibiting hormones (GIHs) have been characterized to date. The first peptide with in vivo GIH activity was isolated from the American lobster Homarus americanus [284]. The mRNA encoding GIH-like peptide has also been found in the Norway lobster Nephrops norvegicus [285] and the shrimp Penaeus monodon [286]. Like other CHH family peptides, they are present in the SG-XO complex. The GIH transcript has also been identified from other tissues, such as the brain and thoracic and abdominal nerve cords in the shrimp [286]. As its name may imply, GIH is a vital peptide hormone in crustacean reproduction. Its inhibition of gonad maturation is achieved by modulating the synthesis of vitellogenin (Vg), which is a precursor for yolk proteins [287]. This is illustrated by a study in which knockdown of Pem-GIH induced by double-stranded RNA increased the transcript level of Vg. Therefore, GIH inhibits Vg at the gene expression level 70 [286]. Although GIH is not as well studied as CHH type I peptides and MIH, its function is understood to be inhibition of gonad maturation and formation of yolk proteins. 2.6.9.5. MOIH in the Crusteacean Another member of the type II CHH subfamily is mandibular organ-inhibiting hormone (MOIH). Mandibular organs (MOs) have a homologous role to the corpora allata in insects. This organ is essential in reproduction and development. The corpora allata is regulated by allatostatins in insects, but these peptides share no sequence similarity to MOIH. Discovery and functional studies of MOIH have been carried out in several crustacean species. In the crab Cancer pagurus, two MOIH peptides were identified, and in an ex vivo assay of isolated MOs, both inhibited synthesis of the terpenoid methyl farnesoate (MF) [288]. A study in the crab Libinia emarginata identified a peptide with a CHH-like sequence. Investigation of its function showed that this peptide inhibits MF synthesisand has hyperglycemic activity, thus giving this peptide a putative assignment as a CHH-family type II peptide similar to MOIH [289]. The inhibition of MF synthesis by MOIH has also been investigated in vivo, although the results do not conclusively verify this function for MOIH. The effects of injecting eyestalk-ablated male Cancer pagurus with synthetic MOIH-1 or MOIH-2 were compared with injection of sinus gland extracts. SG extracts reduced hemolymph levels of MF by 60-80%; however, injection of MOIH-1 or -2 did not dramatically affect MF levels. Treating the sinus gland extracts with protease abolished their activity, which indicates that an additional peptide or protein in the extract—other than MOIH—may be responsible for the in vivo activity of the sinus gland extract [290]. It is also possible that both peptides must be present for activity. Although ex vivo studies suggest that MOIH inhibits MF synthesis by the MO, in vivo studies provide a more complex 71 picture of MF synthesis regulation. Thus, regulation of the mandibular organ may require coordination of multiple hormones produced by the SG. 2.6.10. Pigment-Dispersing Hormone (PDH) Pigment-dispersing hormones (PDH) were discovered along with pigment concentrating hormones (RPCH) in early neuroendocrine studies of the crustacean eyestalk, and these two families are sometimes grouped into a single superfamily [4]. Although the peptides have similar canonical functions—acting on the distribution of pigment granules in specialized colorcontaining cells, they share little sequence similarity and act on different subtypes of cells. For this reason, these two families will be treated separately in this review. Initially called distal retinal pigment hormone (DRPH), PDH was discovered after investigations into the molecular entities that caused migration of pigments in the retinae of crustaceans, as reviewed by a number of authors [4, 291-294]. The first PDH family neuropeptide was sequenced in 1976 as NSGMINSILGIPRVMTEA-amide from the shrimp Pandalus borealis [295]. A different PDH isoform was later found in the crabs Uca pugilator [296] and Cancer magister [297], with the sequence NSELINSILGPRVMTEA-amide. These two types of PDH, designated α and β respectively, were later found in numerous species, with additional isoforms containing only a few amino acid substitutions also identified (reviewed in [298] and [299]). Several postulated PDH sequences, including both α and β types,determined from genomic analysis exist [294], but they are not listed here. As indicated, many more isoforms of β-PDH than α-PDH have been identified, and β-PDH has been discovered in more species. Some species also contain multiple isoforms of β-PDH. It was once postulated that only prawns of the Pandalus genus expressed α-PDH, but it has also been described in Macrobrachium rosenbergii from cDNA clones [300]. Although there are two different types 72 and many isoforms of each type, the overall sequence similarity is high and all members are octadecapeptides. Thus the PDH family is considered a highly conserved family among crustaceans. Differences between the α and β types lie mostly in positions three and 4, which contain large, charged residues at neutral pH (E, L) in β-PDH but small, neutral residues (S, G) in α-PDH. The full significance of these differences is not known. When a species has multiple isoforms of one of the PDH types, they typically vary in only a few amino acids, and these changes typically do not drastically alter the overall properties of the molecule. The presence of multiple isoforms in a single species may indicate intricate regulation of physiological processes, or simply redundancy. 2.6.10.1. Distribution of PDH in Crustacean Tissues PDH localization has recently been described in detail in a review [294]. It is found mostly in the cerebral ganglion (contains eyestalks and brain) and ventral ganglia of crustaceans. The eyestalks are considered to be the lateral part of the protocerebrum, although they may be separated from the rest of the brain by long fiber tracts. These tissues demonstrate a great deal of PDH immunoreactivity in several species, with PDH-reactive cell bodies in the more medial parts whose fibers terminate in the sinus gland—located more laterally, and in the medial protocerebrum and other more medial ganglia. In the medial protocerebrum, immunoreactivity is observed in cell bodies in an area of neuropil with an identified role in photoreception. in the medial protocerebrum, Most of the immunoreactivity for PDH in the brain is in neuropil with cell bodies originating in the eyestalk, but there are a few paired cell bodies that contain PDH that send projections to the circumesophageal ganglion (COG). Differential distribution of isoforms of β-PDH has also been observed, with the general finding that Cancer productus βPDH II is the only isoform found in the SG, and thus it is likely a neurohormone while isoform I 73 is more likely to be a local neurotransmitter [294]. Overall, the distribution of PDH isoforms in the cerebral ganglion (including eyestalk) suggests both local activity, with projections typically originating from cells in the eyestalk, and a neurohormonal role, as many fibers terminate in the secretory structure SG. Also described in the review is localization of PDH in other structures. Outside of the cerebral ganglion, PDH-immunoreactive fibers are found in the ventral nerve cord, COG, and stomatogastric ganglion (STG). They are typically found in neuropil and as fiber terminals, although some species have somata in the COGs that stain for PDH [294]. This localization pattern is more suggestive of a neurotransmitter role for PDH, although its presence in structures far from the eyestalk may indicate that it is mediating circadian effects on ganglia not directly involved in adaptation to light. Overall, the distribution of PDH in crustacean ganglia suggests that it is not simply involved in adaptation of the retinal pigments to light changes. Its presence in a neurosecretory organ and projections and cell bodies that are far from the eyestalk point to additional roles, possibly in using light signals as a trigger for other physiological changes. 2.6.10.2. Function of PDH in Crustaceans The primary function of PDH is translocation of pigment granules in chromatophore cells by moving them toward the distal parts of these stellate cells. This has been reviewed by many authors [4, 294]. Certainly, the effect of PDH on isolated chromatophores is well established: application of it or crude extracts containing it cause pigment cells to express color, as the pigment granules are dispersed centripetally [4]. This color change is of importance for day/night adaptation and camouflage. One genus in which circadian color change has been well-studied is the fiddler crab Uca, which undergoes a darkening of pigment during the day and a lightening during the night, with the darker pigment in the daytime acting as protection from the light of the 74 sun. This pattern of pigment migration based on time occurs even in the absence of light, and two other crab species have been shown to have a similarly persistent time-related color change [294]. Migration of pigments in retinal pigment cells is also a main function of PDH, as it allows adaptation of the eye to low or high ambient light conditions. It also affects the electrical activity of photoreceptor cells, mediating changes in sensitivity that are important for vision at different times of the day [4, 299]. PDH has been shown to cause pigment dispersion in chromatophores and retinal pigment cells. In general, it helps the crustacean adapt to high ambient light conditions by this mechanism. The possibility of PDH as a circadian hormone in crustaceans has been studied in detail due to a number of factors. First, the persistence of circadian color change in the absence of an external stimuluspoints to an internal clock, and the ability of PDH to cause color change suggests it may be part of that system [4]. Secondly, the related pigment dispersing factors (PDFs) in insects have well-established roles in circadian rhythms, mostly as output messengers that relay time-related signals from the central nervous system’s clock circuits to target sites [4, 294]. Few studies have directly shown that the concentration of PDH varies in a circadian manner [301]. However, one recent study found increases in the levels of PDH immunoreactivity in the brain photoreceptor neuropils (BPNs) of Procambarus clarkii and Cherax destructor that could be correlated with the animals’ distinct circadian patterns of locomotor activity [302]. In general, they saw increased PDH immunoreactivity during and immediately preceding periods of increased locomotor activity, although the two species differ in their exact patterns of circadian activity. Significantly, PDH is expressed at the beginning of the dark photoperiod, in contradiction to its role in pigment dispersion in chromatophores. Pigment concentration, not dispersal, should occur during the dark period. This also correlates with a period of increased 75 activity in many crustaceans, including both crayfish species in this study. However, P. clarkii also has a period of increased activity near the end of the dark photoperiod, and in this species, PDH immunoreactivity was also increased during this time. For C. destructor, which does not exhibit activity around the time of the dark-light transition, PDH immunoreactivity also did not increase at this time [302]. These findings, in particular the increase in PDH at the beginning of the dark period, suggest a circadian role for PDH in addition to its chromatophore dispersal role. Unchanged levels of PDH at the end of the dark period in C. destructor are surprising and provide further evidence for this. This study suggests that PDH is mediating time-related signals about the transition from one light phase to another that may signal for increased locomotion. Recently, Solís-Chagoyán et al. (2012) demonstrated that daily injections of P. clarkii β-PDH were sufficient to alter the period and phase of a pattern of electrical activity in the retina that shows a circadian pattern of that species. It also altered spontaneous activity in an area of the brain that is suggested to form part of the circadian system due to connectivity (the cerebroid ganglion). Thus, the authors postulated that PDH acts upstream of the retina, on the cerebroid ganglion, to alter its responsiveness in a circadian manner [303]. This effect, in addition to its downstream effects on the pigment and photoreceptor cells, is likely a hormonal component of the circadian rhythm. A great deal of evidence exists to suggest that PDH is involved in the internal clock of crustaceans. It has been demonstrated to act as an effector of the circadian clock, as it appears to act on chromatophores and neurons directly, similar to the role of PDFs in insects. 2.6.10.3. Molecular Action of PDH and its Receptor The PDH receptor has been identified as a G-protein coupled receptor in Daphnia pulex via analysis of its homology to C. elegans and D. melanogaster pigment-dispersing factor 76 receptor genes [299]. Although the sequences of PDH and PDF receptor genes are known in several invertebrate species, little is known about the intracellular signaling components of the PDF/PDH response. Pharmacological knockout experiments have made some progress in identifying parts of the signal cascade. As reviewed by Nery and Astrucci in 2002, signaling through Gs to adenyl cyclase produces an increase in cAMP, which causes activation of cAMPdependent protein kinase (PKA). The authors also point to phosphorylation of an unknown protein with molecular weight around 58kDa as a possible downstream component of the signaling pathway [304]. Few studies have addressed the mechanisms by which PDH causes pigment dispersion in crustaceans, although Gs, adenyl cyclase, and cAMP have been implicated. Some work has recently been done in Drosophila to dissect the steps of the signaling pathway through genetic techniques. The PDF receptor in the fly is a Class II GPCR similar to calcitonin-CGRP receptors. It causes increases in cAMP, likely through Gs. It was also found to increase intracellular Ca2+ as a second messenger [305]. The cAMP increase has later been determined to be mediated by adenylate cyclase AC3 in vivo in a subset of Drosophila pacemaker cells in which the PDF receptor is active. Other GPCRs in the same cells do not signal through AC3, and it is postulated that a physical association between the PDF receptor and AC3 is responsible for this specificity of second messenger activity. The A-kinase anchoring protein nervy is a component of cellular scaffolding that may be at least partially responsible for this [306]. This has interesting implications in how molecules that can be activated by multiple upstream signaling proteins in theory are specific to only a few signaling pathways in vivo. Although it has been studied in greater detail, it is still unclear how PDF causes pigment dispersion in insect chromatophores. Several of the same intracellular messengers have been identified as in the crustacean studies. 77 2.6.11. Red Pigment Concentrating Hormone (RPCH)/Adipokinetic Hormone (AKH) Red pigment concentrating hormone (RPCH) was first isolated from the shrimp Pandalus borealis in 1967 [307]. The full sequence was then determined in 1972 to be pQLNFSPGWamide [308]. Similar hormones were found in insects, with roles in energy mobilization, in the adipokinetic hormone (AKH) family. These peptides typically have a sequence 8-10 amino acids long with several conserved features: pGlu at the N-terminus, amidation at the C-terminus, Phe or Tyr at position 4, Trp at position 8, Gly at position 9 (if present), and no net charge at physiological pH [309]. Several reviews have discussed this family in insects in great detail [202, 291, 292, 309, 310 , 311-313]. In insects, a large amount of variety is found in the sequences of peptides in the AKH family, but in decapod crustaceans, only one sequence has been identified—pQLNFSPGWamide—although it has been seen in at least 10 species [309]. A variant form, pQVNFSTSWamide, has been predicted from the ESTs of the crustaceans Daphnia magna [84] and Daphnia carinata [314], and the genome of Daphnia pulex [85]. The low sequence variation of this peptide across species and absence of multiple isoforms is uncommon for a neuropeptide in Crustacea, especially as the insect species display a large and varied set of AKH peptides. 2.6.11.1 RPCH Distribution in Crustacean Tissues Red pigment concentrating hormone was first isolated from eyestalks, and its existence in this tissue has been extensively documented. Only a few of the many studies that have demonstrated RPCH in the eyestalk are listed here: [307, 311, 315-317]. Localization in the eyestalk is consistent with the canonical function of RPCH in adaptation of skin coloration to different environments. Early studies found RPCH in the ventral nerve cord (VNC) and substances released from the VNC after electrical stimulation [318]. RPCH has also been 78 identified in releasate from the stomatogastric ganglion upon stimulation of several nerves that have inputs to this ganglion [172], and by immunochemistry in the STG and PO in addition to the eyestalk sinus gland (SG) [319]. The existence of RPCH outside of the eyestalk and its release upon nerve stimulation are indicators that it may be released upon multiple stimuli in addition to alterations in light or color that are perceived in the eyestalk. It may thus represent a more global signal in the animal, altering multiple pathways in response to light and/or color in the environment. Although NPs released by the SG can enter the general circulation of the crustacean, release of RPCH by the PO, VNC, and directly at the STG will have a more direct and concentrated effect. This may also suggest a role RPCH in crustaceans as whole-body modulators of energy mobilization, similar to the identified roles of the related adipokinetic hormones in insects. 2.6.11.2. Function of RPCH in Crustaceans The function for RPCH determined in early studies of the crustacean was alteration of the localization of pigment within chromatophore cells, as reviewed by Lars Josefsson, a researcher largely responsible for the identification and initial sequencing of RPCH from prawns, in 1983 [291]. The author’s interest in RPCH was stimulated by its ability to change the distribution of pigment in the cells of the integument, which permits some species to change color. This article illustrates different stages of pigment distribution in a red pigment-containing cell of the hypodermis (erythropore). In the absence of RPCH, pigment is distributed widely throughout the star-shaped cell. The effect of RPCH is to move pigment particles within the erythropore toward the center of the cell, which results in a loss of overall color in the animal. This allows animals to adapt their coloration to the conditions to which they are exposed. Josefsson also indicates that crustaceans without eyestalks have dispersed pigment in their erythropores, and injection into a 79 de-stalked shrimp became a useful bioassay for the presence of RPCH in the injected substance, as its injection into these animals led to loss of color (blanching). Additional studies reviewed in the article demonstrated that RPCH causes concentration of pigment in all types ofthe hypodermal chromatophore cells found in several other crustacean species—erythrophores (red), leucophores (white), xanthophores (yellow), and melanophores (brown/black)—although not all types in all species were responsive [291]. This article provides a comprehensive summary of the role of RPCH in altering the chromatophore cells in the hypodermis of crustaceans. The canonical function of RPCH described in this article is to concentrate pigment molecules at the centers of chromatophores, with effects seen in several crustacean species and many (but not all) types of chromatophores—concentrating not just red pigments, but white, yellow, and brown/black pigments as well. It is also clear from this article that the majority of RPCH that is responsible for these color changes comes from the eyestalk of the animal. Additional reviews have summarized the role of RPCH in alteration of pigment distribution in chromatophores [5, 292, 293, 320]. As color change has long been associated with ambient light conditions [321], it is possible that RPCH constitutes a molecular signal of light intensity, which may indicate a role in altering other processes that are different during day and night and throughout seasons. Its pigment concentrating effects have been observed in the distal retinal pigment cells of the crayfish Procambarus clarkii [322, 323]. RPCH not only changes the distribution of pigments in the retinal cells of P. clarkii, but it also increases the electroretinogram (ERG—a measure of the sensitivity of the retinal cells to light) in vivo in both light- and dark- adapted animals [323] and in isolated retinas (Orconectes limosus) [214]. This direct effect on the retinal cells’ responsiveness to light, independent of the location of the pigment, suggests that RPCH has an 80 effect on the activity of the photoreceptors themselves. The level of RPCH in the eyestalk of P. clarkii is also seasonally variable [324]. Thus, circadian and seasonal modulation of visual perception in crustaceans is another function of RPCH. Although the primary function of the related adipokinetic hormones in insects is energy regulation, and some studies have found that crustacean RPCHs can induce such changes when introduced to insects [88], RPCH has not been observed to have this role in crustacean studies. A compound with sequence identical to the Pandalus borealis RPCH was identified in the isopod Porcellio scaber, and found to induce hyperglycemia without affecting physical activity level when applied topically to these insects [325]. A potential role in energy mobilization is suggested by the activity of RPCH in the stomatogastric nervous system [172, 326-333], which is primarily responsible for movements of the crustacean stomach. In this system, a number of different neuromodulators including RPCH can induce an inward current in the lateral pyloric (LP) neuron with an amplitude that depends on the cell’s resting potential, resulting in increased excitability of this neuron and as a result, the rest of the STG circuit [331]. Each of these NPs leads to a different STG output and affects a different subset of cells; for RPCH the cells altered are the LP and anterior burst (AB) neurons [332]. Changes in this circuit are not only in activation of certain patterns, but also in modulation of these patterns. Movement of the stomach leads to digestion, which can increase hemolymph glucose levels. The ability of RPCH to cause this in a system that is also regulated by a large number of other neuromodulators suggests that it is part of a mechanism that integrates information from various modalities to produce different outputs. However, inducing hyperglycemia by increasing digestion is a vastly different mechanism than the energy mobilization from stores induced by AKH family hormones in insects. The link between RPCH and stored energy mobilization in crustaceans has not yet been 81 established; however, RPCH is one member of a set of neuromodulators that together provide information to the STG to elicit stomach contraction, which does have the overall effect of increasing available energy. RPCH has also been shown to increase activity in several other crustacean ganglia, and at the neuromuscular junction. The crayfish swimmerets are controlled by a set of neurons in the abdominal ganglia that form pattern-generating circuits. The patterns of output changedin response to RPCH, slowing the swimmeret movement pattern by increasing the length of motor neuron bursts and the period between them [334]. RPCH increased the amplitude of muscle contraction in a simple preparation, likely by increasing the release of the primary neurotransmitter by the motorneuron [335]. It also has been shown to activate and modulate the cardiac sac rhythm and cardiac ganglion (CG) in several crustacean species, leading in general to patterns that would increase heart rate and contraction strength [152, 328, 329]. It can also activate different rhythms depending on the location within a network where it is applied, can cause different networks to synchronize, and make two networks join together, as reviewed in 2001 [336]. In these studies, RPCH has been shown to have diverse modulatory actions. The amplitude of firing is increased, and the timing of bursts is altered differentially depending on the system. They are increased in the CG and neuromuscular junction, but decreased in the crayfish swimmeret system. Overall, it appears that RPCH increases the tendency of crustacean neurons in these systems to fire, which is consistent with observations in the STNS. 2.6.11.3. Molecular Action of RPCH and its Receptor Like most NPs, the receptor for RPCH is expected to be a G-protein coupled receptor (GPCR), although it has not yet been identified in a published work. A putative Daphnia pulex RPCH receptor is present in GenBank [337], with accession no. EU503126, submitted in 2008 82 by Hamers et al. [338]. This receptor was identified by homology to insect AKH receptors, many of which have been published by the same research group [230, 251, 339-341]. Further research and sequencing of additional crustacean genomes may lead to a more confident identification. The mechanism by which binding to this receptor produces pigment aggregation in chromatophores has been studied in detail, but is still not completely understood. A number of works were recently compiled into a model by Milograna et al. that explains the process of how RPCH binding to its receptor leads to translocation of pigment granules along the cytoskeleton [342]. Following binding to its receptor, the typical heterotrimeric G-protein signal transduction pathway is activated. This somehow leads to activation of ryanodine receptors on the intracellular smooth endoplasmic reticulum, which causes release of Ca2+ through channels in an unknown mechanism. This increased intracellular Ca2+ leads to closure of the K+ channels that normally leak K+ when the cell is at rest, which is then balanced by the Na+/K+ ATP-ase. The mechanism by which the increased intracellular Ca2+ leads to closure of these channels is also unknown, although the authors postulate it could be due to the change in Ca2+ concentration itself, a secondary action of the G-protein subunits activated by the RPCH receptor, or phosphorylation by protein kinase C/Ca2+-calmodulin. Closing this channel causes the cell to depolarize greatly, which leads to opening of several types of Ca2+ channels (voltage-gated and others) on the cell surface. This causes extracellular Ca2+ to enter the cell, and once it reaches a threshold concentration, it activates Rho-protein kinase directly and/or through production of cGMP. This protein kinase phosphorylates a specific myosin II molecular motor, which is attached to a pigment granule. The myosin then moves the pigment granule toward the center of the cell [342]. This mechanism may also be responsible for the other functions of RPCH in nonchromatophore cells. The presence or absence of RPCH, leading to efflux of intracellular 83 calcium, could lead to circadian or seasonal alterations in other roles beyond color change. This mechanism would also lead to increased excitability of neurons with RPCH receptors by depolarizing their resting potential; this is consistent with observed evidence that RPCH increases activity in the crustacean in the retina, in several pattern-generating circuits, and at the neuromuscular junction. The mechanisms by which the various insect AKH family hormones exert their metabolic effects have also been studied in detail and reviewed [343]. As mentioned previously, the AHK receptor is a GPCR. It also causes release of Ca2+ from intracellular stores and influx of extracellular stores, although the mechanism is different and better understood. A variety of signaling pathways are employed in insect cells to produce mobilization of a several different stored energy compounds, actuated largely by increases in intracellular Ca2+ and activation of phosphorylases that break down the molecules used for energy storage. These signaling pathways include adenyl cyclase activation of phosphorylase b kinase, phospholipase C (PLC)induced release of Ca2+ from the endoplasmic reticulum via inositol triphosphase, and PLC activation of protein kinase C by diacyl glycerol leading to phosphorylase activation [343]. Although the intricate mechanisms have been simplified here, the overall effect of AKH binding to its GPCR in insect cells is to metabolize stored energy molecules, with cAMP, PLC, and Ca 2+ as mediators. Such a large variety of intracellular signaling pathways for AKH family peptides in insects permits a wide variety of specific signals to be transmitted, as AKH is known to initiate metabolism of three different types of stored energy molecules. Most of the pathways discovered in insects are upstream of the known pathways for RPCH signaling in crustaceans, and it is possible that further study into the “missing links” in the RPCH signaling chain may identify the 84 same messengers that are employed by AKH, in particular those that lead to release of Ca 2+from intracellular stores. 2.6.12. Pryokinins Peptides of the pyrokinin family were first discovered in insects, where their regulation of reproduction is well-characterized in several species. For information about these peptides in insects, recent review articles are suggested [344, 345]. For several aspects of pyrokinin peptides, information only exists in the context of insect systems; they will not be discussed here (pyrokinin receptor, reproductive function, molecular action, interaction with other factors). Insect pyrokinins are defined by having a common C-terminal motif of FXPRLamide, with X as T, S, G, or V [344]. In crustaceans, the common FXPRLamide motif is also observed, although additional amino acids can occur in the X position. The N-terminus typically adds an additional 2 to 5 AAs. For Penaeus vannamei, the first crustacean in which a pyrokinin was identified, S and N occupy this position in the two identified pyrokinins [346]. Homarus americanus has S in the X position in its one identified pyrokinin [57]. In Litopenaeus vannamei, L and N are observed in addition to S [347]. The amino acids A and S occupy this position in the pyrokinins of Carcinus maenas [102] and Cancer borealis [59]. Finally, in Callinectes sapidus, the observed pyrokinin has an S in this position [348]. In the single crustacean with a sequenced genome, Daphnia pulex, pyrokinins were not identified, likely due to the short sequence tag that could be used for interrogation of genetic sequences [85-87]. In Litopenaeus vannamei, a pyrokinin could also not be identified from the known genomic information that was available for that species [347]. It is possible that transcriptomics strategies are not successful in finding sequences with homology tags of this size. Sequences of pyrokinins are characterized by the FXPRLamide motif in insects and crustaceans, although instead of having T, S, G, or V 85 occupying the X position, N, S, A, and L are observed in crustaceans. It is clear that flexibility of the sequence at this position is a common theme among invertebrates, and although the exact AAs observed in crustaceans can differ from those in insects (with the exception of S), many have similar properties on the side chains. G and A are typically classified together as non-polar, small AAs. V and L are considered to be medium-sized, hydrophobic AAs. N and T, however, do not have many similar characteristics. The ability to have different pyrokinins with similar but distinct receptor properties due to slight alterations in sequence is an advantage possible in both insect and crustacean pyrokinins. The localization of pyrokinins in invertebrates suggests multiple modes of action, both directly on identified neural circuits and hormonally through circulating fluid. In insects, pyrokinins are found in the brain-subesophageal ganglion complex, corpora cardiaca, ventral nerve cord (VNC), and abdominal and thoracic ganglia [345]. In crustaceans, they have been identified in cell bodies in the commisural ganglia (CoGs), and neuropil of the STG and CoGs of C. borealis [59, 65]. In this species and C. sapidus, it was found in the pericardial organs (POs) as well [59, 65, 348]. Pyrokinin-family peptides were found in the brain and VNC of H. americanus [57] and L. vannamei [347], and the brain of C. maenas [102]. It was also isolated from an extract of the entire central nervous system of P vannamei [346]. This wide pattern of distribution does not suggest a single role for pyrokinin peptides. The presence within the STNS circuits suggests a role in stomach contraction. PO localization suggests a hormonal role. Presence in the central nervous system indicates a more complex function. No single role is suggested by the patterns of localization of pyrokinin peptides in crustaceans, and thus local neurotransmitter and hormonal roles are both likely. 86 The primary roles of pyrokinin peptides in insects are regulation of reproduction and gut motility, although additional diverse functions have recently been suggested, including regulation of diapause and coloration [344]. Far less is known about pyrokinin function in crustaceans. Only a few studies have investigated its action in the well-characterized circuits of the crustacean STNS, using Cancer borealis. It was determined to activate the gastric mill rhythm, acting on all neurons involved in this rhythm, with all 4 pyrokinin peptides having essentially the same effect [65]. It was later shown that output of the gastric mill rhythm following pyrokinin application was nearly identical to that produced by activation of a single identified STG neuron, MCN1. This is achieved in an interesting manner, by changing the relationships of neurons within the circuit to one another. The same neurons are employed by the MCN1-stimulated gastric mill rhythm as by the pryokinin-stimulated gastric mill rhythm, but the way in which their firing affects each other is altered [349]. The significance of this finding is as yet unknown in the overall physiology of the crab; however it has importance in understanding neuronal circuit organization and modification by neuropeptides. Pyrokinin clearly has an important role in the crustacean STNS, eliciting a vital rhythm for digestion. Another study found that it was decreased in the brain after feeding in C. borealis via MS-based stable isotopelabeling quantitative methods [350]. This is indicative of a release of stored pyrokinin, degradation of extracellular pyrokinin present in the unfed state, reduced synthesis of pyrokinin, or a combination of these factors. It is possible that neurons from the CoG, where cell bodies are positive for pyrokinin, are releasing that substance into the STG or other areas to stimulate the gastric rhythm, instead of into the brain. Other roles are also likely, based on localization of the peptide and functions of pyrokinins in insects. 87 2.6.13. Proctolin 2.6.13.1. Proctolin Discovery and Sequence Identification in Arthropods In 1967, a substance, first termed “gut-factor,” was isolated from the gut of the American cockroach Periplaneta americana [351]. This substance was found to stimulate visceral muscle contractions, and could not be identified as any of the known insect neurotransmitters at the time. Proctolin, as the compound was later known, then was isolated and sequenced (RYLPT) in 1975, followed by its synthesis and description of its function on the cockroach hindgut [352] in an assay that later became a standard preparation for assessing putative NP activity, the hindgut assay [353]. Since its discovery and identification, proctolin has been extensively studied in insects, focusing on its distribution among different species, structure-function relationships, and role in feeding behavior (more recent reviews: [108, 344, 354, 355]). In insects, it acts as a cotransmitter to modulate the postsynaptic response to another NT. This action occurs at the neuromuscular junction, synapses in the central nervous system, and distant receptors in an endocrine manner (reviewed in [354, 356]). After its discovery in P. americana, proctolin was subsequently applied to several different preparations of lobster ganglia and motor neuron synapses, and changes in activity were observed [357-359]. Endogenous proctolin in the crustacean was later demonstrated by a study in the crayfish Procambarus clarkii by a number of methods, including that a component of a muscle extract was chromatographically indistinguishable from [3H]-proctolin [360]. Schwartz and co-workers later verified its presence in an extract from Homarus americanus by fast-atom bombardment MS with a mass measurement error of 2.8 ppm [361]. The authentic sequence of proctolin as RYLPT from crustacean tissue was first obtained by Stangier and co-workers in 1986 using Edman degradation [362]. Subsequent studies have demonstrated that this sequence 88 is identical in many insect and crustacean species [363]. A comprehensive review of proctolin’s identification, distribution, function, and related substances in crustaceans does not exist; and a comprehensive review is beyond the scope of this article. The following section will focus on identification and distribution of proctolin in crustacean species. Its function on neural circuits and muscles is complex, and will not be covered in detail. Other aspects of its biological functions will be described, along with some information on its molecular action. 2.6.13.2. Identification of Proctolin in Crustacean Species Proctolin’s sequence has been positively identified as RYLPT in several crustacean species, using Edman degradation, tandem MS, and/or transcriptomics. Following Edman sequencing of authentic proctolin as RYLPT in Carcinus maenas [362], a number of studies confidently verified the existence of proctolin with an identical sequence in other crustacean species. Proctolin’s presence was confirmed by mass spectrometry in Cancer borealis first by Li et al. in 2003 [55], with further identification in this species in a later work [59]. Other studies went on to identify the peptide RYLPT in Homarus americanus [197], Cancer productus [101], Carcinus maenas [102], and Callinectes sapidus [364] by MS/MS. Tandem MS has been a powerful tool for identification of proctolin across many crustacean species. One study identified both proctolin and [Met-4]-proctolin via MS/MS and genome mining in Daphnia pulex.The MS/MS spectrum for RYLMT was of lower quality, and it has not been seen in insects [86]. The lower quality MS/MS spectrum may indicate a lower abundance of this alternative form. A variety of other genomics studies have met with variable success in identifying proctolin in crustaceans. Searching of D. pulex ESTs in 2009 was unable to identify the sequence of proctolin, but this is likely due to the poor performance of the software for identifying small peptides from genomic information as well as the limited amount of genomic information 89 available at the time, as the authors observed proctolin-like immunoreactivity [87]. Another study also failed to find RYPLT in EST databases for a number of crustacean species in 2008 [84]. Later studies of D. pulex after the genome became available in 2010 (at wfleabase.org), identified the sequences RYLPT and SSWGVDARYLPT, along with the proctolin precursorrelated peptide SDPLSPIGPPRGEDP [85]. EST mining was sufficient to identify proctolin in Litopenaeus vannamei, although the same publication failed to find it via MS [347]. The variable success of genome and EST mining for proctolin demonstrates the importance of a comprehensive database and difficulty when using tools designed to identify large sequences for finding NP sequences, which are typically much shorter. In fact, identification of the proctolin genes was greatly aided by the discovery of the preproproctolin gene in Drosophila melanogaster [365], which provided a larger sequence for searching in some of the studies noted here. Proctolin is found in many crustacean species with the same sequence, RYLPT, and one variant has been observed and verified by both genomics and MS/MS. This high degree of sequence conservation across Crustacea, and indeed Arthropoda, is unusual for a NP and may indicate that this NP is highly important and may exist in more species than previously postulated. The identification of a variant by genome mining and its verification by MS/MS in the one decapod crustacean with a sequenced genome hints, however, that previous studies identifying proctolin as RYLPT may be incomplete. Mining the genome permitted identification of the variant without relying on MS/MS, which has a bias for identification of the highest abundance compounds unless targeted methods are used. Further genomic analysis may provide sequences of putative variant forms, which could be verified by targeted tandem MS. 90 2.6.13.3. Distribution of Protcolin In crustaceans, proctolin is widely distributed across several ganglia. Early studies in H. americanus identified proctolin-like immunoreactivity in nearly every portion of the nervous system [361]. The distribution of proctolin in the nervous systems of several decapod crustacean species has been described in detail. Although there is some inter-species variation in proctolin distribution [209, 366-369], the overall patterns are similar. In general, studies have found the most intense protctolin immunoreactivity in neurosecretory cells, mainly in synaptic varicosities. The neurosecretory organs PO and SG have high proctolin concentrations, primarily in axonal areas [209, 319, 361, 362, 366, 368-372]. The proctolin-containing cell bodies that project to the POs are found in the esophageal ganglia (OG) and brain, and other proctolin-positive cell bodies in the OG project to the commissural ganglia (CoG) and stomatogastric ganglia (STG) [209, 366-368, 371, 373-377]. Protcolin neurons are present in the thoracic and abdominal ganglia, from where they project to other ganglia and/or have a neurosecretory role in function of the hindgut or postural stances [360, 368, 371, 378]. Several motorneurons with proctolin content have been identified, and its role in these neurons appears to be similar to that observed at the insect motor synapse—as a cotransmitter that modulates the response to other transmitters [368, 371, 373]. The STG contains proctolin primarily in neuropil [369, 379]. Finally, proctolin is found in somatosensory/mechanosensory cells of the thoracic and abdominal ganglia [368, 371, 373]. The localization of proctolin within the crustacean nervous system can be generalized as follows: nearly every ganglion contains proctolin, and it is present mainly in cell bodies and axons. Some ganglia contain proctolin primarily in cell bodies (esophageal ganglia, thoracic/abdominal ganglia), and some have it mostly in axonal projections from other areas (pericardial organ, sinus gland, stomatogastric ganglion), but few contain both (brain, 91 commissural ganglia). Axonal projections appear to take the form of either synaptic varicosities (“beads-on-a-string”) or discrete synaptic bouton-like structures. This pattern of localization suggests that proctolin has two main functions: neurohormonal via release into the hemolymph to affect distant targets, and neurotransmission via direct projection between ganglia. Most ganglia either send or receive proctolin signals, not both; thus each ganglion may be a site for integration of multiple peptide signals. Although it is nearly ubiquitous in the crustacean nervous system, the varied distribution of proctolin across different ganglia is indicative of its roles as neurohormone and neurotransmitter. 2.6.13.4. Proctolin Function on Neural Circuits By far, the most research into proctolin’s function has gone into analysis of its effect on central pattern generators (CPGs), neural circuits that fire in rhythmic patterns, typically with an output on muscle movement. Main CPGs in crustaceans include the stomatogastric nervous system, responsible for gut and stomach movements; the cardiac ganglion, responsible for heart movements; some motor neuron circuits involved in walking and posture; and neurons in control of gill ventilation. In general, proctolin has the effect of increasing the activity of a neural circuit [380]. This increased activity could occur at any number of levels in the circuit, includingsensory input neurons that initiate a rhythm or monitor its efficacy, interneurons that integrate signals from multiple other neurons, motor neurons that signal the output to muscles, and muscles themselves. Proctolin may act at multiple levels, and may affect different levels in different circuits. For a more detailed description of its action in several circuits and at the motor neuron synapse, a review article is suggested [380]. The sensitivity of some somatosensory neurons has been shown to be increased by proctolin, specifically in control of ventilation and walking legs. This was first demonstrated by 92 Pasztor and Bush in 1987 [381]. These same authors later found that proctolin was released after stimulation of stretch-sensitive neurons in the oval organ, which is important for regulation of gill ventilation [382]. Further experiments demonstrated that proctolin’s augmentation of the sensitivity of somatosensory neurons was opposite to the effect of the classical neurotransmitters serotonin and octopamine [383]. Thus, the authors postulated that local release of proctolin by the oval organ increases the sensitivity of that organ’s mechanosensory inputs, which in turn causes more proctolin release. This positive feedback loop can also be attenuated by monoamines, and the balance between proctolin and classical neurotransmitters permits regulation of the ventilation rate. A similar function was observed in the stretch receptors of the walking legs in crayfish: sensitization of the mechanosensitive neurons [384]. In contrast, proctolin was found to have no effect on the anterior gastric receptor (AGR), a mechanosensory neuron that receives inputs from stomach muscles, although the AGR was sensitive to a FMRFamide-like peptide. It should be noted that the AGR is located just outside of the STG [385]. Thus proctolin modulates neural circuit activity in some cases by increasing the sensitivity ofsomatosensory neurons. In central pattern generating circuits (CPGs), proctolin has the ability to initiate certain patterns and/or modulate them. It has been studied in the context of nearly all identified crustacean CPGs in several species, starting soon after its discovery. In the cardiac ganglion (CG), proctolin has been shown to initiate the cardiac sac rhythm and increase the frequency and duration of spike bursts, as well as the spike frequency and number in each burst [152, 328]. It also acts on nearly all cell types involved in the pyloric rhythm (one output component of the crustacean STNS), and is able to induce this rhythm and modulate several features of the pattern [332]. This is part of how proctolin and other NPs can produce similar currents in a specific cell 93 of the STNS, via activation of a number of different cells in the network, but lead to different overall circuit outputs [331]. Modulation of the pyloric rhythmis postulated to form the basis for changes in the gastric rhythm (the other output of the STNS) elicited by proctolin, as these two rhythms are distinct yet connected, and proctolin appears to have little effect on most of the cells that form this rhythm [386]. Proctolin has also been studied in the context of crustacean swimmeret [387-390] and leg [391] movements. It was found to typically cause excitation of swimmeret movements, acting alone as a neurotransmitter in the pattern-generating neurons of this system, although this effect occurs only in a subset of the motor neurons [387]. In leg movements, it had a different effect depending on the neurotransmitters with which it was applied [391]. When applied with dopamine or concurrent with activation of descending positive inputs, it led to a leg movement associated with a courtship display. Octopamine was able to prevent proctolin from inducing this behavior, unless dopamine was not also added [391]. In crustaceans, proctolin primarily increases the activity of cells within defined pattern-generating circuits. This is particularly strong in the cardiac ganglion, pyloric rhythm, and leg/swimmeret neurons. It many of these circuits, application of proctolin is sufficient for initiation of the circuit’s characteristic firing pattern, while in others, co-transmitters are necessary. Proctolin’s action in these circuits demonstrates how signals from diverse peptides can lead to similar but distinct outputs by acting on a subset of cells in a neural network or working differently depending on the other transmitters present. Its general tendency to lead to activation of firing in neurons of these circuits is consistent with the overall identification of proctolin as an activating peptide. Proctolin can also act directly on muscles to increase the output of circuits in crustaceans and insects (mentioned previously). Not surprisingly, its effect on the muscles of the hindgut has 94 been investigated in a number of crustacean studies, a few of which will be discussed here. Proctolin causes different types of contractions in muscles of the hindgut: in the long muscles that run parallel to the gut itself (longitudinal muscles), it increases the frequency and strength of contractions that occur spontaneously, and in the ring-shaped muscle fibers that surround the gut (circular muscles), it causes a slow, long-lasting contraction [392]. Similar studies on lobster arterial muscles demonstrated that proctolin induces contractions of these ring-shaped muscles, which leads to decreased arterial diameter and increased resistance to blood flow [393]. Some studies have found that proctolin’s ability to increase muscle function is due in part to increased neurotransmitter release in the presence of proctolin at the neuromuscular junction [335, 394], while others suggest that proctolin changes properties of the post-synaptic cell, permitting it to fire more strongly and with less stimulation [395]. Whether the activation is pre- or postsynaptic, proctolin acts directly on muscle, likely at the neuromuscular junction, to induce contractions and increase their strength. By increasing activity at all levels of a circuit—input, interneuron, and output—proctolin has an overall excitatory effect in the crustacean. 2.6.13.5. Molecular Action of Proctolin A GPCR in Daphnia pulex has been proposed as the receptor for proctolin by homology to insect proctolin receptors [2]. Much more is known about the effects of proctolin on targets further downstream in the signaling pathway. Several studies have indicated that calcium channels are integral to proctolin signaling [394, 396, 397]. Secondary messengers postulated to be responsible for this action on Ca2+ channels include cyclic AMP or GMP [395, 398, 399], protein kinase C (PKC) [399], protein kinase A (PKA) [395], and depolarization by K+ channels [395]. It is possible that different phosphonucleotides and different protein kinases act in different crustacean species. It has also been postulated, however, similar to the action of several 95 other NPs, that this GPCR leads to Ca2+ release from endoplasmic reticulum, and this, with or without additional secondary messengers, leads to changes in the electrical activity of the postsynaptic cell that predispose it to greater intensity and/or frequency of firing, and/or change the phosphorylation state of muscle proteins, leading directly to their activation [395, 399, 400]. This wide variety of possible sites where proctolin may influence firing and/or contraction permits it to have a range of functions, from slight alterations in circuit dynamics to dramatic changes in the animal. 2.7. Conclusions and Future Perspectives Invertebrate model animals have made significant contributions to the study of neuropeptides, both in methodology and in understanding how NPs regulate important processes. Mass spectrometry-based techniques are optimal for studying NPs, due to their high specificity when compared with other techniques. MS-based tools can generate peptide sequence information without tany a priori knowledge from a variety of neural tissue samples, ranging from intact tissues to microdialysate. Mass spectrometry imaging (MSI) is an emerging technique to map the distribution of NPs in a given tissue with greater specificity than antibodybased imaging techniques and the ability to characterize the distribution of thousands of analytes in a single experiment. Direct tissue imaging allows rapid identification of NPs from a tissue of interest, avoiding sample preparation steps that might lead to alterations in the chemical identity of NPs. Liquid samples can be subjected to a number of analytical separation techniques prior to MS analysis for improved confidence of identification. Quantitative methods are also rapidly expanding for MS-based platforms. Assays to determine the function of NPs can gain from the advances in MS techniques described above. One great advance is in the application of microdialysis sampling on alert, behaving animals for NP collection and correlation with 96 activity. MSI is also able to improve function determination by localizing NPs to neurons or subsections of ganglia with known physiological roles. In the age of readily available genome sequencing, additional tools to predict and identify NPs are available, and the sequencing of the first crustacean genome, from Daphnia pulex, has greatly increased these capabilities. Many NP families are known, and many of these have been characterized both chemically and functionally. Although recent advances in MS and genomic technology have been highly useful to the field of invertebrate (and vertebrate) neuropeptide discovery and functional identification, further technological advances are under development and the full suite of crustacean NPs has not been described fully. Advances in MS technology will greatly improve the capabilities of this technique to provide tissue localization, identity, and quantity information. In the area of MSI, methods for tissue coating with matrix are being improved to create smaller, more uniform crystals, to decrease analyte diffusion, provide better spatial resolution, increase sensitivity, and reduce “hot spots” of signal on the tissue. Improvements to laser technology will also improve the spatial resolution of this technique. For identification, faster scan rates, higher accuracy, and better resolution will improve the quality of identifications and allow less abundant compounds to be identified. Similarly, for quantification, increases in MS instrument speed, accuracy, and resolution along with the development of novel isotopic labeling techniques will enable more reproducible quantitation of lower concentration analytes. With increased coverage of crustacean genomes by next-generation sequencing, identification by database search software will likely improve in the future. Computer software and hardware advances have also increased the ability to run complex software, including de novo sequencing software, in more reasonable periods of time. Finally, many crustacean NPs currently have no known function, and a two-stage method 97 for function determination will be of use. First, NPs can be monitored using MS for comprehensive analysis of all compounds potentially involved in a particular function, especially with the aid of the minimally invasive sampling technique of microdialysis. This will narrow down the list of hundreds of NPs to a few “interesting” NPs in an unbiased and systematic manner. Then, those peptides that appear to be of import for that role can be investigated in detail, using targeted MS techniques or more traditional techniques, for a more confident determination of the role of the NPs in that function. All of these advances arepossible due to the incorporation of MS into the NP discovery and functional identification workflow. Further improvements to the analysis methods and increased study of NP functions will greatly increase our understanding of this important class of neurotransmitters, both in crustaceans and in other organisms, including mammals. 2.8. Respective Contributions Statement All authors researched, wrote, and edited the paper. 2.9. Works Cited 1. Li, L.; Sweedler, J. V. Peptides in the brain: mass spectrometry-based measurement approaches and challenges. Annu Rev Anal Chem (Palo Alto Calif). 2008, 1, 451-483. 2. Colbourne, J. K.; Pfrender, M. E.; Gilbert, D.; Thomas, W. K.; Tucker, A.; Oakley, T. H.; Tokishita, S.; Aerts, A.; Arnold, G. J.; Basu, M. K.; Bauer, D. J.; Cáceres, C. E.; Carmel, L.; Casola, C.; Choi, J.-H.; Detter, J. C.; Dong, Q.; Dusheyko, S.; Eads, B. D.; Fröhlich, T.; Geiler-Samerotte, K. A.; Gerlach, D.; Hatcher, P.; Jogdeo, S.; Krijgsveld, J.; Kriventseva, E. V.; Kültz, D.; Laforsch, C.; Lindquist, E.; Lopez, J.; Manak, J. R.; Muller, J.; Pangilinan, J.; Patwardhan, R. P.; Pitluck, S.; Pritham, E. J.; Rechtsteiner, A.; Rho, M.; Rogozin, I. B.; Sakarya, O.; Salamov, A.; Schaack, S.; Shapiro, H.; Shiga, Y.; Skalitzky, C.; Smith, Z.; Souvorov, A.; Sung, W.; Tang, Z.; Tsuchiya, D.; Tu, H.; Vos, H.; Wang, M.; Wolf, Y. I.; Yamagata, H.; Yamada, T.; Ye, Y.; Shaw, J. R.; Andrews, J.; Crease, T. J.; Tang, H.; Lucas, S. M.; Robertson, H. M.; Bork, P.; Koonin, E. V.; Zdobnov, E. M.; Grigoriev, I. V.; Lynch, M.; Boore, J. L. The Ecoresponsive Genome of Daphnia pulex. Science. 2011, 331, 555-561. 3. Marder, E.; Bucher, D. Understanding circuit dynamics using the stomatogastric nervous system of lobsters and crabs. Annu Rev Physiol. 2007, 69, 291-316. 4. Hopkins, P. M. The eyes have it: A brief history of crustacean neuroendocrinology. General and Comparative Endocrinology. 2012, 175, 357-366. 5. Christie, A. E.; Stemmler, E. A.; Dickinson, P. S. Crustacean neuropeptides. Cell. Mol. Life Sci. 2010, 67, 41354169. 6. Seeley, E. H.; Caprioli, R. M. 3D Imaging by Mass Spectrometry: A New Frontier. Analytical Chemistry. 2012, 84, 2105-2110. 7. Zhu, L.; Du, Q.; Blackler, A. R.; Tangrea, M. A.; Emmert-Buck, M. R. In Situ Proteomic Analysis of Histological Sections. Curr Proteomics. 2012, 9, 71-79. 98 8. Seeley, E. H.; Caprioli, R. M. MALDI imaging mass spectrometry of human tissue: method challenges and clinical perspectives. Trends in Biotechnology. 2011, 29, 136-143. 9. Greer, T.; Sturm, R.; Li, L. J. Mass spectrometry imaging for drugs and metabolites. Journal of Proteomics. 2011, 74, 2617-2631. 10. Dill, A. L.; Eberlin, L. S.; Ifa, D. R.; Cooks, R. G. Perspectives in imaging using mass spectrometry. Chem Commun. 2011, 47, 2741-2746. 11. Cazares, L. H.; Troyer, D. A.; Wang, B. H.; Drake, R. R.; Semmes, O. J. MALDI tissue imaging: from biomarker discovery to clinical applications. Anal. Bioanal. Chem. 2011, 401, 17-27. 12. Castellino, S.; Groseclose, M. R.; Wagner, D. MALDI imaging mass spectrometry: bridging biology and chemistry in drug development. Bioanalysis. 2011, 3, 2427-2441. 13. DeKeyser, S. S.; Kutz-Naber, K. K.; Schmidt, J. J.; Barrett-Wilt, G. A.; Li, L. Imaging mass spectrometry of neuropeptides in decapod crustacean neuronal tissues. J Proteome Res. 2007, 6, 1782-1791. 14. Baluya, D. L.; Garrett, T. J.; Yost, R. A. Automated MALDI Matrix Deposition Method with Inkjet Printing for Imaging Mass Spectrometry. Anal. Chem. 2007, 79, 6862-6867. 15. Chaurand, P.; Norris, J. L.; Cornett, D. S.; Mobley, J. A.; Caprioli, R. M. New Developments in Profiling and Imaging of Proteins from Tissue Sections by MALDI Mass Spectrometry. J. Proteome Res. 2006, 5, 28892900. 16. Rubakhin, S. S.; Greenough, W. T.; Sweedler, J. V. Spatial profiling with MALDI MS: distribution of neuropeptides within single neurons. Anal Chem. 2003, 75, 5374-5380. 17. Sugiura, Y.; Shimma, S.; Setou, M. Thin sectioning improves the peak intensity and signal-to-noise ratio in direct tissue mass spectrometry. J. Mass Spectrom. Soc. Jpn. 2006, 54, 45-48. 18. Altelaar, A. F.; Klinkert, I.; Jalink, K.; de Lange, R. P.; Adan, R. A.; Heeren, R. M.; Piersma, S. R. Goldenhanced biomolecular surface imaging of cells and tissue by SIMS and MALDI mass spectrometry. Anal Chem. 2006, 78, 734-742. 19. Sugiura, Y.; Shimma, S.; Setou, M. Two-step matrix application technique to improve ionization efficiency for matrix-assisted laser desorption/ionization in imaging mass spectrometry. Anal Chem. 2006, 78, 82278235. 20. Lemaire, R.; Tabet, J. C.; Ducoroy, P.; Hendra, J. B.; Salzet, M.; Fournier, I. Solid ionic matrixes for direct tissue analysis and MALDI imaging. Anal Chem. 2006, 78, 809-819. 21. Jurchen, J. C.; Rubakhin, S. S.; Sweedler, J. V. MALDI-MS imaging of features smaller than the size of the laser beam. J Am Soc Mass Spectrom. 2005, 16, 1654-1659. 22. Touboul, D.; Piednoel, H.; Voisin, V.; De La Porte, S.; Brunelle, A.; Halgand, F.; Laprevote, O. Changes of phospholipid composition within the dystrophic muscle by matrix-assisted laser desorption/ionization mass spectrometry and mass spectrometry imaging. Eur J Mass Spectrom (Chichester, Eng). 2004, 10, 657-664. 23. Groseclose, M. R.; Andersson, M.; Hardesty, W. M.; Caprioli, R. M. Identification of proteins directly from tissue: in situ tryptic digestions coupled with imaging mass spectrometry. J Mass Spectrom. 2007, 42, 254262. 24. Hsieh, Y.; Chen, J.; Korfmacher, W. A. Mapping pharmaceuticals in tissues using MALDI imaging mass spectrometry. J Pharmacol Toxicol Methods. 2007, 55, 193-200. 25. Drexler, D. M.; Garrett, T. J.; Cantone, J. L.; Diters, R. W.; Mitroka, J. G.; Prieto Conaway, M. C.; Adams, S. P.; Yost, R. A.; Sanders, M. Utility of imaging mass spectrometry (IMS) by matrix-assisted laser desorption ionization (MALDI) on an ion trap mass spectrometer in the analysis of drugs and metabolites in biological tissues. J Pharmacol Toxicol Methods. 2007, 55, 279-288. 26. Monroe, E. B.; Annangudi, S. P.; Hatcher, N. G.; Gutstein, H. B.; Rubakhin, S. S.; Sweedler, J. V. SIMS and MALDI MS imaging of the spinal cord. PROTEOMICS. 2008, 8, 3746-3754. 27. Kruse, R.; Sweedler, J. V. Spatial profiling invertebrate ganglia using MALDI MS. J Am Soc Mass Spectrom. 2003, 14, 752-759. 28. Chen, R.; Hui, L.; Sturm, R. M.; Li, L. Three dimensional mapping of neuropeptides and lipids in crustacean brain by mass spectral imaging. Journal of the American Society for Mass Spectrometry. 2009, 20, 10681077. 29. Rubakhin, S. S.; Sweedler, J. V. Characterizing peptides in individual mammalian cells using mass spectrometry. Nat. Protocols. 2007, 2, 1987-1997. 30. Garden, R. W.; Moroz, L. L.; Moroz, T. P.; Shippy, S. A.; Sweedler, J. V. Excess salt removal with matrix rinsing: direct peptide profiling of neurons from marine invertebrates using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. J Mass Spectrom. 1996, 31, 1126-1130. 99 31. Li, L.; Romanova, E. V.; Rubakhin, S. S.; Alexeeva, V.; Weiss, K. R.; Vilim, F. S.; Sweedler, J. V. Peptide profiling of cells with multiple gene products: combining immunochemistry and MALDI mass spectrometry with on-plate microextraction. Anal Chem. 2000, 72, 3867-3874. 32. Kutz, K. K.; Schmidt, J. J.; Li, L. In situ tissue analysis of neuropeptides by MALDI FTMS in-cell accumulation. Anal Chem. 2004, 76, 5630-5640. 33. Romanova, E. V.; Rubakhin, S. S.; Sweedler, J. V. One-Step Sampling, Extraction, and Storage Protocol for Peptidomics Using Dihydroxybenzoic Acid. Analytical Chemistry. 2008, 80, 3379-3386. 34. Predel, R. Peptidergic neurohemal system of an insect: mass spectrometric morphology. J Comp Neurol. 2001, 436, 363-375. 35. Wang, J.; Chen, R.; Ma, M.; Li, L. MALDI MS sample preparation by using paraffin wax film: systematic study and application for peptide analysis. Anal Chem. 2008, 80, 491-500. 36. Wang, J.; Jiang, X.; Sturm, R. M.; Li, L. Combining tissue extraction and off-line capillary electrophoresis matrix-assisted laser desorption/ionization Fourier transform mass spectrometry for neuropeptide analysis in individual neuronal organs using 2,5-dihydroxybenzoic acid as a multi-functional agent. J Chromatogr A. 2009, 1216, 8283-8288. 37. Monroe, E. B.; Jurchen, J. C.; Koszczuk, B. A.; Losh, J. L.; Rubakhin, S. S.; Sweedler, J. V. Massively Parallel Sample Preparation for the MALDI MS Analyses of Tissues. Analytical Chemistry. 2006, 78, 6826-6832. 38. Jimenez, C. R.; van Veelen, P. A.; Li, K. W.; Wildering, W. C.; Geraerts, W. P.; Tjaden, U. R.; van der Greef, J. Neuropeptide expression and processing as revealed by direct matrix-assisted laser desorption ionization mass spectrometry of single neurons. J Neurochem. 1994, 62, 404-407. 39. Li, K. W.; Hoek, R. M.; Smith, F.; Jimenez, C. R.; van der Schors, R. C.; van Veelen, P. A.; Chen, S.; van der Greef, J.; Parish, D. C.; Benjamin, P. R.; et al. Direct peptide profiling by mass spectrometry of single identified neurons reveals complex neuropeptide-processing pattern. J Biol Chem. 1994, 269, 30288-30292. 40. Li, K. W.; Jimenez, C. R.; Van Veelen, P. A.; Geraerts, W. P. Processing and targeting of a molluscan egglaying peptide prohormone as revealed by mass spectrometric peptide fingerprinting and peptide sequencing. Endocrinology. 1994, 134, 1812-1819. 41. Floyd, P. D.; Li, L.; Rubakhin, S. S.; Sweedler, J. V.; Horn, C. C.; Kupfermann, I.; Alexeeva, V. Y.; Ellis, T. A.; Dembrow, N. C.; Weiss, K. R.; Vilim, F. S. Insulin prohormone processing, distribution, and relation to metabolism in Aplysia californica. J Neurosci. 1999, 19, 7732-7741. 42. Garden, R. W.; Moroz, T. P.; Gleeson, J. M.; Floyd, P. D.; Li, L.; Rubakhin, S. S.; Sweedler, J. V. Formation of N-pyroglutamyl peptides from N-Glu and N-Gln precursors in Aplysia neurons. J Neurochem. 1999, 72, 676-681. 43. Li, L.; Garden, R. W.; Sweedler, J. V. Single-cell MALDI: a new tool for direct peptide profiling. Trends in Biotechnology. 2000, 18, 151-160. 44. Hummon, A. B.; Amare, A.; Sweedler, J. V. Discovering new invertebrate neuropeptides using mass spectrometry. Mass Spectrom Rev. 2006, 25, 77-98. 45. Ma, P. W. K.; Garden, R. W.; Niermann, J. T.; O' Connor, M.; Sweedler, J. V.; Roelofs, W. L. Characterizing the Hez-PBAN gene products in neuronal clusters with immunocytochemistry and MALDI MS. Journal of Insect Physiology. 2000, 46, 221-230. 46. Neupert, S.; Predel, R. Mass spectrometric analysis of single identified neurons of an insect. Biochemical and Biophysical Research Communications. 2005, 327, 640-645. 47. Neupert, S.; Schattschneider, S.; Predel, R. Allatotropin-related peptide in cockroaches: Identification via mass spectrometric analysis of single identified neurons. Peptides. 2009, 30, 489-494. 48. Neupert, S.; Predel, R.; Russell, W. K.; Davies, R.; Pietrantonio, P. V.; Nachman, R. J. Identification of tick periviscerokinin, the first neurohormone of Ixodidae: Single cell analysis by means of MALDI-TOF/TOF mass spectrometry. Biochemical and Biophysical Research Communications. 2005, 338, 1860-1864. 49. Rubakhin, S. S.; Garden, R. W.; Fuller, R. R.; Sweedler, J. V. Measuring the peptides in individual organelles with mass spectrometry. Nat Biotechnol. 2000, 18, 172-175. 50. Jarecki, J. L.; Andersen, K.; Konop, C. J.; Knickelbine, J. J.; Vestling, M. M.; Stretton, A. O. Mapping Neuropeptide Expression by Mass Spectrometry in Single Dissected Identified Neurons from the Dorsal Ganglion of the Nematode Ascaris suum. ACS Chemical Neuroscience. 2010, 1, 505-519. 51. Rubakhin, S. S.; Churchill, J. D.; Greenough, W. T.; Sweedler, J. V. Profiling Signaling Peptides in Single Mammalian Cells Using Mass Spectrometry. Analytical Chemistry. 2006, 78, 7267-7272. 52. Winther, A. M.; Siviter, R. J.; Isaac, R. E.; Predel, R.; Nassel, D. R. Neuronal expression of tachykinin-related peptides and gene transcript during postembryonic development of Drosophila. J Comp Neurol. 2003, 464, 180-196. 100 53. Yew, J. Y.; Dikler, S.; Stretton, A. O. De novo sequencing of novel neuropeptides directly from Ascaris suum tissue using matrix-assisted laser desorption/ionization time-of-flight/time-of-flight. Rapid Commun Mass Spectrom. 2003, 17, 2693-2698. 54. Li, L.; Pulver, S. R.; Kelley, W. P.; Thirumalai, V.; Sweedler, J. V.; Marder, E. Orcokinin peptides in developing and adult crustacean stomatogastric nervous systems and pericardial organs. J Comp Neurol. 2002, 444, 227-244. 55. Li, L.; Kelley, W. P.; Billimoria, C. P.; Christie, A. E.; Pulver, S. R.; Sweedler, J. V.; Marder, E. Mass spectrometric investigation of the neuropeptide complement and release in the pericardial organs of the crab, Cancer borealis. J Neurochem. 2003, 87, 642-656. 56. Ma, M.; Chen, R.; Ge, Y.; He, H.; Marshall, A. G.; Li, L. Combining bottom-up and top-down mass spectrometric strategies for de novo sequencing of the crustacean hyperglycemic hormone from Cancer borealis. Anal Chem. 2009, 81, 240-247. 57. Ma, M.; Chen, R.; Sousa, G. L.; Bors, E. K.; Kwiatkowski, M. A.; Goiney, C. C.; Goy, M. F.; Christie, A. E.; Li, L. Mass spectral characterization of peptide transmitters/hormones in the nervous system and neuroendocrine organs of the American lobster Homarus americanus. Gen Comp Endocrinol. 2008, 156, 395-409. 58. Stemmler, E. A.; Cashman, C. R.; Messinger, D. I.; Gardner, N. P.; Dickinson, P. S.; Christie, A. E. High-massresolution direct-tissue MALDI-FTMS reveals broad conservation of three neuropeptides (APSGFLGMRamide, GYRKPPFNGSIFamide and pQDLDHVFLRFamide) across members of seven decapod crustaean infraorders. Peptides. 2007, 28, 2104-2115. 59. Ma, M.; Wang, J.; Chen, R.; Li, L. Expanding the Crustacean neuropeptidome using a multifaceted mass spectrometric approach. J Proteome Res. 2009, 8, 2426-2437. 60. Stemmler, E. A.; Peguero, B.; Bruns, E. A.; Dickinson, P. S.; Christie, A. E. Identification, physiological actions, and distribution of TPSGFLGMRamide: a novel tachykinin-related peptide from the midgut and stomatogastric nervous system of Cancer crabs. Journal of Neurochemistry. 2007, 101, 1351-1366. 61. Myasein, K. T. Methods for peptide analysis of sub-microliter biological samples using MALDI-TOF mass spectrometry. Chicago, IL: University of Illinois at Chicago; 2009. 62. Ma, M.; Kutz-Naber, K. K.; Li, L. Methyl esterification assisted MALDI FTMS characterization of the orcokinin neuropeptide family. Anal Chem. 2007, 79, 673-681. 63. Ma, M.; Sturm, R. M.; Kutz-Naber, K. K.; Fu, Q.; Li, L. Immunoaffinity-based mass spectrometric characterization of the FMRFamide-related peptide family in the pericardial organ of Cancer borealis. Biochem Biophys Res Commun. 2009, 390, 325-330. 64. Ma, M.; Szabo, T. M.; Jia, C.; Marder, E.; Li, L. Mass spectrometric characterization and physiological actions of novel crustacean C-type allatostatins. Peptides. 2009, 30, 1660-1668. 65. Saideman, S. R.; Ma, M.; Kutz-Naber, K. K.; Cook, A.; Torfs, P.; Schoofs, L.; Li, L.; Nusbaum, M. P. Modulation of rhythmic motor activity by pyrokinin peptides. J Neurophysiol. 2007, 97, 579-595. 66. Wang, J.; Ma, M.; Chen, R.; Li, L. Enhanced neuropeptide profiling via capillary electrophoresis off-line coupled with MALDI FTMS. Anal Chem. 2008, 80, 6168-6177. 67. Chen, R.; Ma, M.; Hui, L.; Zhang, J.; Li, L. Measurement of neuropeptides in crustacean hemolymph via MALDI mass spectrometry. J Am Soc Mass Spectrom. 2009, 20, 708-718. 68. Behrens, H. L.; Chen, R.; Li, L. Combining microdialysis, NanoLC-MS, and MALDI-TOF/TOF to detect neuropeptides secreted in the crab, Cancer borealis. Anal Chem. 2008, 80, 6949-6958. 69. Behrens, H. L.; Li, L. Monitoring neuropeptides in vivo via microdialysis and mass spectrometry. Methods Mol Biol. 2010, 615, 57-73. 70. Falth, M.; Skold, K.; Norrman, M.; Svensson, M.; Fenyo, D.; Andren, P. E. SwePep, a database designed for endogenous peptides and mass spectrometry. Mol. Cell Proteomics. 2006, 5, 998-1005. 71. Amare, A.; Hummon, A. B.; Southey, B. R.; Zimmerman, T. A.; Rodriguez-Zas, S. L.; Sweedler, J. V. Bridging neuropeptidomics and genomics with bioinformatics: Prediction of mammalian neuropeptide prohormone processing. J. Proteome Res. 2006, 5, 1162-1167. 72. Hummon, A. B.; Hummon, N. P.; Corbin, R. W.; Li, L.; Vilim, F. S.; Weiss, K. R.; Sweedler, J. V. From precursor to final peptides: a statistical sequence-based approach to predicting prohormone processing. J. Proteome Res. 2003, 2, 650-656. 73. Southey, B. R.; Amare, A.; Zimmerman, T. A.; Rodriguez-Zas, S. L.; Sweedler, J. V. NeuroPred: a tool to predict cleavage sites in neuropeptide precursors and provide the masses of the resulting peptides. Nucleic Acids Res. 2006, 34, W267-272. 101 74. Ducret, A.; Van Oostveen, I.; Eng, J. K.; Yates, J. R., 3rd; Aebersold, R. High throughput protein characterization by automated reverse-phase chromatography/electrospray tandem mass spectrometry. Protein Sci. 1998, 7, 706-719. 75. Xu, C.; Ma, B. Software for computational peptide identification from MS-MS data. Drug Discov Today. 2006, 11, 595-600. 76. Mann, M.; Wilm, M. Error-tolerant identification of peptides in sequence databases by peptide sequence tags. Anal. Chem. 1994, 66, 4390-4399. 77. Baggerman, G.; Liu, F.; Wets, G.; Schoofs, L. Bioinformatic analysis of Peptide precursor proteins. Ann. N.Y. Acad. Sci. . 2005, 1040, 59-65. 78. Resing, K. A.; Meyer-Arendt, K.; Mendoza, A. M.; Aveline-Wolf, L. D.; Jonscher, K. R.; Pierce, K. G.; Old, W. M.; Cheung, H. T.; Russell, S.; Wattawa, J. L.; Goehle, G. R.; Knight, R. D.; Ahn, N. G. Improving reproducibility and sensitivity in identifying human proteins by shotgun proteomics. Anal Chem. 2004, 76, 3556-3568. 79. Stepanyan, R.; Day, K.; Urban, J.; Hardin, D. L.; Shetty, R. S.; Derby, C. D.; Ache, B. W.; McClintock, T. S. Gene expression and specificity in the mature zone of the lobster olfactory organ. Physiol Genomics. 2006, 25, 224-233. 80. Yamano, K.; Unuma, T. Expressed sequence tags from eyestalk of kuruma prawn, Marsupenaeus japonicus. Comp Biochem Physiol A Mol Integr Physiol. 2006, 143, 155-161. 81. Christie, A. E. Neuropeptide discovery in Ixodoidea: an in silico investigation using publicly accessible expressed sequence tags. Gen Comp Endocrinol. 2008, 157, 174-185. 82. Christie, A. E. In silico analyses of peptide paracrines/hormones in Aphidoidea. Gen Comp Endocrinol. 2008, 159, 67-79. 83. Clynen, E.; Huybrechts, J.; Verleyen, P.; De Loof, A.; Schoofs, L. Annotation of novel neuropeptide precursors in the migratory locust based on transcript screening of a public EST database and mass spectrometry. BMC Genomics. 2006, 7, 201. 84. Christie, A. E.; Cashman, C. R.; Brennan, H. R.; Ma, M.; Sousa, G. L.; Li, L.; Stemmler, E. A.; Dickinson, P. S. Identification of putative crustacean neuropeptides using in silico analyses of publicly accessible expressed sequence tags. Gen Comp Endocrinol. 2008, 156, 246-264. 85. Christie, A. E.; McCoole, M. D.; Harmon, S. M.; Baer, K. N.; Lenz, P. H. Genomic analyses of the Daphnia pulex peptidome. Gen Comp Endocrinol. 2011, 171, 131-150. 86. Dircksen, H.; Neupert, S.; Predel, R.; Verleyen, P.; Huybrechts, J.; Strauss, J.; Hauser, F.; Stafflinger, E.; Schneider, M.; Pauwels, K.; Schoofs, L.; Grimmelikhuijzen, C. J. Genomics, transcriptomics, and peptidomics of Daphnia pulex neuropeptides and protein hormones. J Proteome Res. 2011, 10, 4478-4504. 87. Gard, A. L.; Lenz, P. H.; Shaw, J. R.; Christie, A. E. Identification of putative peptide paracrines/hormones in the water flea Daphnia pulex (Crustacea; Branchiopoda; Cladocera) using transcriptomics and immunohistochemistry. Gen Comp Endocrinol. 2009, 160, 271-287. 88. Marco, H. G.; Gade, G. Biological activity of the predicted red pigment-concentrating hormone of Daphnia pulex in a crustacean and an insect. Gen Comp Endocrinol. 2010, 166, 104-110. 89. Price, D. A.; Greenberg, M. J. Structure of a molluscan cardioexcitatory neuropeptide. Science. 1977, 197, 670671. 90. Lopez Vera, E.; Aguilar, M. B.; Heimer de la Cotera, E. P. FMRFamide and related peptides in the phylum mollusca. Peptides. 2008, 29, 310-317. 91. Li, C.; Kim, K.; Nelson, L. S. FMRFamide-related neuropeptide gene family in Caenorhabditis elegans. Brain Res. 1999, 848, 26-34. 92. Kim, K.; Li, C. Expression and regulation of an FMRFamide-related neuropeptide gene family in Caenorhabditis elegans. J Comp Neurol. 2004, 475, 540-550. 93. McVeigh, P.; Leech, S.; Mair, G. R.; Marks, N. J.; Geary, T. G.; Maule, A. G. Analysis of FMRFamide-like peptide (FLP) diversity in phylum Nematoda. Int J Parasitol. 2005, 35, 1043-1060. 94. Husson, S. J.; Clynen, E.; Baggerman, G.; De Loof, A.; Schoofs, L. Discovering neuropeptides in Caenorhabditis elegans by two dimensional liquid chromatography and mass spectrometry. Biochemical and Biophysical Research Communications. 2005, 335, 76-86. 95. Fujii, K.; Ohta, N.; Sasaki, T.; Sezikawa, Y.; Yamada, C.; Kobayashi, H. Immunoreactive FMRFamide in the central nervous system of the earthworm, Eisenia foetida. Zool. Sci. 1989, 6, 951-961. 96. Michel Salzet, P. B. C. W. J. M. FMRFamide-related peptides in the sex segmental ganglia of the Pharyngobdellid leech Erpobdella octoculata. European Journal of Biochemistry. 1994, 221, 269-275. 102 97. Jacobs, A. A.; Van Herp, F. Immunocytochemical localization of a substance in the eyestalk of the prawn, Palaemon serratus, reactive with an anti-FMRFamide rabbit serum. Cell Tissue Res. 1984, 235, 601-605. 98. Trimmer, B. A.; Kobierski, L. A.; Kravitz, E. A. Purification and characterization of FMRFamide like immunoreactive substances from the lobster nervous system: isolation and sequence analysis of two closely related peptides. . J Comp Neurol. 1987, 266, 16-26. 99. Sithigorngul, P.; Pupuem, J.; Krungkasem, C.; Longyant, S.; Chaivisuthangkura, P.; Sithigorngul, W.; Petsom, A. Seven novel FMRFamide-like neuropeptide sequences from the eyestalk of the giant tiger prawn Penaeus monodon. Comp Biochem Physiol B Biochem Mol Biol. 2002, 131, 325-337. 100. Sithigorngul, P.; Saraithongkum, W.; Longyant, S.; Panchan, N.; Sithigorngul, W.; Petsom, A. Three more novel FMRFamide-like neuropeptide sequences from the eyestalk of the giant freshwater prawn Macrobrachium rosenbergii*. Peptides. 2001, 22, 191-197. 101. Fu, Q.; Kutz, K. K.; Schmidt, J. J.; Hsu, Y. W.; Messinger, D. I.; Cain, S. D.; de la Iglesia, H. O.; Christie, A. E.; Li, L. Hormone complement of the Cancer productus sinus gland and pericardial organ: an anatomical and mass spectrometric investigation. J Comp Neurol. 2005, 493, 607-626. 102. Ma, M.; Bors, E. K.; Dickinson, E. S.; Kwiatkowski, M. A.; Sousa, G. L.; Henry, R. P.; Smith, C. M.; Towle, D. W.; Christie, A. E.; Li, L. Characterization of the Carcinus maenas neuropeptidome by mass spectrometry and functional genomics. General and Comparative Endocrinology. 2009, 161, 320-334. 103. Huybrechts, J.; Nusbaum, M. P.; Bosch, L. V.; Baggerman, G.; Loof, A. D.; Schoofs, L. Neuropeptidomic analysis of the brain and thoracic ganglion from the Jonah crab, Cancer borealis. Biochemical and Biophysical Research Communications. 2003, 308, 535-544. 104. Predel, R.; Neupert, S.; Wicher, D.; Gundel, M.; Roth, S.; Derst, C. Unique accumulation of neuropeptides in an insect: FMRFamide-related peptides in the cockroach, Periplaneta americana. European Journal of Neuroscience. 2004, 20, 1499-1513. 105. Verleyen, P.; Huybrechts, J.; Sas, F.; Clynen, E.; Baggerman, G.; De Loof, A.; Schoofs, L. Neuropeptidomics of the grey flesh fly, Neobellieria bullata. Biochemical and Biophysical Research Communications. 2004, 316, 763-770. 106. Baggerman, G.; Boonen, K.; Verleyen, P.; Loof, A. D.; Schoofs, L. Peptidomic analysis of the larval Drosophila melanogaster central nervous system by two-dimensional capillary liquid chromatography quadrupole time-of-flight mass spectrometry. Journal of Mass Spectrometry. 2005, 40, 250-260. 107. Dickinson, P. S.; Stevens, J. S.; Rus, S.; Brennan, H. R.; Goiney, C. C.; Smith, C. M.; Li, L.; Towle, D. W.; Christie, A. E. Identification and cardiotropic actions of sulfakinin peptides in the American lobster Homarus americanus. J Exp Biol. 2007, 210, 2278-2289. 108. Audsley, N.; Weaver, R. J. Neuropeptides associated with the regulation of feeding in insects. General and Comparative Endocrinology. 2009, 162, 93-104. 109. Nelson, L. S.; Kim, K.; Memmott, J. E.; Li, C. FMRFamide-related gene family in the nematode, Caenorhabditis elegans. Brain Res Mol Brain Res. 1998, 58, 103-111. 110. Mercier, A. J.; Friedrich, R.; Boldt, M. Physiological functions of FMRFamide-like peptides (FLPs) in crustaceans. Microscopy Research and Technique. 2003, 60, 313-324. 111. Lorenzo, G.; Massimiliano, B.; Sara, F.; Laura, G.; Paola, R.; Grazia, T. Distribution of FMRFamide-like immunoreactivity in the alimentary tract and hindgut ganglia of the barnacle Balanus amphitrite (Cirripedia, Crustacea). Microscopy Research and Technique. 2006, 69, 636-641. 112. O'Gara, B. A.; Brown, P. L.; Dlugosch, D.; Kandiel, A.; Ku, J. W.; Geier, J. K.; Henggeler, N. C.; Abbasi, A.; Kounalakis, N. Regulation of pharyngeal motility by FMRFamide and related peptides in the medicinal leech, Hirudo medicinalis. Invertebrate Neuroscience. 1999, 4, 41-53. 113. Duttlinger, A.; Berry, K.; Nichols, R. The different effects of three Drosophila melanogaster dFMRFamidecontaining peptides on crop contractions suggest these structurally related peptides do not play redundant functions in gut. Peptides. 2002, 23, 1953-1957. 114. Kriegsfeld, L. J. Driving reproduction: RFamide peptides behind the wheel. Hormones and Behavior. 2006, 50, 655-666. 115. Di Cristo, C.; Bovi, P. D.; Di Cosmo, A. Role of FMRFamide in the reproduction of Octopus vulgaris: molecular analysis and effect on visual input. Peptides. 2003, 24, 1525-1532. 116. Henry, J.; Zatylny, C.; Boucaud-Camou, E. Peptidergic control of egg-laying in the cephalopod Sepia officinalis: involvement of FMRFamide and FMRFamide-related peptides. Peptides. 1999, 20, 1061-1070. 117. Ringstad, N.; Horvitz, H. R. FMRFamide neuropeptides and acetylcholine synergistically inhibit egg-laying by C. elegans. Nat Neurosci. 2008, 11, 1168-1176. 103 118. Turrigiano, G. G.; Selverston, A. I. A cholecystokinin-like hormone activates a feeding-related neural circuit in lobster. Nature. 1990, 344, 866-868. 119. Shetreat-Klein, A. N.; Cropper, E. C. Afferent-induced changes in rhythmic motor programs in the feeding circuitry of aplysia. J Neurophysiol. 2004, 92, 2312-2322. 120. Hill, S. R.; Orchard, I. The influence of diet and feeding state on FMRFamide-related peptides in the gut of Locusta migratoria L. Peptides. 2004, 25, 105-114. 121. Vilaplana, L.; Pascual, N.; Perera, N.; Leira, D.; Belles, X. Antifeeding properties of myosuppressin in a generalist phytophagous leafworm, Spodoptera littoralis (Boisduval). Regulatory Peptides. 2008, 148, 6875. 122. Wei, Z.; Baggerman, G.; R, J. N.; Goldsworthy, G.; Verhaert, P.; De Loof, A.; Schoofs, L. Sulfakinins reduce food intake in the desert locust, Schistocerca gregaria. J Insect Physiol. 2000, 46, 1259-1265. 123. Aguilar, R.; Maestro, J. L.; Vilaplana, L.; Chiva, C.; Andreu, D.; Belles, X. Identification of leucomyosuppressin in the German cockroach, Blattella germanica, as an inhibitor of food intake. Regul Pept. 2004, 119, 105-112. 124. Lingueglia, E.; Champigny, G.; Lazdunski, M.; Barbry, P. Cloning of the amiloride-sensitive FMRFamide peptide-gated sodium channel. Nature. 1995, 378, 730-733. 125. Lingueglia, E.; Deval, E.; Lazdunski, M. FMRFamide-gated sodium channel and ASIC channels: a new class of ionotropic receptors for FMRFamide and related peptides. Peptides. 2006, 27, 1138-1152. 126. Qiu, J.; Fang, Y.; Bosch, M. A.; Ronnekleiv, O. K.; Kelly, M. J. Guinea pig kisspeptin neurons are depolarized by leptin via activation of TRPC channels. Endocrinology. 2011, 152, 1503-1514. 127. Zhang, C.; Roepke, T. A.; Kelly, M. J.; Ronnekleiv, O. K. Kisspeptin depolarizes gonadotropin-releasing hormone neurons through activation of TRPC-like cationic channels. J Neurosci. 2008, 28, 4423-4434. 128. Zhang, X. B.; Spergel, D. J. Kisspeptin inhibits high-voltage activated ca2+ channels in GnRH neurons via multiple ca2+ influx and release pathways. Neuroendocrinology. 2012, 96, 68-80. 129. Pratt, G. E.; Farnsworth, D. E.; Siegel, N. R.; Fok, K. F.; Feyereisen, R. Identification of an allatostatin from adult Diploptera punctata. Biochem Biophys Res Commun. 1989, 163, 1243-1247. 130. Dircksen, H.; Skiebe, P.; Abel, B.; Agricola, H.; Buchner, K.; Muren, J. E.; Nassel, D. R. Structure, distribution, and biological activity of novel members of the allatostatin family in the crayfish Orconectes limosus. Peptides. 1999, 20, 695-712. 131. Pratt, G. E.; Farnsworth, D. E.; Fok, K. F.; Siegel, N. R.; McCormack, A. L.; Shabanowitz, J.; Hunt, D. F.; Feyereisen, R. Identity of a second type of allatostatin from cockroach brains: an octadecapeptide amide with a tyrosine-rich address sequence. Proc Natl Acad Sci U S A. 1991, 88, 2412-2416. 132. Woodhead, A. P.; Stay, B.; Seidel, S. L.; Khan, M. A.; Tobe, S. S. Primary structure of four allatostatins: neuropeptide inhibitors of juvenile hormone synthesis. Proc Natl Acad Sci U S A. 1989, 86, 5997-6001. 133. Lorenz, M. W.; Kellner, R.; Hoffmann, K. H. A family of neuropeptides that inhibit juvenile hormone biosynthesis in the cricket, Gryllus bimaculatus. J Biol Chem. 1995, 270, 21103-21108. 134. Kramer, S. J.; Toschi, A.; Miller, C. A.; Kataoka, H.; Quistad, G. B.; Li, J. P.; Carney, R. L.; Schooley, D. A. Identification of an allatostatin from the tobacco hornworm Manduca sexta. Proc Natl Acad Sci U S A. 1991, 88, 9458-9462. 135. Stay, B.; Tobe, S. S. The role of allatostatins in juvenile hormone synthesis in insects and crustaceans. Annu Rev Entomol. 2007, 52, 277-299. 136. Stay, B.; Fairbairn, S.; Yu, C. G. Role of allatostatins in the regulation of juvenile hormone synthesis. Arch Insect Biochem Physiol. 1996, 32, 287-297. 137. Bendena, W. G.; Garside, C. S.; Yu, C. G.; Tobe, S. S. Allatostatins: diversity in structure and function of an insect neuropeptide family. Ann N Y Acad Sci. 1997, 814, 53-66. 138. Bendena, W. G.; Donly, B. C.; Tobe, S. S. Allatostatins: a growing family of neuropeptides with structural and functional diversity. Ann N Y Acad Sci. 1999, 897, 311-329. 139. Duve, H.; Johnsen, A. H.; Maestro, J. L.; Scott, A. G.; Jaros, P. P.; Thorpe, A. Isolation and identification of multiple neuropeptides of the allatostatin superfamily in the shore crab Carcinus maenas. Eur J Biochem. 1997, 250, 727-734. 140. Duve, H.; Johnsen, A. H.; Scott, A. G.; Thorpe, A. Allatostatins of the tiger prawn, Penaeus monodon (Crustacea: Penaeidea). Peptides. 2002, 23, 1039-1051. 141. Christie, A. E.; Sousa, G. L.; Rus, S.; Smith, C. M.; Towle, D. W.; Hartline, D. K.; Dickinson, P. S. Identification of A-type allatostatins possessing -YXFGI/Vamide carboxy-termini from the nervous system of the copepod crustacean Calanus finmarchicus. Gen Comp Endocrinol. 2008, 155, 526-533. 104 142. Yin, G. L.; Yang, J. S.; Cao, J. X.; Yang, W. J. Molecular cloning and characterization of FGLamide allatostatin gene from the prawn, Macrobrachium rosenbergii. Peptides. 2006, 27, 1241-1250. 143. Fu, Q.; Tang, L. S.; Marder, E.; Li, L. Mass spectrometric characterization and physiological actions of VPNDWAHFRGSWamide, a novel B type allatostatin in the crab, Cancer borealis. J Neurochem. 2007, 101, 1099-1107. 144. Dickinson, P. S.; Wiwatpanit, T.; Gabranski, E. R.; Ackerman, R. J.; Stevens, J. S.; Cashman, C. R.; Stemmler, E. A.; Christie, A. E. Identification of SYWKQCAFNAVSCFamide: a broadly conserved crustacean Ctype allatostatin-like peptide with both neuromodulatory and cardioactive properties. J Exp Biol. 2009, 212, 1140-1152. 145. Skiebe, P.; Schneider, H. Allatostatin peptides in the crab stomatogastric nervous system: inhibition of the pyloric motor pattern and distribution of allatostatin-like immunoreactivity. J Exp Biol. 1994, 194, 195208. 146. Skiebe, P. Allatostatin-like immunoreactivity in the stomatogastric nervous system and the pericardial organs of the crab Cancer pagurus, the lobster Homarus americanus, and the crayfish Cherax destructor and Procambarus clarkii. J Comp Neurol. 1999, 403, 85-105. 147. Panchan, N.; Bendena, W. G.; Bowser, P.; Lungchukiet, P.; Tobe, S. S.; Sithigorngul, W.; Chaivisuthangkura, P.; Rangsiruji, A.; Petsom, A.; Pewnim, T.; Sithigorngul, P. Immunolocalization of allatostatin-like neuropeptides and their putative receptor in eyestalks of the tiger prawn, Penaeus monodon. Peptides. 2003, 24, 1563-1570. 148. Kwok, R.; Rui Zhang, J.; Tobe, S. S. Regulation of methyl farnesoate production by mandibular organs in the crayfish, Procambarus clarkii: a possible role for allatostatins. J Insect Physiol. 2005, 51, 367-378. 149. Auerswald, L.; Birgul, N.; Gade, G.; Kreienkamp, H. J.; Richter, D. Structural, functional, and evolutionary characterization of novel members of the allatostatin receptor family from insects. Biochem Biophys Res Commun. 2001, 282, 904-909. 150. Jorge-Rivera, J.; Marder, Y. E. Allatostatin decreases stomatogastric neuromuscular transmission in the crab Cancer borealis. J Exp Biol. 1997, 200, 2937-2946. 151. Kreissl, S.; Weiss, T.; Djokaj, S.; Balezina, O.; Rathmayer, W. Allatostatin modulates skeletal muscle performance in crustaceans through pre- and postsynaptic effects. Eur J Neurosci. 1999, 11, 2519-2530. 152. Cruz-Bermudez, N. D.; Marder, E. Multiple modulators act on the cardiac ganglion of the crab, Cancer borealis. J Exp Biol. 2007, 210, 2873-2884. 153. Birmingham, J. T.; Billimoria, C. P.; DeKlotz, T. R.; Stewart, R. A.; Marder, E. Differential and historydependent modulation of a stretch receptor in the stomatogastric system of the crab, Cancer borealis. J Neurophysiol. 2003, 90, 3608-3616. 154. Lange, A. B.; Chan, K. K.; Stay, B. Effect of allatostatin and proctolin on antennal pulsatile organ and hindgut muscle in the cockroach, Diploptera punctata. Arch Insect Biochem Physiol. 1993, 24, 79-92. 155. Price, M. D.; Merte, J.; Nichols, R.; Koladich, P. M.; Tobe, S. S.; Bendena, W. G. Drosophila melanogaster flatline encodes a myotropin orthologue to Manduca sexta allatostatin. Peptides. 2002, 23, 787-794. 156. Rankin, S. M.; Stay, B.; Chan, K.; Jackson, E. S. Cockroach allatostatin-immunoreactive neurons and effects of cockroach allatostatin in earwigs. Arch Insect Biochem Physiol. 1998, 38, 155-165. 157. Stemmler, E. A.; Bruns, E. A.; Cashman, C. R.; Dickinson, P. S.; Christie, A. E. Molecular and mass spectral identification of the broadly conserved decapod crustacean neuropeptide pQIRYHQCYFNPISCF: The first PISCF-allatostatin (Manduca sexta- or C-type allatostatin) from a non-insect. Gen Comp Endocrinol. 2009. 158. Birgul, N.; Weise, C.; Kreienkamp, H. J.; Richter, D. Reverse physiology in drosophila: identification of a novel allatostatin-like neuropeptide and its cognate receptor structurally related to the mammalian somatostatin/galanin/opioid receptor family. EMBO J. 1999, 18, 5892-5900. 159. Hauser, F.; Cazzamali, G.; Williamson, M.; Blenau, W.; Grimmelikhuijzen, C. J. A review of neurohormone GPCRs present in the fruitfly Drosophila melanogaster and the honey bee Apis mellifera. Prog Neurobiol. 2006, 80, 1-19. 160. Lenz, C.; Williamson, M.; Grimmelikhuijzen, C. J. Molecular cloning and genomic organization of a second probable allatostatin receptor from Drosophila melanogaster. Biochem Biophys Res Commun. 2000, 273, 571-577. 161. Johnson, E. C.; Bohn, L. M.; Barak, L. S.; Birse, R. T.; Nassel, D. R.; Caron, M. G.; Taghert, P. H. Identification of Drosophila neuropeptide receptors by G protein-coupled receptors-beta-arrestin2 interactions. J Biol Chem. 2003, 278, 52172-52178. 162. Kreienkamp, H. J.; Larusson, H. J.; Witte, I.; Roeder, T.; Birgul, N.; Honck, H. H.; Harder, S.; Ellinghausen, G.; Buck, F.; Richter, D. Functional annotation of two orphan G-protein-coupled receptors, Drostar1 and - 105 2, from Drosophila melanogaster and their ligands by reverse pharmacology. J Biol Chem. 2002, 277, 39937-39943. 163. Lechner, H. A.; Lein, E. S.; Callaway, E. M. A genetic method for selective and quickly reversible silencing of Mammalian neurons. J Neurosci. 2002, 22, 5287-5290. 164. Tan, E. M.; Yamaguchi, Y.; Horwitz, G. D.; Gosgnach, S.; Lein, E. S.; Goulding, M.; Albright, T. D.; Callaway, E. M. Selective and quickly reversible inactivation of mammalian neurons in vivo using the Drosophila allatostatin receptor. Neuron. 2006, 51, 157-170. 165. Jacobs, S.; Schulz, S. Intracellular trafficking of somatostatin receptors. Mol Cell Endocrinol. 2008, 286, 58-62. 166. Satake, H.; Kawada, T.; Nomoto, K.; Minakata, H. Insight into Tachykinin-Related Peptides, Their Receptors, and Invertebrate Tachykinins: A review. Zoological Science. 2003, 20, 533-549. 167. Severini, C.; Improta, G.; Falconieri-Erspamer, G.; Salvadori, S.; Erspamer, V. The tachykinin peptide family. Pharmacol Rev. 2002, 54, 285-322. 168. Neupert, S.; Russell, W. K.; Russell, D. H.; Lopez Jr, J. D.; Predel, R.; Nachman, R. J. Neuropeptides in Heteroptera: Identification of allatotropin-related peptide and tachykinin-related peptides using MALDITOF mass spectrometry. Peptides. 2009, 30, 483-488. 169. Reinhard, P.; Susanne, N.; Steffen, R.; Christian, D.; Dick, R. N. Tachykinin-related peptide precursors in two cockroach species. FEBS Journal. 2005, 272, 3365-3375. 170. Audsley, N.; Weaver, R. J. Analysis of peptides in the brain and corpora cardiaca-corpora allata of the honey bee, Apis mellifera using MALDI-TOF mass spectrometry. Peptides. 2006, 27, 512-520. 171. Christie, A. E.; Lundquist, C. T.; Nassel, D. R.; Nusbaum, M. P. Two novel tachykinin-related peptides from the nervous system of the crab Cancer borealis. J Exp Biol. 1997, 200, 2279-2294. 172. Billimoria, C. P.; Li, L.; Marder, E. Profiling of neuropeptides released at the stomatogastric ganglion of the crab, Cancer borealis with mass spectrometry. Journal of Neurochemistry. 2005, 95, 191-199. 173. Messinger, D. I.; Kutz, K. K.; Le, T.; Verley, D. R.; Hsu, Y.-W. A.; Ngo, C. T.; Cain, S. D.; Birmingham, J. T.; Li, L.; Christie, A. E. Identification and characterization of a tachykinin-containing neuroendocrine organ in the commissural ganglion of the crab Cancer productus. J Exp Biol. 2005, 208, 3303-3319. 174. Christie, A. E.; Kutz-Naber, K. K.; Stemmler, E. A.; Klein, A.; Messinger, D. I.; Goiney, C. C.; Conterato, A. J.; Bruns, E. A.; Hsu, Y.-W. A.; Li, L.; Dickinson, P. S. Midgut epithelial endocrine cells are a rich source of the neuropeptides APSGFLGMRamide (Cancer borealis tachykinin-related peptide Ia) and GYRKPPFNGSIFamide (Gly1-SIFamide) in the crabs Cancer borealis, Cancer magister and Cancer productus. J Exp Biol. 2007, 210, 699-714. 175. Dickinson, P. S.; Stemmler, E. A.; Cashman, C. R.; Brennan, H. R.; Dennison, B.; Huber, K. E.; Peguero, B.; Rabacal, W.; Goiney, C. C.; Smith, C. M.; Towle, D. W.; Christie, A. E. SIFamide peptides in clawed lobsters and freshwater crayfish (Crustacea, Decapoda, Astacidea): a combined molecular, mass spectrometric and electrophysiological investigation. Gen Comp Endocrinol. 2008, 156, 347-360. 176. Pennefather, J. N.; Lecci, A.; Candenas, M. L.; Patak, E.; Pinto, F. M.; Maggi, C. A. Tachykinins and tachykinin receptors: a growing family. Life Sciences. 2004, 74, 1445-1463. 177. Petersen, P. S.; Woldbye, D. P. D.; Madsen, A. N.; Egerod, K. L.; Jin, C.; Lang, M.; Rasmussen, M.; BeckSickinger, A. G.; Holst, B. In Vivo Characterization of High Basal Signaling from the Ghrelin Receptor. Endocrinology. 2009, 150, 4920-4930. 178. Van Loy, T.; Vandersmissen, H. P.; Poels, J.; Van Hiel, M. B.; Verlinden, H.; Broeck, J. V. Tachykinin-related peptides and their receptors in invertebrates: A current view. Peptides. 2010, 31, 520-524. 179. Winther, A. M.; Acebes, A.; Ferrus, A. Tachykinin-related peptides modulate odor perception and locomotor activity in Drosophila. Molecular and Cellular Neuroscience. 2006, 31, 399-406. 180. Brockmann, A.; Annangudi, S. P.; Richmond, T. A.; Ament, S. A.; Xie, F.; Southey, B. R.; Rodriguez-Zas, S. R.; Robinson, G. E.; Sweedler, J. V. Quantitative peptidomics reveal brain peptide signatures of behavior. Proceedings of the National Academy of Sciences. 2009, 106, 2383-2388. 181. Pascual, N.; Maestro, J. L.; Chiva, C.; Andreu, D.; Bell 閟, X. Identification of a tachykinin-related peptide with orexigenic properties in the German cockroach. Peptides. 2008, 29, 386-392. 182. Casini, G.; Dal Monte, M.; Fornai, F.; Bosco, L.; Willems, D.; Yang, Q.; Zhou, Z. J.; Bagnoli, P. Neurokinin 1 receptor expression and substance P physiological actions are developmentally regulated in the rabbit retina. Neuroscience. 2004, 124, 147-160. 183. Rauscent, A.; Le Ray, D.; Cabirol-Pol, M. J.; Sillar, K. T.; Simmers, J.; Combes, D. Development and neuromodulation of spinal locomotor networks in the metamorphosing frog. J Physiol Paris. 2006, 100, 317-327. 106 184. Cape, S. S.; Rehm, K. J.; Ma, M.; Marder, E.; Li, L. Mass spectral comparison of the neuropeptide complement of the stomatogastric ganglion and brain in the adult and embryonic lobster, Homarus americanus. Journal of Neurochemistry. 2008, 105, 690-702. 185. Rehm, K. J.; Deeg, K. E.; Marder, E. Developmental Regulation of Neuromodulator Function in the Stomatogastric Ganglion of the Lobster, Homarus americanus. J. Neurosci. 2008, 28, 9828-9839. 186. Li, X. J.; Wolfgang, W.; Wu, Y. N.; North, R. A.; Forte, M. Cloning, heterologous expression and developmental regulation of a Drosophila receptor for tachykinin-like peptides. EMBO J. 1991, 10, 32213229. 187. Monnier, D.; Colas, J. F.; Rosay, P.; Hen, R.; Borrelli, E.; Maroteaux, L. NKD, a developmentally regulated tachykinin receptor in Drosophila. J Biol Chem. 1992, 267, 1298-1302. 188. Poels, J.; Birse, R. T.; Nachman, R. J.; Fichna, J.; Janecka, A.; Vanden Broeck, J.; Nasel, D. R. Characterization and distribution of NKD, a receptor for Drosophila tachykinin-related peptide 6. Peptides. 2009, 30, 545-556. 189. Torfs, H.; Oonk, H. B.; Broeck, J. V.; Poels, J.; Van Poyer, W.; De Loof, A.; Guerrero, F.; Meloen, R. H.; Akerman, K.; Nachman, R. J. Pharmacological characterization of STKR, an insect G protein-coupled receptor for tachykinin-like peptides. Arch Insect Biochem Physiol. 2001, 48, 39-49. 190. Kawada, T.; Furukawa, Y.; Shimizu, Y.; Minakata, H.; Nomoto, K.; Satake, H. A novel tachykinin-related peptide receptor. Sequence, genomic organization, and functional analysis. Eur J Biochem. 2002, 269, 4238-4246. 191. Atsuhiro, K.; Kyoko, T.-K.; Masato, A.; Honoo, S. A novel tachykinin-related peptide receptor of Octopus vulgaris -- evolutionary aspects of invertebrate tachykinin and tachykinin-related peptide. FEBS Journal. 2007, 274, 2229-2239. 192. Stangier, J.; Hilbich, C.; Burdzik, S.; Keller, R. Orcokinin: a novel myotropic peptide from the nervous system of the crayfish, Orconectes limosus. Peptides. 1992, 13, 859-864. 193. Bungart, D.; Dircksen, H.; Keller, R. Quantitative determination and distribution of the myotropic neuropeptide orcokinin in the nervous system of astacidean crustaceans. Peptides. 1994, 15, 393-400. 194. Yasuda-Kamatani, Y.; Yasuda, A. Identification of orcokinin gene-related peptides in the brain of the crayfish Procambarus clarkii by the combination of MALDI-TOF and on-line capillary HPLC/Q-Tof mass spectrometries and molecular cloning. Gen Comp Endocrinol. 2000, 118, 161-172. 195. Bungart, D.; Hilbich, C.; Dircksen, H.; Keller, R. Occurrence of analogues of the myotropic neuropeptide orcokinin in the shore crab, Carcinus maenas: evidence for a novel neuropeptide family. Peptides. 1995, 16, 67-72. 196. Skiebe, P.; Dreger, M.; Meseke, M.; Evers, J. F.; Hucho, F. Identification of orcokinins in single neurons in the stomatogastric nervous system of the crayfish, Cherax destructor. J Comp Neurol. 2002, 444, 245-259. 197. Fu, Q.; Goy, M. F.; Li, L. Identification of neuropeptides from the decapod crustacean sinus glands using nanoscale liquid chromatography tandem mass spectrometry. Biochem Biophys Res Commun. 2005, 337, 765-778. 198. Dickinson, P. S.; Stemmler, E. A.; Barton, E. E.; Cashman, C. R.; Gardner, N. P.; Rus, S.; Brennan, H. R.; McClintock, T. S.; Christie, A. E. Molecular, mass spectral, and physiological analyses of orcokinins and orcokinin precursor-related peptides in the lobster Homarus americanus and the crayfish Procambarus clarkii. Peptides. 2009, 30, 297-317. 199. Pascual, N.; Castresana, J.; Valero, M. L.; Andreu, D.; Belles, X. Orcokinins in insects and other invertebrates. Insect Biochem Mol Biol. 2004, 34, 1141-1146. 200. Dircksen, H.; Burdzik, S.; Sauter, A.; Keller, R. Two orcokinins and the novel octapeptide orcomyotropin in the hindgut of the crayfish Orconectes limosus: identified myostimulatory neuropeptides originating together in neurones of the terminal abdominal ganglion. J Exp Biol. 2000, 203, 2807-2818. 201. Skiebe, P.; Dreger, M.; Borner, J.; Meseke, M.; Weckwerth, W. Immunocytochemical and molecular data guide peptide identification by mass spectrometry: orcokinin and orcomyotropin-related peptides in the stomatogastric nervous system of several crustacean species. Cell Mol Biol (Noisy-le-grand). 2003, 49, 851-871. 202. Keller, R. Crustacean neuropeptides: structures, functions and comparative aspects. Experientia. 1992, 48, 439448. 203. Bungart, D.; Kegel, G.; Burdzik, S.; Keller, R. Structure-activity relationships of the crustacean myotropic neuropeptide orcokinin. Peptides. 1995, 16, 199-204. 204. Hofer, S.; Homberg, U. Orcokinin immunoreactivity in the accessory medulla of the cockroach Leucophaea maderae. Cell Tissue Res. 2006, 325, 589-600. 107 205. Hofer, S.; Homberg, U. Evidence for a role of orcokinin-related peptides in the circadian clock controlling locomotor activity of the cockroach Leucophaea maderae. J Exp Biol. 2006, 209, 2794-2803. 206. Stangier, J.; Hilbich, C.; Beyreuther, K.; Keller, R. Unusual cardioactive peptide (CCAP) from pericardial organs of the shore crab Carcinus maenas. Proc Natl Acad Sci U S A. 1987, 84, 575-579. 207. Stangier, J.; Hilbich, C.; Dircksen, H.; Keller, R. Distribution of a novel cardioactive neuropeptide (CCAP) in the nervous system of the shore crab Carcinus maenas. Peptides. 1988, 9, 795-800. 208. Groome, J. R.; Lehman, H. K. Characterization of crustacean cardioactive peptide-like immunoreactivity in the horseshoe crab, Limulus polyphemus. J Comp Neurol. 1995, 357, 36-51. 209. Skiebe, P.; Dietel, C.; Schmidt, M. Immunocytochemical localization of FLRFamide-, proctolin-, and CCAPlike peptides in the stomatogastric nervous system and neurohemal structures of the crayfish, Cherax destructor. J Comp Neurol. 1999, 414, 511-532. 210. McGaw, I. J.; Wilkens, J. L.; McMahon, B. R.; Airriess, C. N. Crustacean cardioexcitatory peptides may inhibit the heart in vivo. J Exp Biol. 1995, 198, 2547-2550. 211. Fort, T. J.; Garcia-Crescioni, K.; Agricola, H. J.; Brezina, V.; Miller, M. W. Regulation of the crab heartbeat by crustacean cardioactive peptide (CCAP): central and peripheral actions. J Neurophysiol. 2007, 97, 34073420. 212. Cooke, I. M. Reliable, responsive pacemaking and pattern generation with minimal cell numbers: the crustacean cardiac ganglion. Biol Bull. 2002, 202, 108-136. 213. Fort, T. J.; Brezina, V.; Miller, M. W. Modulation of an integrated central pattern generator-effector system: dopaminergic regulation of cardiac activity in the blue crab Callinectes sapidus. J Neurophysiol. 2004, 92, 3455-3470. 214. Gaus, G.; Stieve, H. The effect of neuropeptides on the ERG of the crayfish Orconectes limosus. Z Naturforsch C. 1992, 47, 300-303. 215. Richards, K. S.; Marder, E. The actions of crustacean cardioactive peptide on adult and developing stomatogastric ganglion motor patterns. J Neurobiol. 2000, 44, 31-44. 216. Weimann, J. M.; Skiebe, P.; Heinzel, H. G.; Soto, C.; Kopell, N.; Jorge-Rivera, J. C.; Marder, E. Modulation of oscillator interactions in the crab stomatogastric ganglion by crustacean cardioactive peptide. J Neurosci. 1997, 17, 1748-1760. 217. Phlippen, M. K.; Webster, S. G.; Chung, J. S.; Dircksen, H. Ecdysis of decapod crustaceans is associated with a dramatic release of crustacean cardioactive peptide into the haemolymph. J Exp Biol. 2000, 203, 521-536. 218. Cheung, C. C.; Loi, P. K.; Sylwester, A. W.; Lee, T. D.; Tublitz, N. J. Primary structure of a cardioactive neuropeptide from the tobacco hawkmoth, Manduca sexta. FEBS Lett. 1992, 313, 165-168. 219. Stangier, J.; Hilbich, C.; Keller, R. Occurrence of crustacean caridoactive peptide (CCAP) in the nervous system of an insect, Locusta migratoria. J Comp Physiol B. 1989, 159, 5-11. 220. Loi, P. K.; Emmal, S. A.; Park, Y.; Tublitz, N. J. Identification, sequence and expression of a crustacean cardioactive peptide (CCAP) gene in the moth Manduca sexta. J Exp Biol. 2001, 204, 2803-2816. 221. Lehman, H. K.; Murgiuc, C. M.; Miller, T. A.; Lee, T. D.; Hildebrand, J. G. Crustacean cardioactive peptide in the sphinx moth, Manduca sexta. Peptides. 1993, 14, 735-741. 222. Dulcis, D.; Levine, R. B.; Ewer, J. Role of the neuropeptide CCAP in Drosophila cardiac function. J Neurobiol. 2005, 64, 259-274. 223. Wasielewski, O.; Skonieczna, M. Pleiotropic effects of the neuropeptides CCAP and myosuppressin in the beetle, Tenebrio molitor L. J Comp Physiol B. 2008, 178, 877-885. 224. Lange, A. B.; Patel, K. The presence and distribution of crustacean cardioactive peptide in the central and peripheral nervous system of the stick insect, Baculum extradentatum. Regul Pept. 2005, 129, 191-201. 225. Donini, A.; Agricola, H.; Lange, A. B. Crustacean cardioactive peptide is a modulator of oviduct contractions in Locusta migratoria. J Insect Physiol. 2001, 47, 277-285. 226. Donini, A.; Lange, A. B. The effects of crustacean cardioactive peptide on locust oviducts are calciumdependent. Peptides. 2002, 23, 683-691. 227. da Silva, R.; Lange, A. B. The association of crustacean cardioactive peptide with the spermatheca of the African migratory locust, Locusta migratoria. J Insect Physiol. 2006, 52, 399-409. 228. Sakai, T.; Satake, H.; Takeda, M. Nutrient-induced alpha-amylase and protease activity is regulated by crustacean cardioactive peptide (CCAP) in the cockroach midgut. Peptides. 2006, 27, 2157-2164. 229. Park, Y.; Kim, Y. J.; Adams, M. E. Identification of G protein-coupled receptors for Drosophila PRXamide peptides, CCAP, corazonin, and AKH supports a theory of ligand-receptor coevolution. Proc Natl Acad Sci U S A. 2002, 99, 11423-11428. 108 230. Belmont, M.; Cazzamali, G.; Williamson, M.; Hauser, F.; Grimmelikhuijzen, C. J. Identification of four evolutionarily related G protein-coupled receptors from the malaria mosquito Anopheles gambiae. Biochem Biophys Res Commun. 2006, 344, 160-165. 231. Veenstra, J. A. Isolation and structure of corazonin, a cardioactive peptide from the American cockroach. FEBS Lett. 1989, 250, 231-234. 232. Veenstra, J. A. Presence of corazonin in three insect species, and isolation and identification of [His7]corazonin from Schistocerca americana. Peptides. 1991, 12, 1285-1289. 233. Verleyen, P.; Baggerman, G.; Mertens, I.; Vandersmissen, T.; Huybrechts, J.; Van Lommel, A.; De Loof, A.; Schoofs, L. Cloning and characterization of a third isoform of corazonin in the honey bee Apis mellifera. Peptides. 2006, 27, 493-499. 234. Predel, R.; Neupert, S.; Russell, W. K.; Scheibner, O.; Nachman, R. J. Corazonin in insects. Peptides. 2007, 28, 3-10. 235. Cantera, R.; Veenstra, J. A.; Nassel, D. R. Postembryonic development of corazonin-containing neurons and neurosecretory cells in the blowfly, Phormia terraenovae. J Comp Neurol. 1994, 350, 559-572. 236. Hansen, I. A.; Sehnal, F.; Meyer, S. R.; Scheller, K. Corazonin gene expression in the waxmoth Galleria mellonella. Insect Mol Biol. 2001, 10, 341-346. 237. Roller, L.; Tanaka, Y.; Tanaka, S. Corazonin and corazonin-like substances in the central nervous system of the Pterygote and Apterygote insects. Cell Tissue Res. 2003, 312, 393-406. 238. Veenstra, J. A.; Davis, N. T. Localization of corazonin in the nervous system of the cockroach Periplaneta americana. Cell Tissue Res. 1993, 274, 57-64. 239. Hamanaka, Y.; Tanaka, S.; Numata, H.; Shiga, S. Peptide immunocytochemistry of neurons projecting to the retrocerebral complex in the blow fly, Protophormia terraenovae. Cell Tissue Res. 2007, 329, 581-593. 240. Predel, R.; Agricola, H. J.; Linde, D.; Wollweber, L.; Veenstra, J. A.; Penzlin, H. The insect neuropeptide corazonin: physiolgoical and immunocytochemical studies in Blattariae. Zoology. 1994, 98, 35-49. 241. Vandersmissen, T.; Hoste, B.; Baggerman, G.; Huybrechts, J.; De Loof, A.; Chaltin, P.; Proost, P.; Breuer, M. Degradation profile of [His7]-corazonin in the hemolymph of the desert locust Schistocerca gregaria. Peptides. 2006, 27, 539-548. 242. Tawfik, A. I.; Tanaka, S.; De Loof, A.; Schoofs, L.; Baggerman, G.; Waelkens, E.; Derua, R.; Milner, Y.; Yerushalmi, Y.; Pener, M. P. Identification of the gregarization-associated dark-pigmentotropin in locusts through an albino mutant. Proc Natl Acad Sci U S A. 1999, 96, 7083-7087. 243. Yerushalmi, Y.; Pener, M. P. The response of a homochrome grasshopper, Oedipoda miniata, to the darkcolour-inducing neurohormone (DCIN) of locusts. J Insect Physiol. 2001, 47, 593-597. 244. Hoste, B.; Simpson, S. J.; Tanaka, S.; Zhu, D.; De Loof, A.; Breuer, M. Effects of [His(7)]-corazonin on the phase state of isolated-reared (solitarious) desert locusts, Schistocerca gregaria. J Insect Physiol. 2002, 48, 981-990. 245. Hoste, B.; Simpson, S. J.; Tanaka, S.; De Loof, A.; Breuer, M. A comparison of phase-related shifts in behavior and morphometrics of an albino strain, deficient in [His(7)]-corazonin, and a normally colored Locusta migratoria strain. J Insect Physiol. 2002, 48, 791-801. 246. Tanaka, S.; Zhu, D. H.; Hoste, B.; Breuer, M. The dark-color inducing neuropeptide, [His(7)]-corazonin, causes a shift in morphometic characteristics towards the gregarious phase in isolated-reared (solitarious) Locusta migratoria. J Insect Physiol. 2002, 48, 1065-1074. 247. Tanaka, Y.; Hua, Y.; Roller, L.; Tanaka, S. Corazonin reduces the spinning rate in the silkworm, Bombyx mori. J Insect Physiol. 2002, 48, 707-714. 248. Yerushalmi, Y.; Bhargava, K.; Gilon, C.; Pener, M. P. Structure-activity relations of the dark-colour-inducing neurohormone of locusts. Insect Biochem Mol Biol. 2002, 32, 909-917. 249. Kim, Y. J.; Spalovska-Valachova, I.; Cho, K. H.; Zitnanova, I.; Park, Y.; Adams, M. E.; Zitnan, D. Corazonin receptor signaling in ecdysis initiation. Proc Natl Acad Sci U S A. 2004, 101, 6704-6709. 250. Porras, M. G.; De Loof, A.; Breuer, M.; Arechiga, H. Corazonin promotes tegumentary pigment migration in the crayfish Procambarus clarkii. Peptides. 2003, 24, 1581-1589. 251. Cazzamali, G.; Saxild, N.; Grimmelikhuijzen, C. Molecular cloning and functional expression of a Drosophila corazonin receptor. Biochem Biophys Res Commun. 2002, 298, 31-36. 252. Janssen, I.; Schoofs, L.; Spittaels, K.; Neven, H.; Vanden Broeck, J.; Devreese, B.; Van Beeumen, J.; Shabanowitz, J.; Hunt, D. F.; De Loof, A. Isolation of NEB-LFamide, a novel myotropic neuropeptide from the grey fleshfly. Mol Cell Endocrinol. 1996, 117, 157-165. 109 253. Verleyen, P.; Huybrechts, J.; Baggerman, G.; Van Lommel, A.; De Loof, A.; Schoofs, L. SIFamide is a highly conserved neuropeptide: a comparative study in different insect species. Biochem Biophys Res Commun. 2004, 320, 334-341. 254. Terhzaz, S.; Rosay, P.; Goodwin, S. F.; Veenstra, J. A. The neuropeptide SIFamide modulates sexual behavior in Drosophila. Biochem Biophys Res Commun. 2007, 352, 305-310. 255. Verleyen, P.; Huybrechts, J.; Schoofs, L. SIFamide illustrates the rapid evolution in Arthropod neuropeptide research. Gen Comp Endocrinol. 2009, 162, 27-35. 256. Christie, A. E.; Stemmler, E. A.; Peguero, B.; Messinger, D. I.; Provencher, H. L.; Scheerlinck, P.; Hsu, Y. W.; Guiney, M. E.; de la Iglesia, H. O.; Dickinson, P. S. Identification, physiological actions, and distribution of VYRKPPFNGSIFamide (Val1)-SIFamide) in the stomatogastric nervous system of the American lobster Homarus americanus. J Comp Neurol. 2006, 496, 406-421. 257. Christie, A. E.; Kutz-Naber, K. K.; Stemmler, E. A.; Klein, A.; Messinger, D. I.; Goiney, C. C.; Conterato, A. J.; Bruns, E. A.; Hsu, Y. W.; Li, L.; Dickinson, P. S. Midgut epithelial endocrine cells are a rich source of the neuropeptides APSGFLGMRamide (Cancer borealis tachykinin-related peptide Ia) and GYRKPPFNGSIFamide (Gly1-SIFamide) in the crabs Cancer borealis, Cancer magister and Cancer productus. J Exp Biol. 2007, 210, 699-714. 258. Soyez, D. Occurrence and diversity of neuropeptides from the crustacean hyperglycemic hormone family in arthropods. A short review. Ann N Y Acad Sci. 1997, 814, 319-323. 259. Lacombe, C.; Greve, P.; Martin, G. Overview on the sub-grouping of the crustacean hyperglycemic hormone family. Neuropeptides. 1999, 33, 71-80. 260. Fanjul-Moles, M. L. Biochemical and functional aspects of crustacean hyperglycemic hormone in decapod crustaceans: review and update. Comp Biochem Physiol C Toxicol Pharmacol. 2006, 142, 390-400. 261. Chan, S. M.; Gu, P. L.; Chu, K. H.; Tobe, S. S. Crustacean neuropeptide genes of the CHH/MIH/GIH family: implications from molecular studies. Gen Comp Endocrinol. 2003, 134, 214-219. 262. Nakatsuji, T.; Lee, C. Y.; Watson, R. D. Crustacean molt-inhibiting hormone: structure, function, and cellular mode of action. Comp Biochem Physiol A Mol Integr Physiol. 2009, 152, 139-148. 263. Webster, S. G.; Keller, R.; Dircksen, H. The CHH-superfamily of multifunctional peptide hormones controlling crustacean metabolism, osmoregulation, moulting, and reproduction. General and Comparative Endocrinology. 2012, 175, 217-233. 264. Chung, J. S.; Zmora, N.; Katayama, H.; Tsutsui, N. Crustacean hyperglycemic hormone (CHH) neuropeptide family: Functions, titer, and binding to target tissues. General and Comparative Endocrinology. 2010, 166, 447-454. 265. Jaros, P. P.; Keller, R. Immunocytochemical identification of hyperglycemic hormone-producing cells in the eyestalk of Carcinus maenas. Cell Tissue Res. 1979, 204, 379-385. 266. Keller, R.; Jaros, P. P.; Kegel, G. Crustacean hyperglycemic neuropeptides. Am. Zool. 1985, 25, 207-221. 267. Chang, E. S.; Chang, S. A.; Beltz, B. S.; Kravitz, E. A. Crustacean hyperglycemic hormone in the lobster nervous system: localization and release from cells in the subesophageal ganglion and thoracic second roots. J Comp Neurol. 1999, 414, 50-56. 268. Escamilla-Chimal, E. G.; Van Herp, F.; Fanjul-Moles, M. L. Daily variations in crustacean hyperglycaemic hormone and serotonin immunoreactivity during the development of crayfish. J Exp Biol. 2001, 204, 10731081. 269. Chung, J. S.; Dircksen, H.; Webster, S. G. A remarkable, precisely timed release of hyperglycemic hormone from endocrine cells in the gut is associated with ecdysis in the crab Carcinus maenas. Proc Natl Acad Sci U S A. 1999, 96, 13103-13107. 270. Chung, J. S.; Webster, S. G. Expression and release patterns of neuropeptides during embryonic development and hatching of the green shore crab, Carcinus maenas. Development. 2004, 131, 4751-4761. 271. Kummer, G.; Keller, R. High-affinity binding of crustacean hyperglycemic hormone (CHH) to hepatopancreatic plasma membranes of the crab Carcinus maenas and the crayfish Orconectes limosus. Peptides. 1993, 14, 103-108. 272. Santos, E. A.; Keller, R.; Rodriguez, E.; Lopez, L. Effects of serotonin and fluoxetine on blood glucose regulation in two decapod species. Braz J Med Biol Res. 2001, 34, 75-80. 273. Morris, S. Neuroendocrine regulation of osmoregulation and the evolution of air-breathing in decapod crustaceans. J Exp Biol. 2001, 204, 979-989. 274. Liu, L.; Laufer, H.; Wang, Y.; Hayes, T. A neurohormone regulating both methyl farnesoate synthesis and glucose metabolism in a crustacean. Biochem Biophys Res Commun. 1997, 237, 694-701. 110 275. Avarre, J. C.; Khayat, M.; Michelis, R.; Nagasawa, H.; Tietz, A.; Lubzens, E. Inhibition of de Novo Synthesis of a Jelly Layer Precursor Protein by Crustacean Hyperglycemic Hormone Family Peptides and Posttranscriptional Regulation by Sinus Gland Extracts in Penaeus semisulcatus Ovaries. General and Comparative Endocrinology. 2001, 124, 257-268. 276. Khayat, M.; Yang, W. J.; Aida, K.; Nagasawa, H.; Tietz, A.; Funkenstein, B.; Lubzens, E. Hyperglycaemic Hormones Inhibit Protein and mRNA Synthesis inin Vitro-Incubated Ovarian Fragments of the Marine ShrimpPenaeus semisulcatus. General and Comparative Endocrinology. 1998, 110, 307-318. 277. Khalaila, I.; Manor, R.; Weil, S.; Granot, Y.; Keller, R.; Sagi, A. The eyestalk–androgenic gland–testis endocrine axis in the crayfish Cherax quadricarinatus. General and Comparative Endocrinology. 2002, 127, 147-156. 278. Chung, J. S.; Zmora, N. Functional studies of crustacean hyperglycemic hormones (CHHs) of the blue crab, Callinectes sapidus – the expression and release of CHH in eyestalk and pericardial organ in response to environmental stress. FEBS Journal. 2008, 275, 693-704. 279. Lorenzon, S.; Giulianini, P. G.; Ferrero, E. A. Lipopolysaccharide-Induced Hyperglycemia Is Mediated by CHH Release in Crustaceans. General and Comparative Endocrinology. 1997, 108, 395-405. 280. Lorenzon, S.; Edomi, P.; Giulianini, P. G.; Mettulio, R.; Ferrero, E. A. Variation of crustacean hyperglycemic hormone (cHH) level in the eyestalk and haemolymph of the shrimp Palaemon elegans following stress. Journal of Experimental Biology. 2004, 207, 4205-4213. 281. Stentiford, G. D.; Chang, E. S.; Chang, S. A.; Neil, D. M. Carbohydrate Dynamics and the Crustacean Hyperglycemic Hormone (CHH): Effects of Parasitic Infection in Norway Lobsters (Nephrops norvegicus). General and Comparative Endocrinology. 2001, 121, 13-22. 282. Stentiford, G. D.; Neil, D. M.; Coombs, G. H. Alterations in the biochemistry and ultrastructure of the deep abdominal flexor muscle of the Norway lobster Nephrops norvegicus during infection by a parasitic dinoflagellate of the genus Hematodinium. Diseases of Aquatic Organisms. 2000, 42, 133-141. 283. Stentiford, G. D.; Green, M.; Bateman, K.; Small, H. J.; Neil, D. M.; Feist, S. W. Infection by a Hematodiniumlike parasitic dinoflagellate causes Pink Crab Disease (PCD) in the edible crab Cancer pagurus. Journal of Invertebrate Pathology. 2002, 79, 179-191. 284. Soyez, D.; Van Deijnen, J. E.; Martin, M. Isolation and characterization of a vitellogenesis-inhibiting factor from the sinus gland of the lobster, Homarus americanus. J Exp Zool. 1987, 244, 479-484. 285. Edomi, P.; Azzoni, E.; Mettulio, R.; Pandolfelli, N.; Ferrero, E. A.; Giulianini, P. G. Gonad-inhibiting hormone of the Norway lobster (Nephrops norvegicus): cDNA cloning, expression, recombinant protein production, and immunolocalization. Gene. 2002, 284, 93-102. 286. Treerattrakool, S.; Panyim, S.; Chan, S. M.; Withyachumnarnkul, B.; Udomkit, A. Molecular characterization of gonad-inhibiting hormone of Penaeus monodon and elucidation of its inhibitory role in vitellogenin expression by RNA interference. FEBS J. 2008, 275, 970-980. 287. Charmantier, G.; Charmantier-Daures, M.; Van Herp, F. Hormonal regulation of growth and reproduction in crustaceans. Endocrinology and Reproduction, Rcent Advances in Marine Biotechnology. 1997, Vol. 1. 288. Wainwright, G.; Webster, S. G.; Wilkinson, M. C.; Chung, J. S.; Rees, H. H. Structure and significance of mandibular organ-inhibiting hormone in the crab, Cancer pagurus. Involvement in multihormonal regulation of growth and reproduction. J Biol Chem. 1996, 271, 12749-12754. 289. Liu, L.; Laufer, H.; Gogarten, P. J.; Wang, M. cDNA cloning of a mandibular organ inhibiting hormone from the spider crab Libinia emarginata. Invert Neurosci. 1997, 3, 199-204. 290. Borst, D. W.; Wainwright, G.; Rees, H. H. In vivo regulation of the mandibular organ in the edible crab, Cancer pagurus. Proc Biol Sci. 2002, 269, 483-490. 291. Josefsson, L. Invertebrate neuropeptide hormones. Int J Pept Protein Res. 1983, 21, 459-470. 292. Rao, K. R. Crustacean pigment-dispersing hormones: chemistry, distribution and actions. Pigment Cell Res. 1992, Suppl 2, 266-270. 293. Nassel, D. R.; Shiga, S.; Mohrherr, C. J.; Rao, K. R. Pigment-dispersing hormone-like peptide in the nervous system of the flies Phormia and Drosophila: immunocytochemistry and partial characterization. J Comp Neurol. 1993, 331, 183-198. 294. Meelkop, E.; Temmerman, L.; Schoofs, L.; Janssen, T. Signalling through pigment dispersing hormone-like peptides in invertebrates. Progress in Neurobiology. 2011, 93, 125-147. 295. Fernlund, P. Structure of a light-adapting hormone from the shrimp, Pandalus borealis. Biochim Biophys Acta. 1976, 439, 17-25. 111 296. Rao, K. R.; Riehm, J. P.; Zahnow, C. A.; Kleinholz, L. H.; Tarr, G. E.; Johnson, L.; Norton, S.; Landau, M.; Semmes, O. J.; Sattelberg, R. M.; Jorenby, W. H.; Hintz, M. F. Characterization of a pigment-dispersing hormone in eyestalks of the fiddler crab Uca pugilator. Proc Natl Acad Sci U S A. 1985, 82, 5319-5322. 297. Kleinholz, L. H.; Rao, K. R.; Riehm, J. P.; Tarr, G. E.; Johnson, L.; Norton, S. Isolation and Sequence Analysis of a Pigment-Dispersing Hormone from Eyestalks of the Crab, Cancer magister. Biological Bulletin. 1986, 170, 135-143. 298. Strauss, J.; Dircksen, H. Circadian clocks in crustaceans: identified neuronal and cellular systems. Front Biosci. 2010, 15, 1040-1074. 299. Meelkop, E.; Marco, H. G.; Janssen, T.; Temmerman, L.; Vanhove, M. P.; Schoofs, L. A structural and functional comparison of nematode and crustacean PDH-like sequences. Peptides. 2012, 34, 74-81. 300. Rao, K. R. Crustacean Pigmentary-Effector Hormones: Chemistry and Functions of RPCH, PDH, and Related Peptides. Am Zool. 2001, 41, 364-379. 301. Hernández-Falcón, J.; Moreno-Sáenz, E.; Farías, J.; Fuentes-Pardo, B. Role of the sinus gland in crayfish circadian rhythmicity—I. pseudopupil circadian rhythm. Comparative Biochemistry and Physiology Part A: Physiology. 1987, 87, 111-118. 302. Sullivan, J. M.; Genco, M. C.; Marlow, E. D.; Benton, J. L.; Beltz, B. S.; Sandeman, D. C. Brain photoreceptor pathways contributing to circadian rhythmicity in crayfish. Chronobiol Int. 2009, 26, 1136-1168. 303. Solís-Chagoyán, H.; Alvarado, R.; Figueroa, A.; Mendoza-Vargas, L.; Fuentes-Pardo, B. Pigment dispersing hormone modulates spontaneous electrical activity of the cerebroid ganglion and synchronizes electroretinogram circadian rhythm in crayfish Procambarus clarkii. Comparative Biochemistry and Physiology - Part A: Molecular & Integrative Physiology. 2012, 161, 450-455. 304. Nery, L. E.; Castrucci, A. M. L. Crustacean Chromatophore: Endocrine Regulation and Signaling Systems. In: Weise K, editor. The Crustacean Nervous System. Berlin, Germany: Springer-Verlag; 2003. p. 98-112. 305. Mertens, I.; Vandingenen, A.; Johnson, E. C.; Shafer, O. T.; Li, W.; Trigg, J. S.; De Loof, A.; Schoofs, L.; Taghert, P. H. PDF Receptor Signaling in Drosophila Contributes to Both Circadian and Geotactic Behaviors. Neuron. 2005, 48, 213-219. 306. Duvall, L. B.; Taghert, P. H. The Circadian Neuropeptide PDF Signals Preferentially through a Specific Adenylate Cyclase Isoform AC3 in M Pacemakers of Drosophila. PLoS Biol. 2012, 10, e1001337. 307. Josefsson, L. Separation and purification of distal retinal pigment hormone and red pigment concentrating hormone of the crustacean eyestalk. Biochimica et Biophysica Acta (BBA) - General Subjects. 1967, 148, 300-303. 308. Fernlund, P.; Josefsson, L. Crustacean color-change hormone: amino acid sequence and chemical synthesis. Science. 1972, 177, 173-175. 309. Gäde, G. Peptides of the Adipokinetic Hormone/Red Pigment-Concentrating Hormone Family. Annals of the New York Academy of Sciences. 2009, 1163, 125-136. 310. Cebada, J.; Alvarado-Alvarez, R.; Becerra, E.; Neri-Bazan, L.; Rocha, L.; Garcia, U. An improved method for long-term measuring of hemolymph fluctuations of non-essential amino acids, GABA and histamine from freely moving crayfish. J Neurosci Meth. 2006, 153, 1-7. 311. De Kleijn, D. P.; Van Herp, F. Molecular biology of neurohormone precursors in the eyestalk of Crustacea. Comp Biochem Physiol B Biochem Mol Biol. 1995, 112, 573-579. 312. Gruhler, A.; Schulze, W. X.; Matthiesen, R.; Mann, M.; Jensen, O. N. Stable Isotope Labeling of Arabidopsis thaliana Cells and Quantitative Proteomics by Mass Spectrometry. Molecular & Cellular Proteomics. 2005, 4, 1697-1709. 313. Gouw, J. W.; Tops, B. B.; Krijgsveld, J. Metabolic labeling of model organisms using heavy nitrogen (15N). Methods Mol Biol. 2011, 753, 29-42. 314. Christie, A. E.; Durkin, C. S.; Hartline, N.; Ohno, P.; Lenz, P. H. Bioinformatic analyses of the publicly accessible crustacean expressed sequence tags (ESTs) reveal numerous novel neuropeptide-encoding precursor proteins, including ones from members of several little studied taxa. General and Comparative Endocrinology. 2010, 167, 164-178. 315. Preciado, M.; Tsutsumi, V.; Arechiga, H. Ultrastructural features of neurosecretory cells in the medulla externa of the crayfish eyestalk. Gen Comp Endocrinol. 1994, 95, 432-442. 316. Sury, M. D.; Chen, J.-X.; Selbach, M. The SILAC Fly Allows for Accurate Protein Quantification in Vivo. Molecular & Cellular Proteomics. 2010, 9, 2173-2183. 317. Stemmler, E. A.; Gardner, N. P.; Guiney, M. E.; Bruns, E. A.; Dickinson, P. S. The detection of red pigmentconcentrating hormone (RPCH) in crustacean eyestalk tissues using matrix-assisted laser 112 desorption/ionization-Fourier transform mass spectrometry: [M + Na]+ ion formation in dried droplet tissue preparations. J Mass Spectrom. 2006, 41, 295-311. 318. Fingerman, M.; Couch, E. F. The red pigment-dispersing hormone of the abdominal nerve cord and its contribution to the chromatic physiology of the prawn, Palaemonetes vulgaris. Rev Can Biol. 1967, 26, 109-117. 319. Christie, A. E.; Skiebe, P.; Marder, E. Matrix of neuromodulators in neurosecretory structures of the crab Cancer borealis. J Exp Biol. 1995, 198, 2431-2439. 320. Yang, W. J.; Rao, K. R. Cloning of precursors for two MIH/VIH-related peptides in the prawn, Macrobrachium rosenbergii. Biochem Biophys Res Commun. 2001, 289, 407-413. 321. Fingerman, M. Persistent Daily and Tidal Rhythms of Color Change in Callinectes sapidus. The Biological Bulletin. 1955, 109, 255-264. 322. Porras, M. G.; Lopez-Colome, A. M.; Arechiga, H. Red pigment-concentrating hormone induces a calciummediated retraction of distal retinal pigments in the crayfish. J Comp Physiol A. 2001, 187, 349-357. 323. Garfias, A.; Rodr; Iacute; Guez-Sosa, L.; Ar; Eacute; Chiga, H. Modulation of crayfish retinal function by red pigment concentrating hormone. J Exp Biol. 1995, 198, 1447-1454. 324. Rodriguez-Sosa, L.; de la Vega, M. T.; Vergara, P.; Arechiga, H. Seasonal rhythm of red pigment concentrating hormone in the crayfish. Chronobiol Int. 1997, 14, 639-645. 325. Zralá, J.; Kodrík, D.; Zahradníčková, H.; Zemek, R.; Socha, R. A novel function of red pigment-concentrating hormone in crustaceans: Porcellio scaber (Isopoda) as a model species. General and Comparative Endocrinology. 2010, 166, 330-336. 326. Dickinson, P. S.; Fairfield, W. P.; Hetling, J. R.; Hauptman, J. Neurotransmitter interactions in the stomatogastric system of the spiny lobster: one peptide alters the response of a central pattern generator to a second peptide. J Neurophysiol. 1997, 77, 599-610. 327. Dickinson, P. S.; Hauptman, J.; Hetling, J.; Mahadevan, A. RCPH modulation of a multi-oscillator network: effects on the pyloric network of the spiny lobster. J Neurophysiol. 2001, 85, 1424-1435. 328. Dickinson, P. S.; Marder, E. Peptidergic modulation of a multioscillator system in the lobster. I. Activation of the cardiac sac motor pattern by the neuropeptides proctolin and red pigment-concentrating hormone. J Neurophysiol. 1989, 61, 833-844. 329. Dickinson, P. S.; Mecsas, C.; Hetling, J.; Terio, K. The neuropeptide red pigment concentrating hormone affects rhythmic pattern generation at multiple sites. J Neurophysiol. 1993, 69, 1475-1483. 330. Dickinson, P. S.; Stemmler, E. A.; Christie, A. E. The pyloric neural circuit of the herbivorous crab Pugettia producta shows limited sensitivity to several neuromodulators that elicit robust effects in more opportunistically feeding decapods. J Exp Biol. 2008, 211, 1434-1447. 331. Swensen, A. M.; Marder, E. Multiple peptides converge to activate the same voltage-dependent current in a central pattern-generating circuit. J Neurosci. 2000, 20, 6752-6759. 332. Swensen, A. M.; Marder, E. Modulators with convergent cellular actions elicit distinct circuit outputs. J Neurosci. 2001, 21, 4050-4058. 333. Thirumalai, V.; Prinz, A. A.; Johnson, C. D.; Marder, E. Red pigment concentrating hormone strongly enhances the strength of the feedback to the pyloric rhythm oscillator but has little effect on pyloric rhythm period. J Neurophysiol. 2006, 95, 1762-1770. 334. Sherff, C. M.; Mulloney, B. Red pigment concentrating hormone is a modulator of the crayfish swimmeret system. J Exp Biol. 1991, 155, 21-35. 335. Jorge-Rivera, J. C.; Sen, K.; Birmingham, J. T.; Abbott, L. F.; Marder, E. Temporal dynamics of convergent modulation at a crustacean neuromuscular junction. J Neurophysiol. 1998, 80, 2559-2570. 336. Skiebe, P. Neuropeptides are ubiquitous chemical mediators: Using the stomatogastric nervous system as a model system. Journal of Experimental Biology. 2001, 204, 2035-2048. 337. Benson, D. A.; Karsch-Mizrachi, I.; Lipman, D. J.; Ostell, J.; Wheeler, D. L. GenBank. Nucleic Acids Research. 2005, 33, D34-D38. 338. Red-pigment concentrating hormone receptor mRNA, partial cds. [database on the Internet]. GenBank. 2008 [cited 05 July 2012]. 339. Hansen, K. K.; Hauser, F.; Cazzamali, G.; Williamson, M.; Grimmelikhuijzen, C. J. Cloning and characterization of the adipokinetic hormone receptor from the cockroach Periplaneta americana. Biochem Biophys Res Commun. 2006, 343, 638-643. 340. Li, B.; Predel, R.; Neupert, S.; Hauser, F.; Tanaka, Y.; Cazzamali, G.; Williamson, M.; Arakane, Y.; Verleyen, P.; Schoofs, L.; Schachtner, J.; Grimmelikhuijzen, C. J.; Park, Y. Genomics, transcriptomics, and 113 peptidomics of neuropeptides and protein hormones in the red flour beetle Tribolium castaneum. Genome Res. 2008, 18, 113-122. 341. Staubli, F.; Jorgensen, T. J.; Cazzamali, G.; Williamson, M.; Lenz, C.; Sondergaard, L.; Roepstorff, P.; Grimmelikhuijzen, C. J. Molecular identification of the insect adipokinetic hormone receptors. Proc Natl Acad Sci U S A. 2002, 99, 3446-3451. 342. Milograna, S. R.; Bell, F. T.; McNamara, J. C. Signal transduction, plasma membrane calcium movements, and pigment translocation in freshwater shrimp chromatophores. Journal of Experimental Zoology Part A: Ecological Genetics and Physiology. 2010, 313A, 605-617. 343. Gäde, G.; Auerswald, L. Mode of action of neuropeptides from the adipokinetic hormone family. General and Comparative Endocrinology. 2003, 132, 10-20. 344. Larance, M.; Bailly, A. P.; Pourkarimi, E.; Hay, R. T.; Buchanan, G.; Coulthurst, S.; Xirodimas, D. P.; Gartner, A.; Lamond, A. I. Stable-isotope labeling with amino acids in nematodes. Nat Meth. 2011, 8, 849-851. 345. Li, L. J.; Sweedler, J. V. Peptides in the Brain: Mass Spectrometry-Based Measurement Approaches and Challenges. Annu Rev Anal Chem. 2008, 1, 451-483. 346. Morimoto, R.; Satoh, F.; Murakami, O.; Totsune, K.; Saruta, M.; Suzuki, T.; Sasano, H.; Ito, S.; Takahashi, K. Expression of peptide YY in human brain and pituitary tissues. Nutrition. 2008, 24, 878-884. 347. Ma, M.; Gard, A. L.; Xiang, F.; Wang, J.; Davoodian, N.; Lenz, P. H.; Malecha, S. R.; Christie, A. E.; Li, L. Combining in silico transcriptome mining and biological mass spectrometry for neuropeptide discovery in the Pacific white shrimp Litopenaeus vannamei. Peptides. 2010, 31, 27-43. 348. Hui, L.; Xiang, F.; Zhang, Y.; Li, L. Mass spectrometric elucidation of the neuropeptidome of a crustacean neuroendocrine organ. Peptides. 2012, 36, 230-239. 349. Saideman, S. R.; Blitz, D. M.; Nusbaum, M. P. Convergent Motor Patterns from Divergent Circuits. The Journal of Neuroscience. 2007, 27, 6664-6674. 350. Chen, R.; Hui, L.; Cape, S. S.; Wang, J.; Li, L. Comparative Neuropeptidomic Analysis of Food Intake via a Multi-faceted Mass Spectrometric Approach. ACS Chem Neurosci. 2010, 1, 204-214. 351. Brown, B. E. Neuromuscular Transmitter Substance in Insect Visceral Muscle. Science. 1967, 155, 595-597. 352. Perry, M.; Li, Q.; Kennedy, R. T. Review of recent advances in analytical techniques for the determination of neurotransmitters. Anal Chim Acta. 2009, 653, 1-22. 353. O'Shea, M.; Adams, M. Proctolin: From “Gut Factor” to Model Neuropeptide. In: Evans PD, Wigglesworth VB, editors. Adv. Insect Physiol.: Academic Press; 1987. p. 1-28. 354. Skiebe, P. Neuropeptides in the crayfish stomatogastric nervous system. Microsc Res Tech. 2003, 60, 302-312. 355. Konopińska, D.; Rosiński, G. Proctolin, an insect neuropeptide. Journal of Peptide Science. 1999, 5, 533-546. 356. Mykles, D. L.; Adams, M. E.; Gade, G.; Lange, A. B.; Marco, H. G.; Orchard, I. Neuropeptide action in insects and crustaceans. Physiol Biochem Zool. 2010, 83, 836-846. 357. Kravitz, E. A.; Glusman, S.; Harris-Warrick, R. M.; Livingstone, M. S.; Schwarz, T.; Goy, M. F. Amines and a peptide as neurohormones in lobsters: actions on neuromuscular preparations and preliminary behavioural studies. J Exp Biol. 1980, 89, 159-175. 358. Schwarz, T. L.; Harris-Warrick, R. M.; Glusman, S.; Kravitz, E. A. A peptide action in a lobster neuromuscular preparation. J Neurobiol. 1980, 11, 623-628. 359. Miller, M. W.; Sullivan, R. E. Some effects of proctolin on the cardiac ganglion of the Maine Lobster, Homarus americanus (Milne Edwards). J Neurobiol. 1981, 12, 629-639. 360. Bishop, C. A.; Wine, J. J.; O'Shea, M. Neuropeptide proctolin in postural motoneurons of the crayfish. J Neurosci. 1984, 4, 2001-2009. 361. Schwarz, T. L.; Lee, G. M.; Siwicki, K. K.; Standaert, D. G.; Kravitz, E. A. Proctolin in the lobster: the distribution, release, and chemical characterization of a likely neurohormone. J Neurosci. 1984, 4, 13001311. 362. Stangier, J.; Dircksen, H.; Keller, R. Identification and immunocytochemical localization of proctolin in pericardial organs of the shore crab, Carcinus maenas. Peptides. 1986, 7, 67-72. 363. Zhao, S.; Sheibanie, A. F.; Oh, M.; Rabbah, P.; Nadim, F. Peptide Neuromodulation of Synaptic Dynamics in an Oscillatory Network. The Journal of Neuroscience. 2011, 31, 13991-14004. 364. Hui, L.; Cunningham, R.; Zhang, Z.; Cao, W.; Jia, C.; Li, L. Discovery and characterization of the Crustacean hyperglycemic hormone precursor related peptides (CPRP) and orcokinin neuropeptides in the sinus glands of the blue crab Callinectes sapidus using multiple tandem mass spectrometry techniques. J Proteome Res. 2011, 10, 4219-4229. 365. Isaac, R. E.; Taylor, C. A.; Hamasaka, Y.; Nassel, D. R.; Shirras, A. D. Proctolin in the post-genomic era: new insights and challenges. Invert Neurosci. 2004, 5, 51-64. 114 366. Wood, D. E.; Nishikawa, M.; Derby, C. D. Proctolinlike immunoreactivity and identified neurosecretory cells as putative substrates for modulation of courtship display behavior in the blue crab, Callinectes sapidus. J Comp Neurol. 1996, 368, 153-163. 367. Marder, E.; Hooper, S. L.; Siwicki, K. K. Modulatory action and distribution of the neuropeptide proctolin in the crustacean stomatogastric nervous system. J Comp Neurol. 1986, 243, 454-467. 368. Siwicki, K. K.; Bishop, C. A. Mapping of proctolinlike immunoreactivity in the nervous systems of lobster and crayfish. J Comp Neurol. 1986, 243, 435-453. 369. Skiebe, P.; Ganeshina, O. Synaptic neuropil in nerves of the crustacean stomatogastric nervous system: an immunocytochemical and electron microscopical study. J Comp Neurol. 2000, 420, 373-397. 370. Kobierski, L. A.; Beltz, B. S.; Trimmer, B. A.; Kravitz, E. A. FMRFamidelike peptides of Homarus americanus: distribution, immunocytochemical mapping, and ultrastructural localization in terminal varicosities. J Comp Neurol. 1987, 266, 1-15. 371. Siwicki, K. K.; Beltz, B. S.; Schwarz, T. L.; Kravitz, E. A. Proctolin in the lobster nervous system. Peptides. 1985, 6 Suppl 3, 393-402. 372. Dircksen, H. Fine structure of the neurohemal sinus gland of the shore crab, Carcinus maenas, and immunoelectron-microscopic identification of neurosecretory endings according to their neuropeptide contents. Cell Tissue Res. 1992, 269, 249-266. 373. Siwicki, K. K.; Beltz, B. S.; Kravitz, E. A. Proctolin in identified serotonergic, dopaminergic, and cholinergic neurons in the lobster, Homarus americanus. J Neurosci. 1987, 7, 522-532. 374. Nusbaum, M. P.; Marder, E. A modulatory proctolin-containing neuron (MPN). I. Identification and characterization. J Neurosci. 1989, 9, 1591-1599. 375. Christie, A. E.; Baldwin, D. H.; Marder, E.; Graubard, K. Organization of the stomatogastric neuropil of the crab, Cancer borealis, as revealed by modulator immunocytochemistry. Cell Tissue Res. 1997, 288, 135148. 376. Fenelon, V. S.; Casasnovas, B.; Faumont, S.; Meyrand, P. Ontogenetic alteration in peptidergic expression within a stable neuronal population in lobster stomatogastric nervous system. J Comp Neurol. 1998, 399, 289-305. 377. Utting, M.; Agricola, H.; Sandeman, R.; Sandeman, D. Central complex in the brain of crayfish and its possible homology with that of insects. J Comp Neurol. 2000, 416, 245-261. 378. Beltz, B. S.; Kravitz, E. A. Physiological identification, morphological analysis, and development of identified serotonin-proctolin containing neurons in the lobster ventral nerve cord. J Neurosci. 1987, 7, 533-546. 379. Hooper, S. L.; Marder, E. Modulation of a central pattern generator by two neuropeptides, proctolin and FMRFamide. Brain Res. 1984, 305, 186-191. 380. Hooper, S. L.; DiCaprio, R. A. Crustacean Motor Pattern Generator Networks. Neurosignals. 2004, 13, 50-69. 381. Pasztor, V. M.; Bush, B. M. Peripheral modulation of mechano-sensitivity in primary afferent neurons. Nature. 1987, 326, 793-795. 382. Pasztor, V. M.; Lange, A. B.; Orchard, I. Stretch-induced release of proctolin from the dendrites of a lobster sense organ. Brain Res. 1988, 458, 199-203. 383. Pasztor, V. M.; Bush, B. M. Primary afferent responses of a crustacean mechanoreceptor are modulated by proctolin, octopamine, and serotonin. J Neurobiol. 1989, 20, 234-254. 384. el Manira, A.; Rossi-Durand, C.; Clarac, F. Serotonin and proctolin modulate the response of a stretch receptor in crayfish. Brain Res. 1991, 541, 157-162. 385. Combes, D.; Simmers, J.; Moulins, M. Conditional dendritic oscillators in a lobster mechanoreceptor neurone. J Physiol. 1997, 499 ( Pt 1), 161-177. 386. Stein, W.; DeLong, N. D.; Wood, D. E.; Nusbaum, M. P. Divergent co-transmitter actions underlie motor pattern activation by a modulatory projection neuron. Eur J Neurosci. 2007, 26, 1148-1165. 387. Acevedo, L. D.; Hall, W. M.; Mulloney, B. Proctolin and excitation of the crayfish swimmeret system. J Comp Neurol. 1994, 345, 612-627. 388. Braun, G.; Mulloney, B. Cholinergic modulation of the swimmeret motor system in crayfish. J Neurophysiol. 1993, 70, 2391-2398. 389. Mulloney, B.; Acevedo, L. D.; Bradbury, A. G. Modulation of the crayfish swimmeret rhythm by octopamine and the neuropeptide proctolin. J Neurophysiol. 1987, 58, 584-597. 390. Murchison, D.; Chrachri, A.; Mulloney, B. A separate local pattern-generating circuit controls the movements of each swimmeret in crayfish. J Neurophysiol. 1993, 70, 2620-2631. 391. Wood, D. E. Neuromodulation of rhythmic motor patterns in the blue crab Callinectes sapidus by amines and the peptide proctolin. J Comp Physiol A. 1995, 177, 335-349. 115 392. Mercier, A. J.; Lee, J. Differential effects of neuropeptides on circular and longitudinal muscles of the crayfish hindgut. Peptides. 2002, 23, 1751-1757. 393. Wilkens, J. L.; Cavey, M. J.; Shovkivska, I.; Zhang, M. L.; Keurs, H. E. D. J. t. Elasticity, unexpected contractility and the identification of actin and myosin in lobster arteries. Journal of Experimental Biology. 2008, 211, 766-772. 394. Rathmayer, W.; Djokaj, S.; Gaydukov, A.; Kreissl, S. The neuromuscular junctions of the slow and the fast excitatory axon in the closer of the crab Eriphia spinifrons are endowed with different Ca2+ channel types and allow neuron-specific modulation of transmitter release by two neuropeptides. J Neurosci. 2002, 22, 708-717. 395. Erxleben, C. F.; deSantis, A.; Rathmayer, W. Effects of proctolin on contractions, membrane resistance, and non-voltage-dependent sarcolemmal ion channels in crustacean muscle fibers. J Neurosci. 1995, 15, 43564369. 396. Bishop, C.; Krouse, M.; Wine, J. Peptide cotransmitter potentiates calcium channel activity in crayfish skeletal muscle. The Journal of Neuroscience. 1991, 11, 269-276. 397. Bishop, C.; Krouse, M.; Wine, J. Peptide potentiation of calcium channel activity can be seasonally variable. Journal of Experimental Biology. 1991, 156, 607-610. 398. Bishop, C. A.; Krouse, M. E.; Wine, J. J. Peptide cotransmitter potentiates calcium channel activity in crayfish skeletal muscle. J Neurosci. 1991, 11, 269-276. 399. Philipp, B.; Rogalla, N.; Kreissl, S. The neuropeptide proctolin potentiates contractions and reduces cGMP concentration via a PKC-dependent pathway. J Exp Biol. 2006, 209, 531-540. 400. Brustle, B.; Kreissl, S.; Mykles, D. L.; Rathmayer, W. The neuropeptide proctolin induces phosphorylation of a 30 kDa protein associated with the thin filament in crustacean muscle. J Exp Biol. 2001, 204, 2627-2635. 2.10. Figures 116 Figure 2.1. Neuropeptide synthesis, processing and storage. Neuropeptides are synthesized in the cell body by the rough endoplasmic reticulum (RER) and packaged into vesicles as prepropeptides. These are transported down the axon to the terminal, where they are stored until released. During this transport and storage period, the prepropeptides are subjected to a number of processing steps, including cleavage and various types of post-translational modification. In crustacean NPs, conversion of a C-terminal glycine residue into a C-terminal amidation is a common modification that is illustrated here. After release from the axon terminal, NPs diffuse across the synapse to the dendrite, where they come into contact with their receptors. NP degradation occurs following receptor internalization (not shown), or after NPs diffuse to degradation enzymes, which are present in the synapse and in the extracellular space. 117 Figure 2.2. Overview of MS-based methods for neuropeptide discovery in invertebrates. Illustrates extraction of tissue, direct tissue profiling, and mass spectrometry imaging (MSI), and how these methods fit into a workflow. Tissue from an animal can be extracted and subjected to cleanup and analytical separations techniques prior to MS and/or MS/MS analysis to determine NP identity. Direct tissue analysis can quickly give the NP profile of a tissue of interest. MSI can determine the complex distribution of a NP throughout a tissue of interest. The brain of a decapod crustacean is used in the figure for illustration. 118 Figure 2.3. Workflow for sample preparation options for analysis of NPs in liquid samples. Liquid NP samples can be obtained via extraction and protein precipitation of dissected tissue or hemolymph, or from microdialysis. These samples can then be subjected to any number of solution-phase sample preparation techniques, including immunoprecipitation, desalting, and chemical modification. One useful type of chemical modification that can be done at this step is attachment of isotope-coded tags to peptides from different time points or different conditions for quantitative MS analysis. These reduced complexity and/or modified samples can then be subjected to analytical separation techniques, two of which are illustrated here. These samples can then be subjected to MS and/or MS/MS. As indicated by the arrows, some steps can be skipped or repeated depending on the sample and desired analysis. 119 Figure 2.4. Physiological recording techniques to determine NP function in the crustacean stomatogastric nervous system (STNS). One method to determine NP function is to apply NPs to an isolated STNS preparation, which is kept alive in a dish and subjected to extracellular and/or intracellular recordings using sharp electrodes. a) Illustrates the isolated STNS and sites where electrical recordings can be made. b) Changes are observed in the stereotypical firing pattern of the LP neuron and from the lvn nerve when a NP, CbAST-C2 is bath applied to the preparation. Adapted from [64] with permission. Copyright 2009, Elsevier. 120 2.11. Table Table 2.1: Detection of NPs in Hemolymph with Different Sample Preparation Methods Sample Preparation Method Peptide Theoretical In vivo In vitro Hemolymph Peptide sequence Classification (M+H) MD MD extraction A-type YAFGLa 569.31 x x allatostatin YSFGLa 585.3 x x SPYAFGLa 753.39 x DPYAFGLa 781.39 x PDMYAFGLa 912.43 x pERPYSFGLa 949.49 x ERPYSGLa 967.5 x B-type NWNKFQGSWa 1165.55 x allatostatin TSWFKFQGSWa 1182.57 x NNNWSKFQGSWa 1366.63 x Allatostatin DPYAFGLGKRPADL 1519.79 x combination DPYAFGLGKRPADL 2128.09 x YEFGLa TachykininAPSGFQa 605.3 x related GFLGMRa 679.37 x APSGFLGMRa 934.49 x x APSGFLGM(O)Ra 950.49 x TPSGFLGMRa 964.5 x Orcokinin NFDEIDRSGFG 1256.55 x x NFDEIDRSGFGF 1403.62 x NFDEIDRSGFGFV 1502.69 x NFDEIDRSGFGFN 1517.67 x NFDEIDRSSFGFV 1532.7 x NFDEIDRSGFGFH 1540.68 x RFamide RSFLRFa 824.49 x RNFLRFa 851.5 x GGRNFLRFa 965.54 x DRNFLRFa 966.53 x SGRNFLRFa 995.55 x GPRNFLRFa 1005.57 x APRNFLRFa 1019.59 x x SDRNFLRFa 1053.56 x TNRNFLRFa 1066.59 x 121 RYamide Other NP Cryptocyanin fragment b-Actin fragment DGGRNFLRFa GAHKNYLRFa SMPSLRLRFa SENRNFLRFa FVGGSRYa SGFYANRYa pEGFYSQRYa pQLNFSPGWa (RPCH) PFCNAFTGCa (CCAP) TNFAFSPRLa (CabPK-I) I/LNFTHKFa 1080.57 1104.61 1105.63 1181.62 784.41 976.46 1030.51 x x 930.45 x 956.38 x 1051.57 x YKIFEPL 909.51 YKIFEPLR YKIFEPLRE YKIFEPLRES KIFEPLREDNL YKIFEPLRESN YKIFEPLRESNL 1065.61 1194.65 1281.68 1373.74 1395.73 1508.81 LRVAPEESPVL 1209.68 x x 905.5 x x x x x x x x x x x x x x x x x x Total NPs 35 8 11 Total protein fragments 2 0 6 Table 2.1 was created from data compiled from references [67-69]. An x indicates that the compound was detected with that sample preparation method, a blank cell indicates that it was not. In vivo MD indicates experiments where a microdialysis probe was implanted into a live crab and sample was collected from this probe for analysis preceded by sample preparation steps. In vitro MD experiments entailed immersing a MD probe into a beaker of hemolymph freshly collected from a crab, kept on ice, and constantly stirred. The liquid that flowed out of this probe was prepared and analyzed. Hemolymph extraction was conducted by adding the hemolymph immediately to an acidified methanol solution with or without protease inhibitors added, 122 followed by sample preparation and analysis. Analysis was conducted by capLC-ESI-QTOF, nanoLC-ESI-QTOF, MALDI-FTICR, and/or MALDI-TOFTOF. 123 Chapter 3: Methods for Function-Driven Discovery and Functional Assessment of Neuropeptides Adapted from Schmerberg, C. M. and Li, L. Protein Pept. Lett. 2012, May 23. (epub ahead of print) and Schmerberg, C. M., Chen, R., and Li, L. Prog. Neurobiol. 2012 [in preparation]. 3.1. Abstract A number of unique challenges are inherent to the study of neuropeptides (NPs), both in determining their molecular structure and their function. Traditional studies follow a model in which novel NPs are discovered and identified, then investigated for function. These studies frequently use biochemical techniques that can be imprecise and cumbersome. Mass spectrometry (MS)-based tools are becoming important not only in precisely determining the identity of a NP or quantifying a compound with a known sequence, but also in studies where identity and putative function can be determined simultaneously. Tools based on MS and tandem MS (MS/MS) have been developed, both with isotope labeling strategies and label-free methods, that allow accurate quantitation of NP changes associated with behavior or physiological manipulation, concurrent with identification of sequence. MS and MS/MS have also been implemented with sampling methods that incorporate temporal or spatial information while determining functional role of a NP, such as microdialysis (MD) and imaging mass spectrometry (IMS). These advances in MS and sampling techniques allow investigation of a particular biological phenomenon to guide studies aimed to identify and characterize NPs. Permitting function to drive identification of relevant compounds allows for a broader understanding of the molecular underpinnings of these events. The NPs thus identified can then be validated with more conventional techniques, and successive iterations of identification and function 124 determination will provide rich information about these compounds. This function-driven discovery of NPs using MS-based techniques is an important new approach for their study. 3.2. Introduction Neuropeptides (NPs) are a class of neural signaling molecules whose study has greatly expanded in the past several years. The physico-chemical characteristics of NPs relative to classical neurotransmitters require a new set of analytical tools to be developed for their study, but these distinct features also provide greater diversity of NP identity and functional specificity. NPs are short chains of amino acids, typically 4-10 residues in length in invertebrates, or 5-30 in vertebrates (reviewed in [1]). They are also defined by having the ability to signal between neurons, or between neurons and somatic targets. The great structural diversity and common sequence motifs among NPs of the same family allow for each peptide to have distinct and highly specialized functions as well as functions in common with other family members. This great structural diversity has led to challenges, however, in the discovery of NPs and elucidation of their function. NPs are synthesized as prepropeptides in neurons (reviewed in [1]). These long peptides then undergo a number of processing steps, including cleavage into smaller peptides and modification of residues, such as pyroglutamation or amidation of the C-terminus. These processing steps take place as the peptides are packaged into vesicles which are finally sent to the axon terminal for storage prior to release. After release by the neuron, NPs diffuse to the receiving cells, either across the synaptic cleft or after transport to a distant site via circulating fluid. NPs in circulation are commonly protected by binding proteins. The peptides then bind to their receptors, typically members of the G-protein coupled receptor superfamily, to transduce the signal to receiving cells. 125 3.2.1. Neuropeptide Detection and Sequence Determination by Classical Methods and Mass Spectrometry A variety of experimental strategies have been employed to study the identities and functions of NPs. These encompass both targeted and non-targeted methods, and are illustrated in Fig. 3.1. Detection of NPs has historically relied heavily upon targeted identification and quantification, frequently with antibody-based techniques. This is problematic due to the high degree of cross-reactivity that most NP antibodies have for multiple NPs, particularly those within the same family. It has recently become clear that most NP families have many members, and not all of these compounds have the same physiological effects, although antibodies cannot distinguish between them (reviewed in [1]). Cross-reactivity across NP families is also observed, especially in the case of NPs with common post-translational modifications, such as C-terminal amidation. Antibodies raised against the longer mammalian neuropeptides are frequently more specific owing to greater antigen complexity, although it is often unclear what portion of the sequence the antibody recognizes. This can lead to confusion when different NP isoforms occur and may have different function [2]. Determination of the exact identity of the active NP under this scheme requires purification of a large amount of sample to be subjected to Edman degradation, and more recently, mass spectrometry (MS) and tandem mass spectrometry (MS/MS) for sequencing. With the advent of soft ionization techniques for MS, the study of NPs has become more feasible and useful (reviewed in [1]). Slight differences in amino acid sequence among NP isoforms, which can lead to antibody cross-reactivity, create noticeable differences in the massto-charge (m/z) ratio observed by MS. MS/MS analysis provides NP sequence information by selecting analytes with a certain m/z in the first dimension of MS and fragmenting them into 126 smaller parts that are then subjected to MS analysis. Confident identification and sequencing of NPs directly from small biological samples is thus achieved with MS and MS/MS tools. MSbased analysis has identified thousands of putative NPs from a wide variety of species (reviewed in [1, 3-7]). However, many studies finding novel NPs by MS can only identify compounds as putative NPs based on sequence similarity to known NPs and/or the organs from which they are extracted. These identifications are either targeted to a particular molecular entity—as in quantification of NPs via antibody or MS-based techniques that require prior knowledge about the compound—or divorced from functional relevance—as in NP surveys that attempt to describe the entire NP complement of an organism via MS-based techniques. 3.2.2. Classical Methods of NP Function Determination and New Directions Determination of NP function with classical targeted methods also typically requires prior knowledge about its sequence. As mentioned previously, under the classical definition of a NP, it must have activity on neuronal or target cells. This activity frequently leads to a change in the electrical potential of a cell or the conductivity of ion channels by which this potential is established and maintained. Thus the gold standard for NP function determination is targeted electrophysiological study of isolated organs, neurons, or organ systems upon application of the NP in question (reviewed in [8]). These living cells are removed from animals and kept alive in a preparation, while various techniques are used to determine the electrical potential of one or more cells in the system. The NP is applied to the cells, and changes in the potential are noted. Studies of this type are able to accurately pinpoint the cellular action of NPs and their function in small neural circuits, but are difficult to implement—requiring specialized equipment and expertise—and require a pure sample of known composition. Determination of a NP’s function is frequently more complex than determining its sequence. These studies are also targeted to 127 specific putative NPs and particular functions—functional information is only obtained about the compounds applied on the electrical circuits investigated. These targeted techniques for determining NP identity and function provide highly specific detail about compounds for which a priori knowledge is available. However, by focusing on a single NP or physiological process, many other important compounds or functions may not be observed. Therefore, there are advantages to a non-targeted approach. One such alternative strategy for non-targeted MS-based NP study uses manipulation of processes known to be controlled by NPs concurrent with MS analysis of putative NPs (examples: [9, 10]), an approach here referred to as function-driven discovery. In this strategy, all compounds that appear to be altered in the course of the experimental manipulation can be monitored. Changes in these compounds occurring during the experiment can later be verified with more established methods for neuropeptide function validation, such as ex vivo electrophysiology as described above, to confirm its identity as a NP under the classical definition with greater confidence. An advantage is that the identity of a NP need not be determined prior to analysis; the use of MS-based tools allows for discovery and sequence identification of new NPs during the course of the experimental physiological manipulation, and no important compounds are missed by focusing only on a few target NPs. A few candidate compounds discovered in this way can then be the subjects of more targeted investigation with traditional techniques. Mass spectrometry-based function-driven discovery has expanded in recent years, and it will be the focus of this chapter. Several studies illustrating the use of many of these tools will be discussed as examples. Particular attention will be paid to studies of decapod crustacean NPs, as this is an area in which NP research has made great headway. The crustacean is an excellent system for NP discovery and functional study, as its nervous system is simple, contains relatively 128 large neurons in well-defined circuits, and is rich in NPs (reviewed in [1, 5, 8, 11]). Validation of the candidate NPs identified in function-driven discovery will also be discussed. 3.3. Function-Driven Discovery of Neuropeptides using Mass Spectrometry Function-driven discovery of NPs requires the ability to measure and identify putative NP candidates that are differentially expressed or secreted under experimental manipulation, either qualitatively or quantitatively. Although MS is a powerful tool for discovery and accurate identification of NPs in small samples, reliable quantitation using MS can be challenging. Obtaining samples for MS that are correlated with a particular dynamic behavior is also difficult. Toward these ends, a variety of innovative strategies for MS-based quantitation and live-animal sampling have been developed. The MS-based techniques are illustrated in Figure 3.2, and broadly fall into the category of function-driven non-targeted approaches, meaning that they do not require a priori information about analytes’ identities. These techniques, and MS used in conjunction with novel sampling methods, will be discussed in further detail. 3.3.1. Novel Mass Spectrometry Tools for Quantitation in Studies of Neuropeptide Function Quantitative analysis by mass spectrometry is complicated by the heterogeneity of ionization and unpredictable discrimination and suppression effects of complex mixtures. Several strategies have been developed to address these problems. Internal standards can be added to samples to account for day-to-day and run-to-run variability. Many of the most reliable MS-based quantitation methods, including selected reaction monitoring/multiple reaction monitoring (SRM/MRM), absolute quantitation (AQUA) by addition of isotopically labeled internal standard, and absolute quantitation calibration curves, fall into the category of targeted analysis and require prior knowledge of the analyte’s m/z and/or its fragments’ m/z, and these 129 methods are time-consuming to develop (reviewed in [7]). Often times, this prior knowledge is not available in a function-driven discovery study. Of these methods, SRM is more popular for determining NP concentration change as a means to elucidate function. SRM uses a first dimension of MS to isolate a single parent ion. It is then fragmented, as with MS/MS to determine peptide sequence. One daughter ion of this fragmentation is then analyzed by the second dimension of MS. This permits highly accurate quantitation, but only allows a single reaction to be monitored. For MRM, multiple parent-to-daughter transitions are monitored and quantified. The most salient feature of these quantitation modes is that they require prior knowledge of the analytes’ structure and fragmentation patterns, as well as generation of calibration curves to determine the relationship between the instrument response and analyte concentration, and thus can only be used in targeted studies of NP function. For these reasons, function-driven NP discovery can best be achieved with differential display (DD) and relative quantitation (RQ), due to the lack of a priori information. 3.3.1.1. Differential Display vs. Relative Quantitation Differential display (DD) and relative quantitation (RQ) are related and often employ the same analysis strategies. Relative quantitation describes changes in the quantity of a compound across different biological samples, often as a fold-change. DD is more often used to describe the all-or-nothing presence of NPs in some samples but not others. This can be of great importance in determining function, because NPs present in some conditions but absent in others can be expected to have an important role in the neurobiology of those conditions. However, since a fold-change cannot be established, it can be difficult to determine the dose-response effect of these NPs. DD data is often displayed as pairs of mass spectra, mass spectra with isotopic peak pairs (described below), Venn diagrams, or tables. 130 In comparison, relative quantitation (RQ) describes changes in the quantities of NPs across biological samples. Quantity changes are usually expressed as a change from baseline, or fold-change between different states. A variety of MS and MS/MS methods are used in RQ, with important features being the accuracy and reproducibility of quantitation. RQ data allows for determination of the dose-response effects of NPs on progressive behavior; for instance, increases in an anorexigenic NP during feeding may lead to reduced food intake rate, resulting finally in cessation of eating. This allows a more concrete link to be established between the NP and its function. RQ data is usually expressed in bar graphs showing fold-change in different NPs between two states, or line graphs showing fold-change versus time. 3.3.1.2. MS-Based Quantitation Methods for Functional Discovery of NPs RQ and DD data can be obtained using either MS or MS/MS techniques, and these methods can further be characterized by whether or not they employ isotopic labeling (label and label-free). Isotopic labeling is a method to prevent differential MS instrument effects from occurring in different samples. Different isotopes are incorporated into separate biological samples, the samples are mixed, and analysis of the mixed sample allows for determination of NPs present in one condition but not the other (DD) and/or changes in NP concentration (relative quantitation). Because the analytes from different conditions differ only in isotope composition, they experience the same instrumental conditions, and the response received for each can be used as a measure to quantitate them [10]. A number of methods for isotope-based MS quantitation exist. Metabolic labeling of whole animals is an exciting new field. In a scheme similar to stable isotope labeling in cell culture (SILAC), animals are raised on an isotope-enriched diet, provided by algae or bacteria that are grown in media containing a single isotope of a vital element, usually nitrogen-15 131 (reviewed in [12]). An additional experimental group is fed the same food source, with the exception that its food source is raised under normal conditions. Each experimental group can be subjected to different conditions expected to lead to changes in NPs (or left unperturbed, as controls), and samples can be collected and combined prior to analysis by MS. To prevent experimental artifacts due to the metabolic labeling, the groups should be switched. The DD of NPs across conditions can be determined by looking for the presence or absence of normal and isotopically-enriched compounds. In addition, relative quantity changes can be determined by comparing the heights of isotopic peaks on a single MS spectrum or the areas under the curve (AUCs) for the extracted ion chromatograms (XICs) for the normal and isotopic NPs if a separation method is coupled to MS, i.e. ultrahigh performance liquid chromatography (UPLC)MS. Although metabolic labeling of whole animals is a powerful tool, it is expensive, limited to animals that can be raised in the lab, and the global incorporation of isotopic elements into an animal may lead to different phenotypes. Most notably, high-anxiety mice fed 15 N diets had greater depression-like behaviors in a recent study [13]. This has profound implications for using metabolic labeling in studies of NP function. Other methods of isotopic labeling exist for functional discovery of NPs, including several where isotopes are incorporated as part of the sample preparation procedure. Several schemes exist to tag biomolecules with isotopes [14-19]. One scheme that has gained much success is dimethylation with formaldehyde and deuterium formaldehyde [18, 19]. This reagent adds two methyl groups to any primary amine in the peptide, such as the N-terminus or lysine side chain. If deuterium formaldehyde is used, these methyl groups will each contain 2 deuterium atoms, leading to a total mass difference of 4 Da per primary amine. Deuterium formaldehyde is relatively inexpensive and the addition of deuteriums to a molecule usually does not greatly alter 132 its behavior under analytical separation modes, such as liquid chromatography or capillary electrophoresis. As with metabolic labeling, samples of different isotopic composition can then be combined and analyzed concurrently. The ratios of peak heights obtained from a MS spectrum or ratios of AUCs from the XICs if separation is employed prior to MS can then be calculated to yield a fold-change in concentration of that analyte. A schematic illustrating this technique is shown in Fig. 3.2 under the heading “Mass Difference Tags.” This figure illustrates the pairs or triplets of peaks observed in such a MS spectrum. Most such tools use only two reagents, but depending on the isotope and label, more reagents may be possible. This technique has successfully been used to quantitate changes in the NP content of several ganglia of the decapod crustacean Cancer borealis after feeding [10, 19]. In Fig. 3.3, the mass spectrum obtained from the extract of brains of fed and unfed crabs is shown [10]. In this work, 12 NPs had statistically significant (p<0.01) expression level changes after feeding in the brain, and 6 were altered in the pericardial organ (PO). Mass difference tags are a useful tool for global assessment of relative abundance changes of NPs in small samples. Label-free methods for MS-based quantitation also exist, including standard addition (reviewed in [20]). A standard can be added to samples at the same concentration and the response of analytes relative to that of the ITSD can be determined. This method assumes that the response of the instrument to the NPs, relative to the response of a standard compound, is linear in the concentration range over which the analysis is run. With most mass spectrometers and typical working ranges, this is usually true. This technique is similar to absolute quantitation by standard addition, but skips the step where a calibration curve for the analyte and internal standard is created. Since many studies quantify hundreds of NPs at once, and the sequences of important NPs are not always known prior to analysis, it is impractical to generate calibration 133 curves for all analytes prior to analysis and standard addition without calibration can identify interesting target compounds for further study. For this type of analysis, ITSD (usually a NP from another species to avoid potential overlap and interference) is added to the sample, the sample is run on chromatography with MS detection, and software is used to create XICs for compounds that are abundant and that show NP-typical chromatographic profiles, characterized by cohesive peak shape and elution around the same time as the NP ITSD. The AUC is calculated for each and divided by the AUC for the ITSD. The concentration changes of these compounds can thus be tracked, and those that change in response to the experimental manipulation can be selected for further study, including determination of the NP sequence. This technique is illustrated in Fig. 3.2 under the heading “ITSD/AUC,” and is represented by a series of TICs obtained each of the samples. These TICs would then be extracted and normalized to allow quantitation of changes in NPs over time. Data obtained via standard addition and MS analysis must be treated as semi-quantitative and not definitive, but it is useful for the purpose of functional NP discovery. 3.3.1.3. MS/MS-Based Quantitation Methods for Function-Driven Discovery of NPs Tandem MS (MS/MS) can also be used for quantitation of NPs as part of a study to determine their function. MS/MS quantitation techniques have the advantage of providing sequence information in the same instrumental run as quantitative information, but they lack the sensitivity of MS-based techniques. As with MS-based techniques, isotopic labels can be incorporated into samples, or label-free methods can be applied. Tandem mass tags (TMTs) are reagents used for MS/MS quantitation of compounds, and these include isobaric tagging for relative and absolute quantitation (iTRAQ), dimethylated leucine-based tags (DiLeu) [19, 21], and others. A full description of the method by which these tags permit quantitation and 134 sequence identification is out of the scope of this dissertation, and a recent review is referred to for detailed discussion [21]. Briefly, TMTs come in sets of reagents. Each reagent is reacted with a different sample, and the labeled samples are then mixed. The tag adds the same total mass to the peptide for each reagent, but upon MS/MS fragmentation, each tag yields a characteristic low-mass reporter ion due to the incorporation of different numbers of isotopes. The full complement of peptide fragments are also observed, and thus comparing the peak heights of the low-mass reporter ions permits relative quantitation and the remaining sequence-specific fragment peaks allow for sequence identification. A graphical representation of this is shown in Fig. 3.2 under the heading “Tandem Mass Tags.” This shows the MS/MS spectrum from a TMT experiment, with fragment ions corresponding to the NP sequence present as well as low mass reporter ions that correspond to each of four samples. iTRAQ has been used for relative quantitation of NPs from distinct neurons in the F cluster of the cerebral ganglion of Aplysia californica [22]. This method is robust and reliable, although, since it relies upon MS/MS; a larger quantity of peptide must be present compared to MS-only techniques. Label-free techniques also exist for MS/MS-based RQ of NPs in function-based discovery studies as well. Spectral counting is a method that determines the relative abundance of a compound based on the number of MS/MS events for which it is selected. Since ions are selected for MS/MS fragmentation based on their relative intensity, this method is a reasonable estimation of relative abundances of corresponding peptides. A number of additional concerns must be addressed when conducting spectral counting, and these are reviewed in [20]. Data obtained from spectral counting is often used to generate a profile of how NP concentrations change relative to each other at each time point, and this is illustrated in Fig. 3.2 under “Spectral Counting.” Although consecutive runs on a single instrument can be compared for quantitative 135 analysis, especially if the spectral counts can be normalized to a peptide/protein that does not change across the time points, it is more reliably used to assess changes in the proportions of NPs that compose each sample. This is shown in the figure by illustrating changes in the relative amounts of NPs of each of several common crustacean NP families. Spectral counting and TMTs lack the sensitivity of MS-based quantitation because they require more of a compound to be present to provide MS/MS spectra of substantial quality. They also operate in data-dependent acquisition (DDA) mode for switching from MS to MS/MS, which is not always consistent due to sample and instrument variations, which will be discussed further in the following section. Several MS and MS/MS-based tools have been developed in recent years to enable quantitation of NPs without requiring prior knowledge of these compounds’ sequences. The ability to reliably quantitate and identify NPs that change during an important biological process has been a great boon to function-directed discovery of NPs. 3.3.2. Data-Independent MS/MS-Methods for Simultaneous Identification and Quantification 3.3.2.1. The Need to Conduct Data-Independent MS/MS Advances in the last few years have led to the development and implementation of new methods for acquiring MS/MS data that allow true identification and quantification in a single instrumental run by making acquisition of fragment ion spectra data-independent. These dataindependent acquisition (DIA) techniques include MSE, SWATH, etc. A recent review described some aspects of these techniques in the context of phosphoproteomics [23]. As mentioned previously, DDA MS/MS relies upon mass filtering of a single ion to be sent to the collision cell, followed by MS analysis of its daughter ions, and switching from MS to MS/MS mode is dependent on the ions observed in the MS1 spectrum. The addition of MS2 scans to the 136 instrument’s duty cycle decreases the number of instrument scans that can be used for MS1 acquisition. With typical chromatography conditions, in which peak widths are on the scale of minutes, most instruments then cannot obtain enough MS1 data points for proper peak integration due to the allocation of some scans to MS2. In addition to not having enough data points per MS1 peak, the number of points per peak will vary in each sample due to data-dependent MS to MS/MS switching, which is usually triggered by compounds reaching a threshold number of MS1 counts. The time at which switching occurs and the raw number of ion counts can vary due to slight variations in instrumental conditions over time. The concentration of the compound, which is roughly proportional to the number of MS1 counts, will also vary in each sample. Thus, instrumental and sample variability will influence how the instrument allocates scans to MS1 or MS2, and thus lead to different numbers of MS1 points per peak in each run. For these reasons, MS1 traces obtained via data-dependent acquisition (DDA) cannot be used reliably for area under the curve (AUC) methods of quantitation, TMT methods may not obtain information about the same precursors in different runs, and spectral counting will also experience variability. Data-independent acquisition methods attempt to solve these problems associated with DDA by producing fragment ions for all precursors equally, instead of selecting single highintensity parent ions. Prior to the advent of electrospray ionization (ESI), matrix-assisted laser desorption ionization (MALDI), and related “soft” ionization techniques, “hard” ionization methods including electron impact (EI) and chemical ionization (CI) were used as ion sources. These techniques not only ionized analytes, but also forced them to degrade into many components, and thus could be termed the first data-independent MS/MS instruments, as all analytes experienced a degree of fragmentation upon ionization. These methods had limited utility for biological samples (as opposed to small molecules) due to the complexity of the 137 resultant spectra. This is a result of the increased numbers and masses of molecular species in biological samples and the larger variety of functional groups where fragmentation can occur in biomolecules. Highly efficient molecular separation was required prior to hard ionization in order to provide interpretable spectra, and methods for separating hydrophilic biomolecules were not as well developed or as compatible with ionization sources (which typically required analytes to be in the gas phase) as were required. 3.3.2.2. Instrumental Techniques for Acquisition of Data-Independent MS/MS Several of the earliest schemes for data-independent MS/MS of biomolecules relied upon in-source fragmentation, a data-independent fragmentation method in which all analytes are fragmented prior to entering the mass analyzer. One can alternate MS acquisitions or scans between low fragmentation conditions and high fragmentation conditions to obtain scans of parent and fragment ions, but this typically cannot be done on the time scale of a LC experiment in order to provide quantitative information in addition to sequence identification. The first experiment using alternating scans in a single LC run to identify peptides was reported by Kosaka and colleagues in 2003 [24]. Infrared multiphoton dissociation (IRMPD) was applied on alternating scans of Fourier transform ion cyclotron resonance (FTICR) MS of nanoLC eluent in a method termed alternating-scan nano-LC/IRMPD-FTICR-MS. Two TICs were reconstructed from these alternating scans, with one corresponding to normal FTICR (parents) and the other to IRMPD-FTICR (fragments). Precursor masses from the parent scan were used to aid peptide sequencing in the fragment scan. The authors used this method to analyze tryptic digests of BSA and a protein purified from rat liver, with 55 and 46% coverage respectively [24]. Although the authors did not conduct quantitation, the TIC traces obtained appear to be suitable for AUC calculation. In-source fragmented bovine serum albumin (BSA) digests were also used by 138 Masselon and co-workers in 2003 to demonstrate that MS/MS spectra of mixtures of peptides could be interpreted without prior knowledge of which peaks corresponded to precursors and which were fragments, provided accurate masses were obtained [25]. Although this work demonstrated that it is not necessary to acquire spectra of parent and daughter ions separately, this is a major component of most data-independent acquisition methods used in the intervening years to reduce the complexity of MS/MS interpretation. Alternating in-source fragmentation methods was first used for quantification in addition to identification in 2003 [26, 27]. These techniques alternate source conditions between those that produce high or low degrees of fragmentation to produce spectra containing primarily parent or fragment ions, similar to the work by Kosaka et al. [24]. Purvine and colleagues termed this “shotgun” CID, employed it for simultaneous ID and quantification, and suggested a method to assign fragment ions to the appropriate parent ion using chromatographic retention profiles [27]. This same approach was also conducted in 2003 by Williams and colleagues, who also used a sequence of multiple levels of in-source fragmentation for more complete peptide fragmentation [26]. These methods were later incorporated into DIA techniques, i.e. alternating precursor and fragment scans, using retention time profiles to assign daughter ions to parent ions, and multiple levels of fragmentation efficiency (morphed into a collision energy ramp from low to high). Another technique for DIA, using large precursor mass isolation windows and rapid MS/MS scan acquisition, was proposed by Venable and colleagues in 2004 [28]. In this technique, a rapidly scanning ion trap mass spectrometer was used. For a single MS/MS scan, precursor molecules spanning a 10 Da window are accumulated in the ion trap, fragmented, and analyzed. The entire m/z range of interest is divided into these 10 Da segments, which are sequentially analyzed, and with the rapid scan speed of this type of MS instrument, 139 approximately 40 of these scans can be conducted in 10 s, corresponding to a total range of about 400 Da, with larger mass ranges possible if lower quality scans or larger precursor windows are used. Since the half-widths of most UPLC peaks are at least 30 s, this permits enough MS/MS spectra to be obtained across the elution peak for quantitation in addition to the identification information provided. Indeed, quantification was improved by increasing the signal-to-noise ratio and the greater specificity that comes from quantification based on a daughter ion as opposed to a parent ion. In addition to improved quantification concurrent with identification, the number of identifications increased [28]. This technique of selecting subsequent “chunks” of the mass range for fragmentation in a fast-scanning instrument is also currently used. These two schema for data-independent analysis—no isolation of precursors, or isolation of “chunks” of precursors in segments spanning the mass range of interest—underlie the mature forms of DIA that have become commercially available in recent years. A major hurdle in advancing DIA methods was that researchers outside of instrument companies often did not have the ability to alter instrument firmware to employ these strategies. Thus, further advancement was made primarily by researchers in instrument companies or in labs with home-built instruments. Patent protection of these techniques has also limited the ability of researchers outside of the companies that hold the patents to experiment with the techniques. The two schemes of DIA initially developed, as well as some similar strategies, are thus now proprietary technologies defined by the companies that hold the patents. The idea of alternating between parent and daughter ion spectra by changing global fragmentation efficiency, as put forth by the work of Kosaka and Purvine, was further advanced by using different energies in the collision cell, instead of at the source, with a high-resolution, high mass accuracy mass spectrometer by Waters Corporation. Beginning in 2005, Silva, 140 Hughes, and co-workers advanced one version of this technique, termed MSE—as the data acquired contains both normal and elevated Energy mass spectra—for protein/peptide analysis [29-32]. The resulting daughter ions are then detected in spectra containing fragments from multiple precursors (called chimeric spectra), and deconvolution is conducted to assign daughter ions to parents, based on elution profiles and the knowledge that a fragment ion cannot have a higher mass than the parent. This technique has been successfully used for simultaneous identification and quantification of proteins in a number of studies, in addition to those already mentioned [33-53], and is exclusive to instruments made by Waters. Another universal fragmentation technique has been implemented with the Orbitrap (Thermo-Fisher) mass spectrometer, called all-ion fragmentation (AIF). This technique sends alternating packets of ions either to the detector directly, or to a collision cell before sending them to the detector [54]. Fragmentation of all precursors, a technique that grew out of alternating fragmentation efficiency at the ion source, has been developed into two mature techniques for DIA, MSE on Waters instruments and AIF on Thermo Orbitrap instruments. The other main scheme for DIA MS—scanning large precursor ion windows as suggested by Venable et al.—has also been developed commercially. Some non-commercial research was initially conducted on this technique [55, 56], but it has subsequently been developed for use with ABSCIEX TripleTOF instruments and termed sequential window acquisition of the total high resolution mass spectrum (SWATH-MS) or MS/MSALL [57]. This technique is newer than MSE, and studies using it are less common. A similar technique that employs smaller mass ranges—2.5-3 Da isolated per MS/MS spectrum, isolation windows separated by 1.5-3 Da, 10-30 isolation windows monitored per run—and multiple sequential instrument runs to cover the entire m/z range of interest has been developed, called precursor acquisition independent from 141 ion count (PAcIFIC) [58-61]. This technique consumes large amounts of instrument time but provides high quality identification and quantification data, and is platform-independent. A final technique employs very large isolation windows (100 Da) on a Fourier transform ion trap mass spectrometer, and is termed Fourier transform-all reaction monitoring (FT-ARM) [62]. For this technique to work, the user must be able to set up the isolation windows for MS/MS acquisition (i.e., the instrument firmware must allow this), and the software to interpret the data is opensource but currently only works with Thermo files. “Chunk” isolation of precursors has been developed into SWATH-MS (MS/MSALL), PAcIFIC, and FT-ARM. Slight variations upon the technique permit more independent research in this technique. 3.3.2.3. Data Analysis in Data-Independent MS Chimeric spectra are the main challenge of DIA MS methods with both global and sequential segmented precursor selection and fragmentation methods. New methods for data analysis had to be developed to reduce the complexity of interpreting these spectra, as they have challenges not typically encountered in DDA MS/MS spectrum identification: more than one precursor mass is possible per spectrum, and the fragment ions present in a given spectrum can correspond to one or more of these parent ions. In contrast, DDA spectra have an identified precursor (the mass isolated in the first mass filter for fragmentation) and all fragments present in the spectrum ideally correspond to that single precursor. A number of strategies have been developed to interpret these spectra, and most are dependent on the method of acquisition of spectra. In 2003, Masselon and co-workers addressed the complexity of automated identification of precursors for MS/MS spectra that contain fragments of multiple compounds, both when there is limited precursor information (multiplexed MS/MS, a DDA method) or no precursor 142 information (true DIA, due here to in-source fragmentation) [25]. The identification of fragments produced by in-source decay was conducted by generating a list of possible peptides for the species and enzyme specified. The b- and y- ions of these potential precursor peptides were then calculated, and searched against the DIA MS/MS spectra obtained. Using this method, peptides from a BSA digest were identified by searching databases from the E. coli and C. elegans genomes, with the sequence of BSA added to them. Many identifications were made for BSA peptides (138 true positive peptide matches), with only a small number of false positives— 3 (2.8%) with the E. coli database, and 11 (7.8%) with the C. elegans database. This work bridges the gap between multiplexed DDA MS, for which interpretation of chimeric spectra was difficult but possible, and DIA MS, which has less information than multiplex MS/MS because a discrete set of possible precursor molecules is not known. Interpretation of chimeric spectra typically requires them to be “deconvoluted,” a process that describes the assignment of the mixed fragment ions to the appropriate precursors. This is normally achieved by using high resolution MS data with optimal separation. Ideal separation will allow every molecular entity in the sample to be distinguished by their analytical separation efficiency, and daughter ions can thus be assigned to parent ion. This degree of separation, in which all distinct compounds have different separation profiles, is not easy to achieve. The addition of another dimension of separation to the traditional UPLC-MS/MS experiment has been beneficial in this regard. Several studies have conducted multistage LC prior to ionization [36, 48]. MSE is marketed in some instruments that also have ion mobility separation capabilities, and this additional gas phase separation provides better resolution of analytes. Retention time (or drift time, in the case of ion mobility) can aid in deconvolution of chimeric DIA spectra. 143 Other information can be used to aid in deconvolution of chimeric spectra. In the cases of mass spectra acquired from sequential segments of the m/z range selected independently (as in SWATH-MS and related techniques), limited information about the precursor is available, namely that its m/z falls within the segment isolated at that moment. General principles of fragmentation can also be used for deconvolution, such as the fact that daughter ions will not have a greater monoisotopic mass than their parents. Instrument-specific software is typically used to make DIA MS data more easily interpreted by software originally developed for DDA data, but open-source software is becoming more common for deconvolution of chimeric spectra, including FT-ARM [62] and DeMux [55]. 3.4. Novel Sampling Methods for MS-Based Functional Discovery of Neuropeptides In addition to new MS tools for quantification of NPs, different sampling methods for use with MS-based analysis have also been established. These techniques can provide more significant temporal information about changes in NP content, or can be used to determine the localization of NPs within well-defined neural organs or neural circuits that have areas with known function. 3.4.1. Live-Animal Sampling for Correlation of Neuropeptide Changes with Behavior Many MS-based functional discovery studies rely on the use of tissue-based sampling techniques which typically require sacrificing multiple animals for each biological sample, such as brain or ganglion homogenates. Although these sampling methods have been used to assign function to many NPs, high degrees of inter-animal variability in baseline levels of NPs can obfuscate net changes that may be important to NP function. Obtaining multiple samples from a single animal is thus highly desirable. Bodily fluids may be sampled via needle, but in mammals, the blood-brain-barrier (BBB) prevents changes in many NPs from being reflected in circulating 144 levels of these compounds, and obtaining cerebrospinal fluid (CSF) is invasive. For crustaceans, the BBB is less complex, and sampling from the hemolymph provides greater information about active NPs [63]. However, hemolymph and CSF contain a wide variety of other components, and obtaining a sample of hemolymph from a marine animal via conventional syringe withdrawal technique induces a great deal of stress to the animal. The optimal sampling method would allow multiple samples to be taken from the animal on a time course relevant to the experimental manipulation without disturbing the animal. Thus, liquid-based sampling is preferred, and dynamic sampling on-line with the animal (i.e. microdialysis (MD), introduced previously) would be the best. On-line sampling allows continual collection of sample without disturbing the animal’s behavior. Two main methods for this type of sampling exist: microdialysis and push-pull perfusion. Cannulation can also be conducted but can lead to animal perturbation due to removing liquid from the animal without replacing the volume and salts lost. Push-pull perfusion is a variation on cannulation that employs an inlet cannula in addition to the withdrawal cannula in order to replace the liquid removed with an equal volume of physiological fluid. To the authors’ knowledge, the only online sampling technique successfully implemented in crustaceans is microdialysis, likely due to the difficulty of surgery on the crab. In the authors’ experience and previously published work, crabs do not heal well from insult to the carapace as opposed to the soft tissue at joints due to limited glue adhesion to the shell. However, this technique has been used successfully to sample from crayfish [64] and the crabs Cancer borealis [65, 66], Callinectes sapidus, Carcinus maenas, and Cancer irroratus. MD has long been used in neuroscience to sample NPs and neurotransmitters directly from the brains of mammals, but its use in conjunction with MS-based analysis approach has 145 only recently been established (reviewed in [67, 68]). MD employs a probe that is surgically implanted into the tissue of interest. At the tip of this probe is a dialysis membrane, through which molecules from the area outside the probe can diffuse into the probe. Liquid is perfused through the probe at a low flow rate (µL/min), and is collected at the probe outlet. This liquid carries with it molecules that diffuse through the dialysis membrane into the probe according to their concentration gradient. After recovery from the surgical implantation, animals can behave normally, and thus with MD, molecules can be collected from an animal while it is behaving normally with minimal perturbation. This allows maximal correlation of behavior with neurochemistry, and permits multiple samples to be taken from the same animal, to limit the impact of inter-animal variability. The pores of the dialysis membrane also only permit molecules below a certain molecular weight (the molecular weight cutoff (MWCO)) to pass into the probe, thus reducing sample complexity, an advantage not obtained with push-pull perfusion. Additional work needs to be conducted, however, to improve the sensitivity of analysis for microdialysates, as the temporal resolution currently available with the technique is 4 hours or greater. This is due primarily to the low concentration of NPs present in the hemolymph, as well as the dialysis membrane impeding the influx of molecules with weights below the MWCO but greater than a few hundred Da. The percent transfer of molecules through the membrane decreases in an exponential manner as mass increases, with the MWCO typically representing the mass at which 95% of molecules will not pass through. Different probe membrane materials may also impact the recovery of larger molecules. Affinity agents can be added to the liquid perfusing the probe to increase the net flux of analyte into the probe. These affinity agents, usually solids with some molecule with NP affinity attached to their surface, remove the bound analyte from the diffusion equation that controls flow of the analyte inside at the probe tip. This 146 method, termed affinity-enhanced microdialysis (AE-MD), was pioneered by Stenken and colleagues in vitro and in mammalian systems [69-81]. Application of AE-MD in crabs has increased temporal resolution to 30 minutes, and the concentration change of neuropeptides following feeding can be observed with this technique. On-line sampling from invertebrates is not simple, particularly for smaller invertebrates, due to the challenge of implanting a MD probe or cannulae into an animal with an exoskeleton. It also suffers from reduced sensitivity due to the low concentration of NPs in the extracellular fluid. However, the benefits of sampling from the animal without disturbing it and the ability to obtain multiple time points from the same animal make it a very attractive option for generating functionally relevant information about NPs. 3.4.1.1. Methods of Analysis for Microdialysates Typical methods for quantitative analysis of microdialysates (the liquid collected at the probe outlet) include immunochemistry and chromatographic separations coupled with electrochemical, ultraviolet-visible spectroscopy, photometric, and/or SRM/MRM-based MS detection methods (reviewed in [7, 67]), all of which require a priori knowledge of the NP’s structure or chromatographic properties. A number of NP MD studies using immunochemical detection have been conducted, however, these analyses often suffer from antibody crossreactivity with the NP of interest. As a result, it is often difficult to clearly assign functions to single molecular entities, and there are instances in which NP isoforms with slight variations in sequence have different biological effects. Although these quantitative analyses of MD samples are often part of studies of NP function, they usually rely upon quantitation that is less specific (antibody-based) and/or require prior knowledge of the NP’s structure (targeted MS techniques 147 such as SRM and AQUA), and thus are less well suited for discovery of NPs based on global comparative analysis of NPs from different functional states. Studies where MS-based techniques are used for discovery of NPs found in dialysate, in contrast, usually do not also include quantitative analysis, and thus provide only minimal functional information [65, 66, 82, 83]. A limitation of MS-based functional discovery for NPs is that it is difficult to obtain sequence information about novel NPs concurrent with quantitative analysis methods described previously, except when TMTs or spectral counting are used. Most tandem MS instruments are operated in a mode in which they dynamically switch between MS and MS/MS. Sequence information is available only from MS/MS scans, which are added to the duty cycle of the instrument in place of MS scans. This addition to the duty cycle leads to less frequent acquisition of MS data. Therefore, the number of MS data points obtained per peak decreases during the run in a data-dependent manner, and this is not reproducible between runs. Integrating the AUC for the ion of interest is not reproducible for data acquired when dynamic MS to MS/MS switching is used. Separate instrument runs must be conducted for identification based on MS/MS sequence and quantification based on MS, for which ions can be identified by their m/z and chromatographic properties. 3.4.1.2. Quantitation of NPs in MD Samples with Targeted MS Methods for Elucidation of Function MS quantitation methods that require prior knowledge of NP sequence to quantitate these compounds in dialysates from mammals have been reported previously [84-88]. MD studies in invertebrates are less common. Several related papers that demonstrate such a targeted approach to MS-based quantitation of NPs in MD samples will be highlighted here to demonstrate the potential of MD used in conjunction with MS. Although they are not function-driven 148 experiments, the same sampling techniques could be used with non-targeted MS approaches to conduct function-driven NP discovery. In these studies, capillary LC (capLC)-MS/MS [89] and capLC-MS3 [90-92] using SRM of known fragmentation reactions for selected NPs was conducted to quantitate NPs collected via MD from the basal ganglia of anesthetized and freelymoving rats under baseline and stimulated conditions. As mentioned previously, SRM requires that the structure of the analyte and its daughter ions is known prior to analysis. MS3 for SRM adds another level of MS analysis, in which the daughter ions generated upon MS/MS analysis are selected and fragmented, and a specific grand-daughter ion of this 2nd order fragmentation is monitored. These targeted MS techniques provide stronger evidence for a correlation between NP concentration changes and stimulation, but require a priori knowledge of the NPs in question and thus cannot be used in a function-driven discovery experiment. These studies used two types of stimulation, which provide different amounts of information about the function of the NPs monitored. Several employed K+-induced widespread neuronal depolarization in the globus pallidus (GP) [89] and striatum [90, 91] to induce the release of enkephalins [89-91] and dynorphins [91]. Although the non-specific nature of this stimulation does not permit assignment of an exact functional role for these NPs, correlating their release more generally with activity in the basal ganglia may point toward a functional role related to the overall functions of these subnuclei. The most recent of these studies also employed capLC-MS3 quantitation but used more biologically relevant manipulations concurrent with behavioral monitoring to determine a more specific function for these compounds [92]. This study quantitated enkephalins in GP microdialysate from rats along with locomotor behavioral analysis, monitoring neurotransmitter (NT) concentration changes, and pharmacological manipulations. The SRM capLC-MS3 149 quantitation system mentioned above was used to quantitate [Met]- and [Leu]-enkephalin (ME and LE) concurrent with administration of drugs that increase dopamine levels (amphetamine) or are dopamine receptor 1 or 2 (D1R or D2R) blockers. The researchers were able to determine that ME and LE increase after administration of amphetamine, and that this effect primarily requires the D1R. Further monitoring of NT concentrations and locomotor behavior during administration of D1R, D2R, and opioid receptor antagonists permitted the development of a model for the role of ME and LE in locomotor function mediated by the GP. This study is highly significant due to the great specificity with which the authors were able to determine excitatory and inhibitory effects of neurochemicals. This was accomplished through the combined use of MS-based quantitation of NPs, a novel method for NT quantitation, behavioral modeling, and selective pharmacologic agents. This targeted study determined a specific function for ME and LE, based on MS analysis of MD samples. 3.4.1.3. Discovery of NPs in MD with MS/MS Tools MS tools can also be used to identify unknown NPs, and a number of such studies have been conducted [82, 93-95]. One such work is complementary to the ME and LE quantitation studies, and consists of MS/MS-based discovery of NPs from the striata of rats under K+stimulated conditions [83]. This study identified 29 peptides from 6 proteins, 25 of which were isoforms that had not previously been observed. The combination of NP identification with the quantitation studies discussed previously comprises an alternative approach to function-driven NP discovery. The decision of which NPs to quantify was based on previously known information about these NPs. A function-driven discovery experiment would instead look for compounds with interesting changes throughout the course of an experimental manipulation, and then attempt to identify them [65]. The power of functional discovery of NPs via MS-based 150 quantitation methods discussed in this article is that the data obtained upon experimental manipulation itself determines which NPs are quantified and subjected to further study. 3.4.1.4. Experimental Design for Functional Discovery of NPs in MD Samples True function-driven discovery of NPs requires functional relevance to guide the choice of which compounds to study further. This can be achieved in a number of ways, but has rarely been done with MD. MS-based relative quantitation of several isoforms of dynorphin A (Dyn A) was conducted on samples collected from rats with a unilateral lesion of the striatum [96]. The identity of the isoforms was then determined by peak mass matching and MS/MS analysis. This study revealed qualitative and quantitative differences in the processing of Dyn A in the lesioned striatum. By quantitating all peaks of interest and later determining their identity, this study conducted function-driven discovery of NPs. MS/MS-based quantitation techniques for function-driven discovery have been used rarely with MD samples. These techniques permit quantitation and identification in a single run, without prior information, but they often require larger amounts of sample, which makes them unpopular for MD. A study of this type used spectral counting to characterize the NP content of the striatum of the rat brain under baseline and K+-stimulated conditions [97], similar to the studies mentioned previously. In this work, 88 proteins and 100 endogenous peptides from 26 protein precursors were identified and quantified. The relative abundances of these compounds in dialysate based on spectral counts were determined, and these relative abundances were observed to change upon K+-depolarization. NPs made up a greater proportion of the components observed in microdialysate after depolarization. In addition, the relative abundances of two NPs, proenkephalin A precursor and a VGF-related peptide, were greater postdepolarization. As the method of stimulation and location of probe placement was similar to 151 previously mentioned studies, the amount and type of functional information obtained from this study is also similar, although additional compounds not previously detected were found and their quantity changes determined. One caveat is that a number of large proteins were detected, including many above the MWCO of probes used. This suggests possible damage to the tissue surrounding the probe causing large-scale protein degradation. However, this study acquired both structural and quantitative information in the same LC-MS/MS run, which is a great boon for function-driven discovery of NPs. MD has been conducted on alert Jonah crabs (Cancer borealis) under baseline conditions with MS and MS/MS analysis of the resulting samples [65, 66]. Although no quantitative results were available in that study, 35 NPs previously identified in C. borealis were detected. A major limitation in the application of this scheme for function-related discovery of NPs was the long collection time of MD samples. Collections were at minimum 4 hours, which is not compatible with studying most of the dynamic processes thought to be mediated by NPs. This long collection time was required due to the low sensitivity of the systems used for detection. New advances in UPLC-MS and improvements to the MD collection technique termed affinityenhanced microdialysis have made it possible to follow concentration changes in NPs on a shorter time scale with 30 min sampling intervals [98]. These enhancements have made it possible to determine the concentration changes in NPs induced by feeding in C. borealis, and have provided additional evidence that several NPs have feeding-related functions. Further refinement of the techniques used may allow for collection of samples on an even faster time scale, which will provide useful information about the time-resolved changes in these compounds with respect to feeding, and thus aid in elucidating the NPs involved in this important physiological event. MD sampling of alert, behaving animals, combined with MS and 152 MS/MS techniques described previously, allows for tight correlation of NP changes with behavior, and thus is a useful technique. 3.4.2. Mass Spectrometry Imaging of Tissue to Correlate Neuropeptide Location with Function Not only do MS-based tools permit NP function to be elucidated by correlating behavior with release over time, they allow determination of NP localization within tissue as a guide for function. Mass spectrometry imaging (MSI) is an emerging technique, in which a tissue of interest is sectioned and analyzed with MS (recent reviews [4, 99-104]. A coordinate system is superimposed on the tissue, and mass spectra are acquired for and registered to positions within this system. The intensity of a given peak in the spectra can be compiled into a heat map image to represent the distribution of that compound throughout the tissue. This technique has been applied with great success to determine the localization of NPs within the brains of several species (see selected reviews and relevant invertebrate papers [3, 99-115]). Previous knowledge about the function of certain brain structures can then be extrapolated to NPs that are heavily concentrated in these structures. The brain of the decapod crustacean has been studied extensively, and several regions have known functions due to the locations from which they receive inputs and to which they send outputs. For instance, the olfactory neuropil (ON) and antennal lobes (AL) in the brains of decapod crustaceans both receive input from olfactory receptors, although the AL receives higher-order olfactory input—the olfactory signals are relayed through a set of interneurons before they reach the AL—and input from other senses [116]. In an IMS study of the brain of H. americanus, a number of patterns of NP expression were determined, including a greater expression of two SIFamide family peptides in the ON, and increased levels of two orcokinins in 153 the AL [113] (shown in Fig. 3.4). The difference in localization of these two families of peptides may indicate different functional roles, i.e. SIFamides are more important in primary processing of olfactory signals, and orcokinins are involved in higher-order processing, including integration of signals from multiple sensory modalities. MSI is a useful tool that allows assignment of function to a NP based on its localization patterns within nervous structures with known functional areas. In conjunction with novel tools for quantitation with MS, advances in sampling have allowed the identification and assignment of NP function by providing information about their temporal and spatial distribution. 3.5. Validation of Potential Function for NPs Discovered via MS-Based Methods Although function-driven discovery of neuropeptides using MS-based tools provides definitive information about NP identity, the functional information is often less complete; more targeted studies are frequently required for definitive assignment of function. Putative functions identified by MS-based functional discovery of NPs can be further investigated using a variety of targeted tools, including more traditional bioassays, “omics”-based approaches, and some previously mentioned MS techniques. Similarly, many of the methods used for functional discovery of NPs can also be used to determine the identities of compounds with functions already known. Thus, a cycle of identifying compounds and determining their functions forms the basis of neuropeptide research (Fig. 3.1). Some of the techniques commonly used to validate the putative functions of NPs discovered by non-targeted MS strategies will be described. There are many methods to determine the role a NP plays in a biological system. NPs can be administered to an animal, and neurochemical and/or behavioral changes can be monitored. Endogenous NP content also can be monitored during the course of a physiological change to provide correlations between NPs and functions. Although the first technique provides a more 154 definitive causal relationship, it requires the administration of a pure sample of a known compound, and a well-defined behavior or physiochemical change to be monitored. Thus, a great deal of foreknowledge is required to carry out this type of functional relationship study, and it is best suited to verification of postulated functions. The second type permits discovery of previously unknown NP-function relationships but provides less definitive information. 3.5.1. Causal Studies of NP Function Two main methods have been used to monitor NP action in decapods via direct administration. These are ex vivo nervous system or chromatophore preparations, monitored by electrochemical recording or microscopy, and in vivo injection of the decapod with the substance, followed by monitoring behavioral and/or hemolymph chemical changes. The ex vivo preparation allows—via clever use of pharmacological agents and electrophysiological techniques—determination of the mechanism of NP action at the molecular level. An in vivo experiment captures whole-animal behavioral changes that do not translate well to the cellular level and changes that require the involvement of multiple organ systems, such as metabolism of stored energy. 3.5.1.1. Ex Vivo NP Function Experiments A common method for determining the function of a putative NP is to isolate a neural organ, muscle, or portion of the nervous system, and apply the NP to the preparation. Changes in the electrical profiles of neurons and muscles or in the contraction frequency or strength of a muscle can then be recorded, and thus are correlated with the NP applied. These studies, in which an organ or muscle is removed but kept alive and used as a test bed for NP action, can be termed ex vivo studies. A number of different ex vivo preparations have been described in invertebrates, but the classic model is the isolated crustacean stomatogastric nervous system 155 (STNS), a set of ganglia connected by nerves located near the stomach. For detailed instructions on this preparation, see videos and articles by Gutierrez and Grashow and Tobin and Bierman [117, 118]. Fig. 3.4 illustrates the basic preparation and how the function of a bath-applied NP can be determined using this system. The STNS comprises a central-pattern generating circuit in control of feeding, with each cell in each ganglion having a characteristic rhythmic firing pattern. The cell number, location, and connectivity in the STNS are nearly identical in all mature members of each species. For causal studies of NP function using this system, the STNS is dissected and placed in an electrophysiological recording dish, and a variety of electrodes are placed in selected cells and on nerves. NPs can then be applied to certain cells, ganglia, or the entire preparation, and alterations in the firing frequency and pattern of these cells can be recorded. Two distinct patterns are observed, one to control chewing—the gastric mill rhythm— and one to control filtering of food—the pyloric rhythm. The addition of pharmacological agents with known function along with the NPs or modern electrophysiological techniques such as voltage clamping can allow for elucidation of their exact molecular targets. This system was pioneered by Maynard and extensively used by Marder and colleagues to determine the function of several NPs on the rhythmic patterns of the gastric mill and pylorus. For a review of the known functions of NPs on this system, see a review by Marder and Bucher [119]. The ex vivo STNS preparation allows for identification of the molecular mechanisms by which a NP acts on the cells in this system, and has provided important information on the function of many crustacean NPs. Isolated chromatophores (cells in the epithelium or eye that change color) have been used to determine the activity of several pigment-related hormones. Simply, chromatophores are isolated from the eyestalk, placed on a microscope slide, and NPs are applied [120, 121]. This 156 causes pigments in the cell to change their distribution, which can be observed via microscopy. The molecular signaling pathways by which externally applied NPs cause redistribution of pigments in a crustacean chromatophore are varied depending on whether the NP is pigmentdispersing or pigment-concentrating. For further information, see the sections on pigmentaffecting hormones in Chapter 2. The ex vivo STNS and chromatophore preparations can be used to determine the molecular targets and functions of certain NPs that act on cells in these systems. This is useful for NPs that act very specifically there, but less useful if the action of a NP is distributed across multiple organs. 3.5.1.2. In Vivo Administration of NPs to Determine Function NPs can also be administered to the animal as a whole, and changes in behavior or bioactive chemicals can then be monitored. Provided administration of the NP causes a change greater than that seen following administration of a carrier solution (physiological saline), that NP can be assumed to have exacted that change. Administration is typically conducted via injection of a solution into the soft tissue near a joint, although implantation of a permanent cannula would also be appropriate if multiple doses were required. Behavioral changes monitored might include the rapidity of movement, frequency of a certain pose being assumed, or movement of certain parts of the body, such as the tail or mouth parts. Biochemical changes caused by NP administration can be determined by sampling the extracellular fluid/blood of the crustacean, again through the soft tissue at a leg joint. Although microdialysis is appropriate for both NP administration and sampling from the hemolymph in this type of experiment, and is indeed preferable in terms of invasiveness, it has not yet been applied for either of these roles in the crustacean. 157 An NP can also be administered in vivo and hemolymph chemical changes or behavioral changes can be monitored. A recent study of this type has been presented by Chung et al. [122], in which crustacean hyperglycemic hormone (CHH) was administered to eyestalk-ablated Chionoecetes bairdi, after which hemolymph glucose levels were monitored.The eyestalk is the primary endogenous source of CHH in the crustacean, and removing it provides better causal evidence that the CHH injected led to the increases in hemolymph glucose level observed. By this method, Chung and colleagues were able to demonstrate CHH’s role in the mobilization of stored energy, a process that requires the NP to act on multiple organs. Both ex vivo and in vivo studies can be used to determine the causal relationship between NPs and functions, on a protein, cell, or organismal level. However, these causal relationship studies require administration of a pure NP sample and monitoring of a well-defined output that is already postulated to be the function of the NP. In addition, they typically only allow for determination of the function of a single NP at a time, and it is well known that NPs can attenuate and alter each other’s function. 3.5.2. Correlational Studies of NP Function in Invertebrates Due to the drawbacks of causal studies of NP function in invertebrates mentioned above, correlational studies are often more suitable. When mass spectrometry is used, many methods for causal study of NP function are identical to the function-driven discovery methods discussed previously. Most of previously mentioned MS tools that improve quantitation of multiple NPs across different time points can be used when the chemical identity of the NP is known, often with better success than when it is unknown. There is an inherent disadvantage to correlational studies in that they do not have the specificity in determining NP-function relationships or functional unit resolution that is seen in causal experiments, but they are an important part of determining the putative function of a NP. 158 For correlational studies, the neuropeptide content in several time points or different conditions must be determined. A number of challenges arise with this requirement; namely what type of sample to obtain (liquid or tissue) and how to conduct quantitation. The previously discussed quantitation schemes and novel sampling methods are of particular use for this component of causal studies of NP function. 3.5.3. Peptidomics to Genomics: Function Determination by Homology Search of Public Databases MS-based discovery of NPs can also be combined with homology searches to verify function. The function of many NPs is conserved throughout evolution [1, 123-128]. Recent reviews have discussed the technique of database mining for discovery of novel NPs [8, 129131], and searching sequences obtained via MS for homologous peptides in other related species follows a similar procedure. A novel NP sequence identified by MS/MS can be searched against the publicly available translated nucleotide database of expressed sequence tags (ESTs) using tblastn. The EST database contains a large number of sequences from many organisms and is constantly being expanded. Protein BLAST is not suitable for most peptides due to their short sequences. Searches of this type may yield a homologous peptide in a different species, for which the function is already known. Due to the conservation of function for many NPs across disparate species, a putative function for a NP can thus be verified. Many NPs have been predicted from bioinformatics analysis of publicly accessible databases, verified to exist in vivo via immunochemical or MS methods, and/or determined to have biological function via bioassays (notable invertebrate examples [127, 130-145]). Homology searching is not only a tool for identification of NPs, but also provides information to their function and can verify the results of a function-driven discovery study. 159 3.6. Summary and Perspective New tools and approaches that rely upon mass spectrometry and novel sampling methods have been developed that allow researchers to pursue non-targeted function-driven discovery of new compounds. These tools have the unique advantages of allowing functional information about NPs, whether it comes from following concentration changes over time or determining the distribution in a tissue, to be acquired concurrent with or before determination of the sequences of these compounds. The power of this approach is that all NPs with relevance to a particular function can be analyzed; one does not have to determine which compounds will be the focus of an experiment prior to conducting it. The added benefit of MS’s ability to distinguish between related NP isoforms increases the power of these analyses and permits knowledge of the exact molecular entity that is active in the biological system. These powerful tools also enable deciphering functional consequences of NP multiplicity and structural diversity. In fact, previously unknown and unpredicted isoforms of mammalian NPs have been observed in MSbased functional discovery studies via the analysis of microdialysate [89, 97]. Previously, a molecular entity had to be characterized prior to determination of its function; now MS-based function-driven discovery can identify NPs with putative functions that can then be validated with more conventional means. In this way, NP identification and functional determination can be accomplished in a cycle (Fig. 3.1), with multiple iterations providing a more complete picture of both the NPs’ identities and functions. By eliminating the need for prior knowledge, new and exciting NPs may be discovered with important functional roles, and vice versa. In addition, the field of mass spectrometry has experienced constant growth and innovation, and advancements in MS technology improve the sensitivity, speed, dynamic range, and accuracy of analysis on a regular basis. Although the NPs observed most in current MS 160 studies are likely to be the most abundant in vivo, future improvements will lead to elucidation of roles for those NPs that are currently below the limits of detection or quantitation for current instruments. By employing MS in function-driven non-targeted NP discovery workflows, even greater amount of information will be able to be achieved as new technology is developed. The types of experiments that will be possible with future generations of MS instrumentation will allow greater information to be gained about the molecules that mediate intrinsic physiological properties. 3.7. Works Cited 1. Li, L. J.; Sweedler, J. V. Peptides in the Brain: Mass Spectrometry-Based Measurement Approaches and Challenges. Annu Rev Anal Chem. 2008, 1, 451-483. 2. Morimoto, R.; Satoh, F.; Murakami, O.; Totsune, K.; Saruta, M.; Suzuki, T.; Sasano, H.; Ito, S.; Takahashi, K. Expression of peptide YY in human brain and pituitary tissues. Nutrition. 2008, 24, 878-884. 3. Hummon, A. B.; Amare, A.; Sweedler, J. V. Discovering new invertebrate neuropeptides using mass spectrometry. Mass Spectrom Rev. 2006, 25, 77-98. 4. Boonen, K.; Landuyt, B.; Baggerman, G.; Husson, S. J.; Huybrechts, J.; Schoofs, L. Peptidomics: the integrated approach of MS, hyphenated techniques and bioinformatics for neuropeptide analysis. J Sep Sci. 2008, 31, 427-445. 5. Christie, A. E.; Durkin, C. S.; Hartline, N.; Ohno, P.; Lenz, P. H. Bioinformatic analyses of the publicly accessible crustacean expressed sequence tags (ESTs) reveal numerous novel neuropeptide-encoding precursor proteins, including ones from members of several little studied taxa. General and Comparative Endocrinology. 2010, 167, 164-178. 6. Fricker, L. D. Analysis of mouse brain peptides using mass spectrometry-based peptidomics: implications for novel functions ranging from non-classical neuropeptides to microproteins. Mol Biosyst. 2010, 6, 1355-1365. 7. Perry, M.; Li, Q.; Kennedy, R. T. Review of recent advances in analytical techniques for the determination of neurotransmitters. Anal Chim Acta. 2009, 653, 1-22. 8. Christie, A. E.; Stemmler, E. A.; Dickinson, P. S. Crustacean neuropeptides. Cell. Mol. Life Sci. 2010, 67, 4135-4169. 9. Cape, S. S.; Rehm, K. J.; Ma, M.; Marder, E.; Li, L. Mass spectral comparison of the neuropeptide complement of the stomatogastric ganglion and brain in the adult and embryonic lobster, Homarus americanus. Journal of Neurochemistry. 2008, 105, 690702. 10. Chen, R.; Hui, L.; Cape, S. S.; Wang, J.; Li, L. Comparative Neuropeptidomic Analysis of Food Intake via a Multi-faceted Mass Spectrometric Approach. ACS Chem Neurosci. 2010, 1, 204-214. 161 11. Skiebe, P. Neuropeptides in the crayfish stomatogastric nervous system. Microsc Res Tech. 2003, 60, 302-312. 12. Gouw, J. W.; Krijgsveld, J.; Heck, A. J. Quantitative proteomics by metabolic labeling of model organisms. Mol Cell Proteomics. 2010, 9, 11-24. 13. Frank, E.; Kessler, M. S.; Filiou, M. D.; Zhang, Y.; Maccarrone, G.; Reckow, S.; Bunck, M.; Heumann, H.; Turck, C. W.; Landgraf, R.; Hambsch, B. Stable isotope metabolic labeling with a novel N-enriched bacteria diet for improved proteomic analyses of mouse models for psychopathologies. PLoS One. 2009, 4, e7821. 14. Ahrends, R.; Pieper, S.; Kuhn, A.; Weisshoff, H.; Hamester, M.; Lindemann, T.; Scheler, C.; Lehmann, K.; Taubner, K.; Linscheid, M. W. A metal-coded affinity tag approach to quantitative proteomics. Mol Cell Proteomics. 2007, 6, 1907-1916. 15. Guo, K.; Ji, C.; Li, L. Stable-isotope dimethylation labeling combined with LC-ESI MS for quantification of amine-containing metabolites in biological samples. Anal Chem. 2007, 79, 8631-8638. 16. Morano, C.; Zhang, X.; Fricker, L. D. Multiple isotopic labels for quantitative mass spectrometry. Anal Chem. 2008, 80, 9298-9309. 17. Xu, M.; Yan, X.; Xie, Q.; Yang, L.; Wang, Q. Dynamic labeling strategy with 204Hgisotopic methylmercurithiosalicylate for absolute peptide and protein quantification. Anal Chem. 2010, 82, 1616-1620. 18. Wang, J.; Zhang, Y.; Xiang, F.; Zhang, Z.; Li, L. Combining capillary electrophoresis matrix-assisted laser desorption/ionization mass spectrometry and stable isotopic labeling techniques for comparative crustacean peptidomics. J Chromatogr A. 2010, 1217, 44634470. 19. Xiang, F.; Ye, H.; Chen, R.; Fu, Q.; Li, L. N,N-dimethyl leucines as novel isobaric tandem mass tags for quantitative proteomics and peptidomics. Anal Chem. 2010, 82, 2817-2825. 20. Neilson, K. A.; Ali, N. A.; Muralidharan, S.; Mirzaei, M.; Mariani, M.; Assadourian, G.; Lee, A.; van Sluyter, S. C.; Haynes, P. A. Less label, more free: approaches in label-free quantitative mass spectrometry. Proteomics. 2011, 11, 535-553. 21. Treumann, A.; Thiede, B. Isobaric protein and peptide quantification: perspectives and issues. Expert Rev Proteomics. 2010, 7, 647-653. 22. Rubakhin, S. S.; Sweedler, J. V. Quantitative Measurements of Cell−Cell Signaling Peptides with Single-Cell MALDI MS. Analytical Chemistry. 2008, 80, 7128-7136. 23. Blackburn, K.; Goshe, M. B. Challenges and strategies for targeted phosphorylation site identification and quantification using mass spectrometry analysis. Brief Funct Genomic Proteomic. 2009, 8, 90-103. 24. Kosaka, T.; Yoneyama-Takazawa, T.; Kubota, K.; Matsuoka, T.; Sato, I.; Sasaki, T.; Tanaka, Y. Protein identification by peptide mass fingerprinting and peptide sequence tagging with alternating scans of nano-liquid chromatography/infrared multiphoton dissociation Fourier transform ion cyclotron resonance mass spectrometry. Journal of Mass Spectrometry. 2003, 38, 1281-1287. 25. Masselon, C.; Paša-Tolić, L.; Lee, S.-W.; Li, L.; Anderson, G. A.; Harkewicz, R.; Smith, R. D. Identification of tryptic peptides from large databases using multiplexed tandem mass spectrometry: simulations and experimental results. Proteomics. 2003, 3, 1279-1286. 26. Williams, J. D.; Flanagan, M.; Lopez, L.; Fischer, S.; Miller, L. A. D. Using accurate mass electrospray ionization–time-of-flight mass spectrometry with in-source collision-induced 162 dissociation to sequence peptide mixtures. Journal of Chromatography A. 2003, 1020, 11-26. 27. Purvine, S.; Eppel*, J.-T.; Yi, E. C.; Goodlett, D. R. Shotgun collision-induced dissociation of peptides using a time of flight mass analyzer. Proteomics. 2003, 3, 847-850. 28. Venable, J. D.; Dong, M.-Q.; Wohlschlegel, J.; Dillin, A.; Yates, J. R. Automated approach for quantitative analysis of complex peptide mixtures from tandem mass spectra. Nat Meth. 2004, 1, 39-45. 29. Silva, J. C.; Denny, R.; Dorschel, C.; Gorenstein, M. V.; Li, G.-Z.; Richardson, K.; Wall, D.; Geromanos, S. J. Simultaneous qualitative and quantitative analysis of the E. coli proteome: A sweet tale. Molecular & Cellular Proteomics. 2006. 30. Silva, J. C.; Denny, R.; Dorschel, C. A.; Gorenstein, M.; Kass, I. J.; Li, G.-Z.; McKenna, T.; Nold, M. J.; Richardson, K.; Young, P.; Geromanos, S. Quantitative Proteomic Analysis by Accurate Mass Retention Time Pairs. Analytical Chemistry. 2005, 77, 2187-2200. 31. Silva, J. C.; Gorenstein, M. V.; Li, G. Z.; Vissers, J. P.; Geromanos, S. J. Absolute quantification of proteins by LCMSE: a virtue of parallel MS acquisition. Mol Cell Proteomics. 2006, 5, 144-156. 32. Hughes, M. A.; Silva, J. C.; Geromanos, S. J.; Townsend, C. A. Quantitative proteomic analysis of drug-induced changes in mycobacteria. J Proteome Res. 2006, 5, 54-63. 33. Doneanu, C. E.; Xenopoulos, A.; Fadgen, K.; Murphy, J.; Skilton, S. J.; Prentice, H.; Stapels, M.; Chen, W. Analysis of host-cell proteins in biotherapeutic proteins by comprehensive online two-dimensional liquid chromatography/mass spectrometry. MAbs. 2012, 4, 2444. 34. Finamore, F.; Pieroni, L.; Ronci, M.; Marzano, V.; Mortera, S. L.; Romano, M.; Cortese, C.; Federici, G.; Urbani, A. Proteomics investigation of human platelets by shotgun nUPLCMSE and 2DE experimental strategies: a comparative study. Blood Transfus. 2010, 8 Suppl 3, s140-148. 35. Kong, W. M.; Chik, Z.; Ramachandra, M.; Subramaniam, U.; Aziddin, R. E.; Mohamed, Z. Evaluation of the effects of Mitragyna speciosa alkaloid extract on cytochrome P450 enzymes using a high throughput assay. Molecules. 2011, 16, 7344-7356. 36. Levin, Y.; Hradetzky, E.; Bahn, S. Quantification of proteins using data-independent analysis (MSE) in simple andcomplex samples: a systematic evaluation. Proteomics. 2011, 11, 3273-3287. 37. Mbeunkui, F.; Goshe, M. B. Investigation of solubilization and digestion methods for microsomal membrane proteome analysis using data-independent LC-MSE. Proteomics. 2011, 11, 898-911. 38. Pacheco, L. G.; Slade, S. E.; Seyffert, N.; Santos, A. R.; Castro, T. L.; Silva, W. M.; Santos, A. V.; Santos, S. G.; Farias, L. M.; Carvalho, M. A.; Pimenta, A. M.; Meyer, R.; Silva, A.; Scrivens, J. H.; Oliveira, S. C.; Miyoshi, A.; Dowson, C. G.; Azevedo, V. A combined approach for comparative exoproteome analysis of Corynebacterium pseudotuberculosis. BMC Microbiol. 2011, 11, 12. 39. Pieroni, L.; Finamore, F.; Ronci, M.; Mattoscio, D.; Marzano, V.; Mortera, S. L.; Quattrucci, S.; Federici, G.; Romano, M.; Urbani, A. Proteomics investigation of human platelets in healthy donors and cystic fibrosis patients by shotgun nUPLC-MSE and 2DE: a comparative study. Mol Biosyst. 2011, 7, 630-639. 40. Ternette, N.; Wright, C.; Kramer, H. B.; Altun, M.; Kessler, B. M. Label-free quantitative proteomics reveals regulation of interferon-induced protein with tetratricopeptide repeats 163 3 (IFIT3) and 5'-3'-exoribonuclease 2 (XRN2) during respiratory syncytial virus infection. Virol J. 2011, 8, 442. 41. Yang, H. Y.; Kwon, J.; Choi, H. I.; Park, S. H.; Yang, U.; Park, H. R.; Ren, L.; Chung, K. J.; Kim, Y. U.; Park, B. J.; Jeong, S. H.; Lee, T. H. In-depth analysis of cysteine oxidation by the RBC proteome: advantage of peroxiredoxin II knockout mice. Proteomics. 2012, 12, 101-112. 42. Blackburn, K.; Mbeunkui, F.; Mitra, S. K.; Mentzel, T.; Goshe, M. B. Improving Protein and Proteome Coverage through Data-Independent Multiplexed Peptide Fragmentation. Journal of Proteome Research. 2010, 9, 3621-3637. 43. Geromanos, S. J.; Vissers, J. P. C.; Silva, J. C.; Dorschel, C. A.; Li, G.-Z.; Gorenstein, M. V.; Bateman, R. H.; Langridge, J. I. The detection, correlation, and comparison of peptide precursor and product ions from data independent LC-MS with data dependant LCMS/MS. Proteomics. 2009, 9, 1683-1695. 44. Acera, A.; Vecino, E.; Rodriguez-Agirretxe, I.; Aloria, K.; Arizmendi, J. M.; Morales, C.; Duran, J. A. Changes in tear protein profile in keratoconus disease. Eye (Lond). 2011, 25, 1225-1233. 45. Xie, H.; Doneanu, C.; Chen, W.; Rininger, J.; Mazzeo, J. R. Characterization of a recombinant influenza vaccine candidate using complementary LC-MS methods. Curr Pharm Biotechnol. 2011, 12, 1568-1579. 46. Xie, H.; Chakraborty, A.; Ahn, J.; Yu, Y. Q.; Dakshinamoorthy, D. P.; Gilar, M.; Chen, W.; Skilton, S. J.; Mazzeo, J. R. Rapid comparison of a candidate biosimilar to an innovator monoclonal antibody with advanced liquid chromatography and mass spectrometry technologies. MAbs. 2010, 2. 47. Xie, H.; Gilar, M.; Gebler, J. C. Characterization of protein impurities and site-specific modifications using peptide mapping with liquid chromatography and data independent acquisition mass spectrometry. Anal Chem. 2009, 81, 5699-5708. 48. Gilar, M.; Olivova, P.; Chakraborty, A. B.; Jaworski, A.; Geromanos, S. J.; Gebler, J. C. Comparison of 1-D and 2-D LC MS/MS methods for proteomic analysis of human serum. Electrophoresis. 2009, 30, 1157-1167. 49. Cheng, F. Y.; Blackburn, K.; Lin, Y. M.; Goshe, M. B.; Williamson, J. D. Absolute protein quantification by LC/MS(E) for global analysis of salicylic acid-induced plant protein secretion responses. J Proteome Res. 2009, 8, 82-93. 50. Schaff, J. E.; Mbeunkui, F.; Blackburn, K.; Bird, D. M.; Goshe, M. B. SILIP: a novel stable isotope labeling method for in planta quantitative proteomic analysis. Plant J. 2008, 56, 840-854. 51. Vissers, J. P.; Pons, S.; Hulin, A.; Tissier, R.; Berdeaux, A.; Connolly, J. B.; Langridge, J. I.; Geromanos, S. J.; Ghaleh, B. The use of proteome similarity for the qualitative and quantitative profiling of reperfused myocardium. J Chromatogr B Analyt Technol Biomed Life Sci. 2009, 877, 1317-1326. 52. Chakraborty, A. B.; Berger, S. J.; Gebler, J. C. Use of an integrated MS – multiplexed MS/MS data acquisition strategy for high-coverage peptide mapping studies. Rapid Communications in Mass Spectrometry. 2007, 21, 730-744. 53. Martins-de-Souza, D.; Guest, P. C.; Guest, F. L.; Bauder, C.; Rahmoune, H.; Pietsch, S.; Roeber, S.; Kretzschmar, H.; Mann, D.; Baborie, A.; Bahn, S. Characterization of the human primary visual cortex and cerebellum proteomes using shotgun mass spectrometry-data-independent analyses. Proteomics. 2012, 12, 500-504. 164 54. Geiger, T.; Cox, J.; Mann, M. Proteomics on an Orbitrap Benchtop Mass Spectrometer Using All-ion Fragmentation. Molecular & Cellular Proteomics. 2010, 9, 2252-2261. 55. Bern, M.; Finney, G.; Hoopmann, M. R.; Merrihew, G.; Toth, M. J.; MacCoss, M. J. Deconvolution of mixture spectra from ion-trap data-independent-acquisition tandem mass spectrometry. Anal Chem. 2010, 82, 833-841. 56. Hopfgartner, G.; Tonoli, D.; Varesio, E. High-resolution mass spectrometry for integrated qualitative and quantitative analysis of pharmaceuticals in biological matrices. Anal Bioanal Chem. 2012, 402, 2587-2596. 57. Gillet, L. C.; Navarro, P.; Tate, S.; Röst, H.; Selevsek, N.; Reiter, L.; Bonner, R.; Aebersold, R. Targeted Data Extraction of the MS/MS Spectra Generated by Data-independent Acquisition: A New Concept for Consistent and Accurate Proteome Analysis. Molecular & Cellular Proteomics. 2012, 11. 58. Panchaud, A.; Scherl, A.; Shaffer, S. A.; von Haller, P. D.; Kulasekara, H. D.; Miller, S. I.; Goodlett, D. R. Precursor Acquisition Independent From Ion Count: How to Dive Deeper into the Proteomics Ocean. Analytical Chemistry. 2009, 81, 6481-6488. 59. Panchaud, A.; Jung, S.; Shaffer, S. A.; Aitchison, J. D.; Goodlett, D. R. Faster, Quantitative, and Accurate Precursor Acquisition Independent From Ion Count. Analytical Chemistry. 2011, 83, 2250-2257. 60. Acosta-Martin, A. E.; Panchaud, A.; Chwastyniak, M.; Dupont, A.; Juthier, F.; Gautier, C.; Jude, B.; Amouyel, P.; Goodlett, D. R.; Pinet, F. Quantitative mass spectrometry analysis using PAcIFIC for the identification of plasma diagnostic biomarkers for abdominal aortic aneurysm. PLoS One. 2011, 6, e28698. 61. Hengel, S. M.; Murray, E.; Langdon, S.; Hayward, L.; O'Donoghue, J.; Panchaud, A.; Hupp, T.; Goodlett, D. R. Data-independent proteomic screen identifies novel tamoxifen agonist that mediates drug resistance. J Proteome Res. 2011, 10, 4567-4578. 62. Weisbrod, C. R.; Eng, J. K.; Hoopmann, M. R.; Baker, T.; Bruce, J. E. Accurate peptide fragment mass analysis: multiplexed peptide identification and quantification. J Proteome Res. 2012, 11, 1621-1632. 63. Butt, A. M. Modulation of a glial blood-brain barrier. Ann N Y Acad Sci. 1991, 633, 363-377. 64. Cebada, J.; Alvarado-Alvarez, R.; Becerra, E.; Neri-Bazan, L.; Rocha, L.; Garcia, U. An improved method for long-term measuring of hemolymph fluctuations of non-essential amino acids, GABA and histamine from freely moving crayfish. J Neurosci Meth. 2006, 153, 1-7. 65. Behrens, H. L.; Chen, R.; Li, L. Combining microdialysis, NanoLC-MS, and MALDITOF/TOF to detect neuropeptides secreted in the crab, Cancer borealis. Anal Chem. 2008, 80, 6949-6958. 66. Behrens, H. L.; Li, L. Monitoring neuropeptides in vivo via microdialysis and mass spectrometry. Methods Mol Biol. 2010, 615, 57-73. 67. Lanckmans, K.; Sarre, S.; Smolders, I.; Michotte, Y. Quantitative liquid chromatography/mass spectrometry for the analysis of microdialysates. Talanta. 2008, 74, 458-469. 68. Lee, G. J.; Park, J. H.; Park, H. K. Microdialysis applications in neuroscience. Neurol Res. 2008, 30, 661-668. 69. Ao, X.; Sellati, T. J.; Stenken, J. A. Enhanced microdialysis relative recovery of inflammatory cytokines using antibody-coated microspheres analyzed by flow cytometry. Anal Chem. 2004, 76, 3777-3784. 165 70. Ao, X.; Stenken, J. A. Water-soluble cyclodextrin polymers for enhanced relative recovery of hydrophobic analytes during microdialysis sampling. Analyst. 2003, 128, 1143-1149. 71. Ao, X.; Stenken, J. A. Microdialysis sampling of cytokines. Methods. 2006, 38, 331-341. 72. Andreis, P. G.; Malendowicz, L. K.; Rebuffat, P.; Spinazzi, R.; Ziolkowska, A.; Nussdorfer, G. G. Galanin enhances corticosterone secretion from dispersed rat adrenocortical cells through the activation of GAL-R1 and GAL-R2 receptors coupled to the adenylate cyclase-dependent signaling cascade. Int J Mol Med. 2007, 19, 149-155. 73. Ao, X.; Stenken, J. A. No delayed temporal response to sample concentration changes during enhanced microdialysis sampling using cyclodextrins and antibody-immobilized microspheres. Analyst. 2006, 131, 62-67. 74. Ao, X.; Wang, X.; Lennartz, M. R.; Loegering, D. J.; Stenken, J. A. Multiplexed cytokine detection in microliter microdialysis samples obtained from activated cultured macrophages. J Pharm Biomed Anal. 2006, 40, 915-921. 75. Duo, J.; Fletcher, H.; Stenken, J. A. Natural and synthetic affinity agents as microdialysis sampling mass transport enhancers: current progress and future perspectives. Biosens Bioelectron. 2006, 22, 449-457. 76. Duo, J.; Stenken, J. A. In vitro and in vivo affinity microdialysis sampling of cytokines using heparin-immobilized microspheres. Anal Bioanal Chem. 2011, 399, 783-793. 77. Duo, J.; Stenken, J. A. Heparin-immobilized microspheres for the capture of cytokines. Anal Bioanal Chem. 2011, 399, 773-782. 78. Fletcher, H. J.; Stenken, J. A. An in vitro comparison of microdialysis relative recovery of Met- and Leu-enkephalin using cyclodextrins and antibodies as affinity agents. Anal Chim Acta. 2008, 620, 170-175. 79. Khramov, A. N.; Stenken, J. A. Enhanced microdialysis recovery of some tricyclic antidepressants and structurally related drugs by cyclodextrin-mediated transport. Analyst. 1999, 124, 1027-1033. 80. Sun, L.; Stenken, J. A. Improving microdialysis extraction efficiency of lipophilic eicosanoids. J Pharm Biomed Anal. 2003, 33, 1059-1071. 81. Wang, Y.; Stenken, J. A. Affinity-based microdialysis sampling using heparin for in vitro collection of human cytokines. Anal Chim Acta. 2009, 651, 105-111. 82. Reed, B.; Bidlack, J. M.; Chait, B. T.; Kreek, M. J. Extracellular biotransformation of betaendorphin in rat striatum and cerebrospinal fluid. J Neuroendocrinol. 2008, 20, 606-616. 83. Haskins, W. E.; Watson, C. J.; Cellar, N. A.; Powell, D. H.; Kennedy, R. T. Discovery and neurochemical screening of peptides in brain extracellular fluid by chemical analysis of in vivo microdialysis samples. Anal Chem. 2004, 76, 5523-5533. 84. Lanckmans, K.; Stragier, B.; Sarre, S.; Smolders, I.; Michotte, Y. Nano-LC-MS/MS for the monitoring of angiotensin IV in rat brain microdialysates: limitations and possibilities. J Sep Sci. 2007, 30, 2217-2224. 85. Nyitrai, G.; Kekesi, K. A.; Emri, Z.; Szarics, E.; Juhasz, G.; Kardos, J. GABA(B) receptor antagonist CGP-36742 enhances somatostatin release in the rat hippocampus in vivo and in vitro. Eur J Pharmacol. 2003, 478, 111-119. 86. Wilson, S. R.; Boix, F.; Holm, A.; Molander, P.; Lundanes, E.; Greibrokk, T. Determination of bradykinin and arg-bradykinin in rat muscle tissue by microdialysis and capillary column-switching liquid chromatography with mass spectrometric detection. J Sep Sci. 2005, 28, 1751-1758. 166 87. Andren, P. E.; Caprioli, R. M. Determination of extracellular release of neurotensin in discrete rat brain regions utilizing in vivo microdialysis/electrospray mass spectrometry. Brain Res. 1999, 845, 123-129. 88. Emmett, M. R.; Andren, P. E.; Caprioli, R. M. Specific molecular mass detection of endogenously released neuropeptides using in vivo microdialysis/mass spectrometry. J Neurosci Methods. 1995, 62, 141-147. 89. Haskins, W. E.; Wang, Z.; Watson, C. J.; Rostand, R. R.; Witowski, S. R.; Powell, D. H.; Kennedy, R. T. Capillary LC-MS2 at the attomole level for monitoring and discovering endogenous peptides in microdialysis samples collected in vivo. Anal Chem. 2001, 73, 5005-5014. 90. Baseski, H. M.; Watson, C. J.; Cellar, N. A.; Shackman, J. G.; Kennedy, R. T. Capillary liquid chromatography with MS3 for the determination of enkephalins in microdialysis samples from the striatum of anesthetized and freely-moving rats. Journal of Mass Spectrometry. 2005, 40, 146-153. 91. Li, Q.; Zubieta, J. K.; Kennedy, R. T. Practical aspects of in vivo detection of neuropeptides by microdialysis coupled off-line to capillary LC with multistage MS. Anal Chem. 2009, 81, 2242-2250. 92. Mabrouk, O. S.; Li, Q.; Song, P.; Kennedy, R. T. Microdialysis and mass spectrometric monitoring of dopamine and enkephalins in the globus pallidus reveal reciprocal interactions that regulate movement. J Neurochem. 2011, 118, 24-33. 93. Reed, B.; Zhang, Y.; Chait, B. T.; Kreek, M. J. Dynorphin A(1-17) biotransformation in striatum of freely moving rats using microdialysis and matrix-assisted laser desorption/ionization mass spectrometry. J Neurochem. 2003, 86, 815-823. 94. Zhang, H.; Stoeckli, M.; Andren, P. E.; Caprioli, R. M. Combining solid-phase preconcentration, capillary electrophoresis and off-line matrix-assisted laser desorption/ionization mass spectrometry: intracerebral metabolic processing of peptide E in vivo. J Mass Spectrom. 1999, 34, 377-383. 95. Prokai, L.; Kim, H. S.; Zharikova, A.; Roboz, J.; Ma, L.; Deng, L.; Simonsick, W. J., Jr. Electrospray ionization mass spectrometric and liquid chromatographic-mass spectrometric studies on the metabolism of synthetic dynorphin A peptides in brain tissue in vitro and in vivo. J Chromatogr A. 1998, 800, 59-68. 96. Klintenberg, R.; Andren, P. E. Altered extracellular striatal in vivo biotransformation of the opioid neuropeptide dynorphin A(1-17) in the unilateral 6-OHDA rat model of Parkinson's disease. J Mass Spectrom. 2005, 40, 261-270. 97. Bernay, B.; Gaillard, M. C.; Guryca, V.; Emadali, A.; Kuhn, L.; Bertrand, A.; Detraz, I.; Carcenac, C.; Savasta, M.; Brouillet, E.; Garin, J.; Elalouf, J. M. Discovering new bioactive neuropeptides in the striatum secretome using in vivo microdialysis and versatile proteomics. Mol Cell Proteomics. 2009, 8, 946-958. 98. Schmerberg, C. M.; Li, L. Mass Spectrometric Detection of Neuropeptides Using AffinityEnhanced Microdialysis with Antibody-Coated Magnetic Nanoparticles Analytical Chemistry. 2012, [resubmitted]. 99. Ye, H.; Greer, T.; Li, L. From pixel to voxel: a deeper view of biological tissue by 3D mass spectral imaging. Bioanalysis. 2011, 3, 313-332. 100. Seeley, E. H.; Schwamborn, K.; Caprioli, R. M. Imaging of intact tissue sections: moving beyond the microscope. J Biol Chem. 2011, 286, 25459-25466. 167 101. Bruand, J.; Sistla, S.; Meriaux, C.; Dorrestein, P. C.; Gaasterland, T.; Ghassemian, M.; Wisztorski, M.; Fournier, I.; Salzet, M.; Macagno, E.; Bafna, V. Automated querying and identification of novel peptides using MALDI mass spectrometric imaging. J Proteome Res. 2011, 10, 1915-1928. 102. Chen, R.; Li, L. Mass spectral imaging and profiling of neuropeptides at the organ and cellular domains. Anal Bioanal Chem. 2010, 397, 3185-3193. 103. Wisztorski, M.; Croix, D.; Macagno, E.; Fournier, I.; Salzet, M. Molecular MALDI imaging: an emerging technology for neuroscience studies. Dev Neurobiol. 2008, 68, 845-858. 104. Burnum, K. E.; Frappier, S. L.; Caprioli, R. M. Matrix-assisted laser desorption/ionization imaging mass spectrometry for the investigation of proteins and peptides. Annu Rev Anal Chem (Palo Alto Calif). 2008, 1, 689-705. 105. Garden, R. W.; Sweedler, J. V. Heterogeneity within MALDI samples as revealed by mass spectrometric imaging. Anal Chem. 2000, 72, 30-36. 106. Herbert, Z.; Rauser, S.; Williams, L.; Kapan, N.; Guntner, M.; Walch, A.; Boyan, G. Developmental expression of neuromodulators in the central complex of the grasshopper Schistocerca gregaria. J Morphol. 2010, 271, 1509-1526. 107. Hofer, S.; Dircksen, H.; Tollback, P.; Homberg, U. Novel insect orcokinins: characterization and neuronal distribution in the brains of selected dicondylian insects. J Comp Neurol. 2005, 490, 57-71. 108. Kruse, R.; Sweedler, J. V. Spatial profiling invertebrate ganglia using MALDI MS. J Am Soc Mass Spectrom. 2003, 14, 752-759. 109. Verhaert, P. D.; Pinkse, M. W.; Strupat, K.; Conaway, M. C. Imaging of similar mass neuropeptides in neuronal tissue by enhanced resolution MALDI MS with an ion trap Orbitrap hybrid instrument. Methods Mol Biol. 2010, 656, 433-449. 110. Zimmerman, T. A.; Rubakhin, S. S.; Romanova, E. V.; Tucker, K. R.; Sweedler, J. V. MALDI mass spectrometric imaging using the stretched sample method to reveal neuropeptide distributions in aplysia nervous tissue. Anal Chem. 2009, 81, 9402-9409. 111. Chen, R.; Cape, S. S.; Sturm, R. M.; Li, L. Mass spectrometric imaging of neuropeptides in decapod crustacean neuronal tissues. Methods Mol Biol. 2010, 656, 451-463. 112. Chen, R.; Hui, L.; Sturm, R. M.; Li, L. Three dimensional mapping of neuropeptides and lipids in crustacean brain by mass spectral imaging. Journal of the American Society for Mass Spectrometry. 2009, 20, 1068-1077. 113. Chen, R.; Jiang, X.; Conaway, M. C.; Mohtashemi, I.; Hui, L.; Viner, R.; Li, L. Mass spectral analysis of neuropeptide expression and distribution in the nervous system of the lobster Homarus americanus. J Proteome Res. 2010, 9, 818-832. 114. DeKeyser, S. S.; Kutz-Naber, K. K.; Schmidt, J. J.; Barrett-Wilt, G. A.; Li, L. Imaging mass spectrometry of neuropeptides in decapod crustacean neuronal tissues. J Proteome Res. 2007, 6, 1782-1791. 115. Goodwin, R. J.; Pennington, S. R.; Pitt, A. R. Protein and peptides in pictures: imaging with MALDI mass spectrometry. Proteomics. 2008, 8, 3785-3800. 116. Sullivan, J. M.; Beltz, B. S. Neural pathways connecting the deutocerebrum and lateral protocerebrum in the brains of decapod crustaceans. J Comp Neurol. 2001, 441, 9-22. 117. Gutierrez, G. J.; Grashow, R. G. Cancer Borealis Stomatogastric Nervous System Dissection. J Vis Exp. 2009, e1207. 168 118. Tobin, A.-E.; Bierman, H. S. Homarus Americanus Stomatogastric Nervous System Dissection. J Vis Exp. 2009, e1171. 119. Marder, E.; Bucher, D. Understanding Circuit Dynamics Using the Stomatogastric Nervous System of Lobsters and Crabs. Annual Review of Physiology. 2007, 69, 291-316. 120. Rao, K. R. Crustacean pigment-dispersing hormones: chemistry, distribution and actions. Pigment Cell Res. 1992, Suppl 2, 266-270. 121. Rao, K. R. Crustacean Pigmentary-Effector Hormones: Chemistry and Functions of RPCH, PDH, and Related Peptides. Am Zool. 2001, 41, 364-379. 122. Chung, J. S.; Bembe, S.; Tamone, S.; Andrews, E.; Thomas, H. Molecular cloning of the crustacean hyperglycemic hormone (CHH) precursor from the X-organ and the identification of the neuropeptide from sinus gland of the Alaskan Tanner crab, Chionoecetes bairdi. Gen Comp Endocrinol. 2009, 162, 129-133. 123. Janssen, T.; Lindemans, M.; Meelkop, E.; Temmerman, L.; Schoofs, L. Coevolution of neuropeptidergic signaling systems: from worm to man. Ann N Y Acad Sci. 2010, 1200, 1-14. 124. Coast, G. M.; Schooley, D. A. Toward a consensus nomenclature for insect neuropeptides and peptide hormones. Peptides. 2011, 32, 620-631. 125. Husson, S. J.; Mertens, I.; Janssen, T.; Lindemans, M.; Schoofs, L. Neuropeptidergic signaling in the nematode Caenorhabditis elegans. Prog Neurobiol. 2007, 82, 33-55. 126. Roch, G. J.; Busby, E. R.; Sherwood, N. M. Evolution of GnRH: diving deeper. Gen Comp Endocrinol. 2011, 171, 1-16. 127. Veenstra, J. A. Neuropeptide evolution: neurohormones and neuropeptides predicted from the genomes of Capitella teleta and Helobdella robusta. Gen Comp Endocrinol. 2011, 171, 160-175. 128. Fukusumi, S.; Fujii, R.; Hinuma, S. Recent advances in mammalian RFamide peptides: The discovery and functional analyses of PrRP, RFRPs and QRFP. Peptides. 2006, 27, 10731086. 129. Clynen, E.; Liu, F.; Husson, S. J.; Landuyt, B.; Hayakawa, E.; Baggerman, G.; Wets, G.; Schoofs, L. Bioinformatic approaches to the identification of novel neuropeptide precursors. Methods Mol Biol. 2010, 615, 357-374. 130. Liu, F.; Baggerman, G.; D'Hertog, W.; Verleyen, P.; Schoofs, L.; Wets, G. In silico identification of new secretory peptide genes in Drosophila melanogaster. Mol Cell Proteomics. 2006, 5, 510-522. 131. Christie, A. E.; Cashman, C. R.; Brennan, H. R.; Ma, M.; Sousa, G. L.; Li, L.; Stemmler, E. A.; Dickinson, P. S. Identification of putative crustacean neuropeptides using in silico analyses of publicly accessible expressed sequence tags. Gen Comp Endocrinol. 2008, 156, 246-264. 132. Jarecki, J. L.; Frey, B. L.; Smith, L. M.; Stretton, A. O. Discovery of neuropeptides in the nematode Ascaris suum by database mining and tandem mass spectrometry. J Proteome Res. 2011, 10, 3098-3106. 133. Christie, A. E.; Nolan, D. H.; Ohno, P.; Hartline, N.; Lenz, P. H. Identification of chelicerate neuropeptides using bioinformatics of publicly accessible expressed sequence tags. Gen Comp Endocrinol. 2011, 170, 144-155. 134. Stemmler, E. A.; Bruns, E. A.; Cashman, C. R.; Dickinson, P. S.; Christie, A. E. Molecular and mass spectral identification of the broadly conserved decapod crustacean 169 neuropeptide pQIRYHQCYFNPISCF: the first PISCF-allatostatin (Manduca sexta- or Ctype allatostatin) from a non-insect. Gen Comp Endocrinol. 2010, 165, 1-10. 135. Gard, A. L.; Lenz, P. H.; Shaw, J. R.; Christie, A. E. Identification of putative peptide paracrines/hormones in the water flea Daphnia pulex (Crustacea; Branchiopoda; Cladocera) using transcriptomics and immunohistochemistry. Gen Comp Endocrinol. 2009, 160, 271-287. 136. Dickinson, P. S.; Stemmler, E. A.; Cashman, C. R.; Brennan, H. R.; Dennison, B.; Huber, K. E.; Peguero, B.; Rabacal, W.; Goiney, C. C.; Smith, C. M.; Towle, D. W.; Christie, A. E. SIFamide peptides in clawed lobsters and freshwater crayfish (Crustacea, Decapoda, Astacidea): a combined molecular, mass spectrometric and electrophysiological investigation. Gen Comp Endocrinol. 2008, 156, 347-360. 137. Ma, M.; Gard, A. L.; Xiang, F.; Wang, J.; Davoodian, N.; Lenz, P. H.; Malecha, S. R.; Christie, A. E.; Li, L. Combining in silico transcriptome mining and biological mass spectrometry for neuropeptide discovery in the Pacific white shrimp Litopenaeus vannamei. Peptides. 2010, 31, 27-43. 138. Christie, A. E. In silico analyses of peptide paracrines/hormones in Aphidoidea. Gen Comp Endocrinol. 2008, 159, 67-79. 139. Christie, A. E. Neuropeptide discovery in Ixodoidea: an in silico investigation using publicly accessible expressed sequence tags. Gen Comp Endocrinol. 2008, 157, 174-185. 140. Christie, A. E.; Stevens, J. S.; Bowers, M. R.; Chapline, M. C.; Jensen, D. A.; Schegg, K. M.; Goldwaser, J.; Kwiatkowski, M. A.; Pleasant, T. K., Jr.; Shoenfeld, L.; Tempest, L. K.; Williams, C. R.; Wiwatpanit, T.; Smith, C. M.; Beale, K. M.; Towle, D. W.; Schooley, D. A.; Dickinson, P. S. Identification of a calcitonin-like diuretic hormone that functions as an intrinsic modulator of the American lobster, Homarus americanus, cardiac neuromuscular system. J Exp Biol. 2010, 213, 118-127. 141. Lovejoy, D. A.; Barsyte-Lovejoy, D. Characterization of a corticotropin-releasing factor (CRF)/diuretic hormone-like peptide from tunicates: insight into the origins of the vertebrate CRF family. Gen Comp Endocrinol. 2010, 165, 330-336. 142. Montagne, N.; Desdevises, Y.; Soyez, D.; Toullec, J. Y. Molecular evolution of the crustacean hyperglycemic hormone family in ecdysozoans. BMC Evol Biol. 2010, 10, 62. 143. Yasuda, A.; Yasuda-Kamatani, Y.; Nozaki, M.; Nakajima, T. Identification of GYRKPPFNGSIFamide (crustacean-SIFamide) in the crayfish Procambarus clarkii by topological mass spectrometry analysis. Gen Comp Endocrinol. 2004, 135, 391-400. 144. Fu, Q.; Goy, M. F.; Li, L. Identification of neuropeptides from the decapod crustacean sinus glands using nanoscale liquid chromatography tandem mass spectrometry. Biochem Biophys Res Commun. 2005, 337, 765-778. 145. Ma, M.; Szabo, T. M.; Jia, C.; Marder, E.; Li, L. Mass spectrometric characterization and physiological actions of novel crustacean C-type allatostatins. Peptides. 2009, 30, 16601668. 146. Schmerberg, C. M.; Li, L. Function-Driven Discovery of Neuropeptides with Mass Spectrometry-Based Tools. Protein Pept Lett. 2012. 170 3.8. Figures Figure 3.1. Schematic representing the different types of MS-based tools that can be used for function-driven NP discovery, by providing quantitative information about different samples. In this schematic, samples are taken from animals either before and after an experimental manipulation, or several times during the manipulation. Each sample is characterized with a distinct color. MS and MS/MS-based techniques, both those that incorporate isotopic labels and those that do not, are illustrated in the chart below. Further details of these techniques and explanations of abbreviations are found in the text. From [146] Copyright 2012 Bentham Science Publishers. 171 Figure 3.2. The role of function-driven NP discovery in the iterative cycle of neuropeptide identity and function determination. A number of techniques exist to determine NP function that require prior identification of the compounds, which was previously done with methods devoid of input from functional studies. Function-driven MS discovery permits discovery of functionally-relevant NPs via comparative peptidomic analysis. The putative functions thus identified can be validated with other tools. Functions previously determined with less specific analytical tools can also be verified and the exact identities of functionally-relevant compounds determined. This permits cyclical study of NPs to better understand their identities and functions. From [146] Copyright 2012 Bentham Science Publishers. 172 Figure 3.3. A) Workflow for differential display (DD) of NPs with expression changed after feeding in C. borealis. B) Representative MALDI-TOF/TOF mass spectrum of labeled mixture of brain extracts from fed and unfed crabs. The heavy labeled peaks (fed crabs, n=3) are indicated with open circles, and the light labeled peaks are indicated with closed circles (unfed crabs, n=3). Several abundant neuropeptides are indicated and labeled with their corresponding amino acid sequences. Adapted with permission from Chen, R., Hui, L., Cape, S. S., Wang, J, and Li, L. ACS Chem. Neurosci. 2010, 1 (3), pp 2-4-214. Copyright 2009 American Chemical Society. (Ref. [10]) From [146] Copyright 2012 Bentham Science Publishers. 173 Figure 3.4. I) schematic illustrating MALDI imaging workflow. Adapted with permission from Chen, R., Hui, L., Cape, S. S., Wang, J, and Li, L. ACS Chem. Neurosci. 2010, 1 (3), pp 2-4-214. Copyright 2009 American Chemical Society (Ref. [10]). II) MALDI imaging of neuropeptide localization in the H. americanus brain using both MALDI TOF/TOF and MALDI LTQ Orbitrap mass spectrometers. (a) Schematic drawing of the lobster brain, which contains multiple neuropils, including anterior (AMPN) and posterior medial protocerebral neuropils (PMPN), olfactory lobe (ON), accessory lobe (AL), antenna I neuropil (AnN) and lateral II antenna neuropil (LAN). Ion images of (b) VYRKPPFNGSIFamide (m/z 1423.8) and (c) NFDEIDRSGFGFN (m/z 1517.7) were obtained using MALDI TOF/TOF. Ion images of multiple known neuropeptides were acquired by MALDI LTQ Orbitrap, including: (d) VYRKPPFNGSIFamide (m/z 1423.8); (e) APSGFLGMRamide (m/z 934.5); (f) NFDEIDRSGFGFN (m/z 1517.7); and (g) overlay of the above three neuropeptides. A novel peptide HI/LASLYKPR (m/z 1084.6) in the lobster brain was also mapped using MALDI LTQ Orbitrap instrument by (h) precursor ion scanning of m/z 1084.6 and (i) selected reaction monitoring of transition between m/z 1084.6 and sequence-specific fragment ion m/z 685.4 (b6). Adapted with permission from Chen, R., Jiang, X., Conaway, M. C. P., Mohtashemi, I., Hui, L, Viner, R., and Li, L. J. Proteome Res.. 2010, 9 (2), pp 818-832. Copyright 2009 American Chemical Society (Ref. [113]). From [146] Copyright 2012 Bentham Science Publishers. 174 175 Chapter 4. Mass Spectrometric Detection of Neuropeptides Using Affinity-Enhanced Microdialysis with Antibody-Coated Magnetic Nanoparticles Adapted from Schmerberg, C. M. and Li, L. Anal. Chem. [resubmitted] 4.1. Abstract Microdialysis (MD) is a useful sampling tool for many applications due to its ability to permit sampling from an animal concurrent with normal activity. MD is of particular importance in the field of neuroscience, in which it is used to sample neurotransmitters (NTs) while the animal is behaving in order to correlate dynamic changes in NTs with behavior. One important class of signaling molecules, the neuropeptides (NPs), however, presented significant challenges when studied with MD, due to the low relative recovery (RR) of NPs by this technique. Affinityenhanced microdialysis (AE-MD) has previously been used to improve recovery of NPs and similar molecules. For AE-MD, an affinity agent (AA), such as an antibody-coated particle or free antibody, is added to the liquid perfusing the MD probe. This AA provides an additional mass transport driving force for analyte to pass through the dialysis membrane, and thus increases the RR. In this work, a variety of AAs have been investigated for AE-MD of NPs in vitro and in vivo, including particles with C18 surface functionality and antibody-coated particles. Antibody-coated magnetic nanoparticles (AbMnP) provided the best RR enhancement in vitro, with statistically significant (p<0.05) enhancements for 4 out of 6 NP standards tested, and RR increases up to 41-fold. These particles were then used for in vivo MD in the Jonah crab, Cancer borealis, during a feeding study, with mass spectrometric (MS) detection. 31 NPs were detected in a 30 min sample, compared to 17 when no AA was used. The use of AbMnP also increased the temporal resolution from 4-18 hrs in previous studies to just 30 min in this study. The levels of NPs detected were also sufficient for reliable quantitation with the MS system in 176 use, permitting quantitative analysis of the concentration changes for 7 identified NPs on a 30 min time course during feeding. 4.2. Introduction Microdialysis (MD) is a sampling technique that allows collection of signaling molecules from an animal while it is alert and behaving, with minimal disturbance to the animal. In this technique, a MD probe is implanted into the tissue of interest and perfused with liquid at a flow rate in the range of 0.1-10 µL/min. The tip of this MD probe consists of a dialysis membrane, having pores with a defined molecular weight cutoff (MWCO). Molecules below this MWCO near the tip of the probe passively diffuse into the probe and are then carried by the slowlymoving liquid out of the probe, through a length of tubing, and finally to a sample collection vial or analysis system. This technique has been used successfully to collect a variety of different molecules from a number of tissues in several species, and has provided important insights into the action of compounds in vivo in a minimally perturbed animal [1, 2]. MD is of great utility in neuroscience, in which time-resolved changes in neurochemistry during the performance of a behavior or exposure to a stimulus are of interest. Continual collection of neurochemicals without disturbing the animal to obtain the samples allows the experimenter to determine the molecular underpinnings of neuronal activity related to these events, in the absence of any sampling-induced neuronal changes. MD has been used successfully to monitor small molecule neurotransmitter (NT) changes in vertebrate animals under a variety of different conditions, and has contributed greatly to our understanding of the effects of NT release on behavior [1, 3-5]. One area that is particularly challenging for MD sampling is the analysis of larger molecules, such as neuropeptides (NPs), which are below the MWCO of the probe but are in the mass range of 500-10,000 [5-9]. A number of complex factors make recovery of NPs difficult. 177 One such reason is the lower relative recovery (RR) of NPs in comparison to small molecules due to their larger size hindering passage through the dialysis membrane. The RR is calculated by taking the concentration of an analyte collected through MD divided by the concentration outside the probe, and is usually expressed as a percentage. This RR is inversely related to the mass of the molecule in a nonlinear manner, with larger molecules typically having RRs of less than 50%. Another challenge is the low endogenous concentration of these compounds. NPs are present in vivo at the nM - pM concentration range [2]. Therefore, the concentration collected, governed by the laws of passive diffusion, is reduced compared to NTs due not only to their low endogenous concentrations but also their reduced RR. Furthermore, RR is also governed by the amount of time the liquid is in contact with the membrane (the MD flow rate, FR), with lower flow rates leading to greater RRs.[2, 8, 10] If increased amounts of analyte are desired, a longer collection time can be employed. If a short collection time is desired, an experiment will detect NPs reliably only if the RR is improved by other means,[7] or a more sensitive detection technique is employed. Progress has been made in using highly sensitive and specific detection methods for NPs in samples obtained via microdialysis, relying mainly on mass spectrometry (MS) [1, 3, 4, 1113]. Some of these studies use MS for surveys of NP content and identity [14-23]. Other studies use MS for quantitation of identified NPs in microdialysate, mostly with selected reaction monitoring (SRM) of daughter or granddaughter ions [2, 10, 24-30]. Finally, microdialysates can be analyzed via MS-based techniques for NP discovery combined with less precise quantification methods commonly used in proteomics [31-36]. In addition to MS-based analysis of dialysates, other sensitive techniques, including those that rely on immunochemical or spectrophotometric detection, have been used for quantitation of NPs in dialysates, but these methods lack specificity [1, 3, 13]. Although MS instruments are highly sensitive, not all perform adequately in the 178 concentration range at which NPs are present in vivo, and other methods to increase sensitivity must be investigated. An important method to increase the sensitivity of NP detection in MD is to increase the RR. The relative recovery can be increased by adding affinity agents (AAs) to the liquid perfusing the probe. The analytes form interactions with the AAs, thus increasing the quasiequilibrium driving force for further analyte to diffuse into the probe [7]. This technique is termed affinity-enhanced microdialysis (AE-MD), and has been used by a number of different researchers with a wide variety of compounds [5-7, 17, 34, 35, 37-52]. Stenken and colleagues, among others, have achieved success in improving the recovery of cytokines in vitro and in vivo [7, 39, 51, 53, 54] and neuropeptides in vitro [43], using free antibody, cyclodextrins, and micron-sized beads coated with antibodies or heparin. AE-MD is not yet optimal, as saturation of the beads can occur, leading to non-linear recovery enhancement. Clogging or settling of the beads in solution is also a major concern. The technique has also not yet been applied to study NPs in vivo (although the cytokine CCL2 has been studied in rats using AE-MD [54]), nor have smaller beads been employed as affinity agents. In this work, several AAs are tested for enhancement of NP recovery. Nanoscale magnetic beads are developed for use as AAs, with the advantages of reduced settling rate and greater binding capacity. They enhance recovery of 4 out of 6 NP standards tested in vitro. They are also employed in vivo to study the time course of NP release following feeding in the Jonah crab, Cancer borealis. This new affinity agent for AE-MD greatly increases the utility of this technique for monitoring peptide secretion during behavior. 4.3. Materials and Methods More detail for this section can be found in Appendix A. 4.3.1. Reagents 179 Peptide standards (bradykinin (BK), somatostatin-14 (SMT), substance P (SP), Homarus americanus FMRFamide-like peptide I (FLP I), H. americanus FMRFamide-like peptide II (FLP II), and FMRFamide) were purchased from American Peptide Co. (Sunnyvale, CA, USA) and used without further purification. C18 silica microparticles (C18SµP) were purchased from Varian (now Agilent Technologies, Santa Clara, CA, USA) and were 5µm in size with 300Å pore size. They were used in perfusate at a concentration of 0.2mg/mL, or 3.04 x 103 beads/µL. C18 magnetic microparticles of 1µm diameter (C18MµP) were purchased from Varian at a stock concentration of 2 x 106 beads/µL and used in perfusate at 3.3 x 104 beads/µL Magnetic microparticles pre-coated with protein G were purchased from New England Biolabs (Ipswich, MA, USA), with a stock concentration of 3.11 x 104 beads/µL and a final perfusate concentration of 518 beads/µL. Magnetic nanoparticles of 100nm diameter pre-coated with protein G were purchased from Chemicell GmbH (Berlin, Germany) with a stock concentration of 1.8 x 1010 beads/µL, and thus perfusate concentrations of 3.0 x 108 beads/µL and 1.8 x 109 beads/µL as indicated below. Bovine serum albumin (BSA) and formic acid (FA) were purchased from Sigma-Aldrich (St. Louis, MO, USA). Polyclonal rabbit anti-FMRFa antibody was purchased from Abcam (Cambridge, MA, USA). All other chemicals were purchased from Fisher Scientific (Pittsburgh, PA, USA) at ACS reagent-grade and used without further purification. ACS reagentgrade solvents and Milli-Q water were used for sample preparation. Optima grade solvents were used for operation of the UPLC-QTOF. C18-coated magnetic beads and antibody-linked beads were prepared and used as recommended by the manufacturers. Details of preparation, unbinding, and in vitro bead binding assays can be found in the supplementary information. 4.3.2. Animals Jonah crabs (Cancer borealis) were purchased from Ocean Resources, Inc. (Sedgwick, ME, USA) and The Fresh Lobster Company (Gloucester, MA, USA). These crabs were wild- 180 caught and shipped overnight packed on ice. The crabs were then maintained in an artificial seawater tank at 10-12°C, with crushed gravel as a substrate. The water from two 55 gal and one 40 gal tanks was combined and continually recirculated through a filtration apparatus (total system volume ~160 gal) with a protein skimmer and water chiller. The tank system was modified from a system purchased previously from Aquatic Eco-Systems, Inc. (Apopka, FL, USA). Levels of salinity, dissolved oxygen, nitrate, nitrite, pH, and ammonia were checked regularly and adjusted with commercial aquarium maintenance products or by adjusting filtration settings. The room was kept on a 12hr/12hr dark/light schedule, with the dark photoperiod from 10AM to 10PM so animals could be observed during their normal active period. 4.3.3. Microdialysis Supplies CMA/20 Elite probes with 4 mm membranes of polyarylether sulfone (PAES) were purchased from CMA Microdialysis (Harvard Apparatus, Holliston, MA, USA). All MD probes were rinsed with water prior to use. Several pumps were used, including a CMA/102, a KD Scientific 100 (KD Scientific Inc., Holliston, MA, USA), and a Harvard 22 (Harvard Apparatus, Holliston, MA, USA). When required, additional FEP (CMA) or PEEK (Upchurch-Scientific, Idex Health and Science, Oak Harbor, WA, USA) tubing was connected to the tubing of the probe by flanged connectors from CMA, and BASi (West Lafayette, IN, USA). BD (Franklin Lakes, NJ, USA) plastic syringes were typically used. Flanged connectors were used to connect 21 gauge Luer-lock needles (included with CMA 20 series probes) blunted by grinding with a rotary tool (Dremel, Robert Bosch LLC, Farmington Hills, MI, USA) to the probe tubing. 4.3.4. In Vitro MD Experiments For in vitro experiments, the tip of the probe was immersed into a vial with a home-built apparatus to hold the probe in place. Typically, 3 different probes’ tips were immersed in the solution in the vial concurrently to provide multiple experimental replicates. The vial contained 181 microdialysis medium, which consisted of a phosphate buffered saline (PBS) solution with neuropeptide standards of interest dissolved in it at known concentrations, in the 1-5 micromolar range, except for C18 silica particles, which used 50µM. The vial with probe holder was placed on an orbital rotating platform to produce constant mixing. A sample of the medium was taken prior to starting microdialysis and following the experiment. The probe was allowed to equilibrate at the flow rate of the experiment (0.5 µL/min) for 30 min before starting dialysate collection. Technical replicates were taken as consecutive 30 min samples of the liquid flowing out of the tubing. Samples from the medium taken before and after the experiment were used to determine the relative recovery percentage. A minimum of three technical replicates were obtained per experiment, and a minimum of three experimental replicates were obtained, each coming from either a different probe or a different instance of setting up and conducting the experiment. Medium samples and samples containing no AA were placed immediately in a 96well sample plate for UPLC-QTOF analysis. For AE-MD, NPs were unbound from AAs as recommended by the manufacturer and combined with the liquid portion of the sample in a 96well plate for analysis. The percent of beads passing through the probe was determined by counting on a hemacytometer for micron-sized beads, and by comparing the dry mass of particles for nanoscale beads. 4.3.4.1. Preparation and use of Magnetic Beads C18 silica microparticles (C18SµP) were dissolved in water with 0.5% BSA to aid solubility and 0.1% FA to improve NP binding to the beads. Following MD, the beads were separated from the solution by employing paper filter spin cups that fit into 1.5mL tubes. The solution containing the solid particles is loaded into the cup, which is inserted into a tube. The tube is then centrifuged, upon which the liquid flows through the filter into the tube, and the solid particles are retained. For MD experiments, the flow-through was saved and added to the 182 liquid recovered from washing the beads in order to collect all analytes present in the dialysate. Analyte was eluted from the C18SµP by incubating them with a solution of 50/50/0.1 ACN/water/formic acid (FA) and centrifuging. The flow-through was recombined with the initial flow-through and solvent evaporated to dryness with a SpeedVac (Savant SC 110, Thermo). It was then resuspended in 0.1% FA and placed in UPLC vials for further analysis. A similar procedure was used for C18 magnetic microparticles, with the exceptions of using a magnet instead of a spin cup for separating solid particles and not adding BSA to the perfusate. All magnetic beads in this study were separated from the solution by holding a neodymium magnet to the side of the vial for 30 sec followed by pipetting out the liquid. Protein G magnetic microbeads were linked to anti-FMRFa antibody following a procedure similar to that presented by the manufacturer. Briefly, the beads were rinsed several times with a phosphate buffered saline (PBS) solution and incubated with shaking at 4º C overnight with the antibody at a dilution of 1:500 (ratio of 50µL beads: 1µL of a 1:10 dilution of serum). The beads were then again rinsed and stored in PBS. These are termed antibody-coated magnetic microparticles, abbreviated AbMµP. For in vitro assessments of bead-binding efficiency, 30µL of a solution containing neuropeptide standards in the 10-5-10-6M range was incubated with 30µL of beads for 30 min. The beads were then washed with PBS to remove unbound compounds, and this flowthrough was saved for analysis. Unbinding of the analytes from the beads was achieved by incubating them with 2% formic acid (FA) at room temperature for 10 min. Protein G magnetic nanobeads were prepared in much the same way as AbMµP, with the exception of shorter incubation times for all steps and a second concentration for antibody-bead incubation, as recommended by the manufacturer. The same antibody concentration used with the microbeads was employed, 1:500, and these are termed antibody-coated magnetic nanoparticles (AbMnP). In addition, an antibody concentration ten times higher (1:50) was 183 employed in vivo and in bead-binding assays, as this was the appropriate antibody concentration indicated by the nanobead manufacturer. Incubation with the antibody was conducted for 15 min at room temperature, and incubation with the test solution for in vitro binding assays was carried out for 15 min. The unbinding step involved 2 min incubation at room temperature with 2% FA in water. C18 magnetic microparticles (C18MµP) were washed with PBS prior to use. Elution was conducted by incubating the beads with a solution of 50/50/0.1 ACN/water/FA for 2 min at room temperature. For desalting physiological samples, the solution containing NPs was dissolved in 0.1% FA and incubated with the beads for 2 min at room temperature. They were then washed 3 times with 0.1% FA. The NPs were eluted from the beads twice with the 50/50/0.1 ACN/water/FA solution into UPLC vials, and additional water (to reduce the strength of the injection solvent) and a known amount of bradykinin was added. 4.3.4.2. Affinity-Enhanced MD In Vitro When affinity agent was used, a clean steel ball bearing of appropriate size (1/8 inch, Wheels Manufacturing, Louisville, CO, USA) was added inside the barrel of the syringe delivering microdialysate. The pump was placed into a rocking platform shaker with the syringe placed at an angle to the axis of rotation of the shaker. The rolling of the ball bearing inside the syringe kept the affinity agent in solution [55]. For the affinity agent perfusate, the equivalent of 50µL of bead solution was diluted to 3mL with PBS (a dilution of 1:60), with one exception. Although the concentrations of beads in mg/mL varied, it was determined that they had equal activity per mL, as the manufacturers’ protocols recommended the same ratio of beads to sample, i.e. 50µL beads with 0.5 mL cell lysate, a 1:10 ratio. In one set of experiments, a higher concentration of affinity agent was used, as it was possible to increase bead concentration 184 without adverse experimental effects. This trial is noted as 6x AbMnP, containing six times as many nanoparticles per unit volume (50µL of bead solution diluted to 0.5mL of perfusate). 4.3.5. In Vivo Microdialysis The procedure for implantation of a MD probe was adapted from previous publications [14, 15]. A Jonah crab was anesthetized for 15 min on ice. The crab’s shell in the area immediately above the pericardial sinus was washed with 30% bleach followed by ethanol. It was then scored in diagonal lines using a grinding wheel on a rotary tool. A small amount of hot glue was applied to the MD probe to mark the deepest point to which the probe would be inserted. A 1/32” hole was drilled in a location estimated to be immediately above the heart, and the probe was inserted. Loctite super glue gel (Loctite, Westlake, OH, USA) was applied in a small circle around the probe. Mighty Putty (epoxy materials suspended in a clay-like base, Mighty Putty, North Wales, PA, USA) was applied on top of this glue circle and then formed into a cone shape around the exposed part of the probe. Any remaining voids were filled with super glue gel. The crab was kept on ice until the glue was no longer pliable (10-15 min) and then replaced in the tank. The day of surgery was designated as day 1 and further experiments were conducted following a surgical recovery period of 2 days. 4.3.5.1. In Vivo MD feeding study The probe was surgically implanted in the crab 2 days prior to the first feeding experiment, and the last feeding experiment was conducted 8 days after surgery. This time window was chosen to avoid effects from surgery (stress of anesthesia and being out of water, trauma to the hypodermis) and tissue growth over the probe’s active membrane. Artificial crab saline (440 mM NaCl;11 mM KCl; 13 mM CaCl2; 26 mM MgCl2; 10 mM HEPES acid, pH 7.4) was used as the basis for perfusate. The flow rate was 0.5 µL/min, supplied by a programmable syringe pump (KD Scientific Model 100, Holliston, MA, USA) and samples were collected 185 every 30 min with a refrigerated fraction collector (BASi Honeycomb, Bioanalytical Systems, Inc. Indianapolis, IN, USA). The dead volume of the system from the probe tip to the collection tube was estimated to be 30µL, equal to 60 min at our flow rate. Flow was initiated through the system 30 minutes before feeding the animal. The first 30 min fraction was discarded as it was collected while the probe was not yet in equilibrium with its surroundings. Collection of feedingrelated samples was begun and the animal was fed. Samples were collected every 30 min up to 180 min post-feeding. The first two samples, taken before the animal was presented with food, were designated as baseline samples. Typical MD was conducted on days 3, 5, and 6, and AEMD using AbMnP was conducted on day 8. For AE-MD in vivo, a 1:10 dilution of nanobeads was used (equal to the high AbMnP concentration for in vitro AE-MD studies). Upon collection, 1.5 µL of formic acid was added to each sample to improve NP stability [2] and unbind NPs from the antibody-coated nanoparticles, and an internal standard (bradykinin, 1µM) was added for quantitation. Samples collected without affinity agent were directly injected onto the UPLCMS system, and magnetic beads were removed from AE-MD samples prior to addition of internal standard and MS analysis. 4.3.6. UPLC-MS and UPLC-MS/MS Analysis and Data Processing In vitro MD samples were analyzed via a UPLC-MS approach. A Waters nanoAcquity UPLC system (Waters, Millford, MA, USA) was used in conjunction with a home-packed capillary column (360 µm OD, 75 µm ID, 10cm long, Magic C18 particles (Michrom, Auburn, CA, USA), 3 µm diameter, 100Å pore size) with integrated laser-pulled (approx. 7 µm diameter, with a Sutter Instruments P-2000 (Novato, CA, USA)) ESI emitter tip. This was coupled directly to a Micromass (currently Waters) QTOF micro with capillary voltage of 3500 V run in MS only mode over the mass range m/z 500-700. Medium samples flanked dialysate samples. Table 5.1 186 enumerates the retention times of the peptides using a reverse phase separation (H2O/ACN/0.1% formic acid) with gradient from 95% aqueous to 95% organic over 30 min. For quantitation, QuanLynx software (Waters) was employed. The extracted ion chromatogram (XIC) for the dominant peak of each of the 6 NPs was generated and the area under the curve was calculated. These were all divided by the total ion current (TIC) value for that chromatogram to normalize for ion response variations throughout the run. Percent recovery was calculated as the corrected area divided by the average value obtained from the two medium samples. Statistical significance was determined using JMP statistical software (Version 9.0.2 SAS Institute, Inc., Cary, NC, USA). For data obtained using C18 silica microparticles, a student’s t-test was conducted. For the remaining data, one-way analysis of variance (ANOVA) tests were conducted, followed by post-hoc Tukey-Kramer honestly significant difference (HSD) tests with α=0.05. Means, standard errors, and p-values are reported. Graphs illustrating this data were generated in Microsoft Excel 2010 (Microsoft Corporation, Redmond, WA, USA). The linear quantitation range of the UPLC-MS system in use was also determined by creating a series of samples containing FLP I and FLP II at a range of concentrations with a constant amount of BK. These samples were analyzed via the same method and linear regrssions were generated in Microsoft Excel. In vivo MD samples were analyzed using the same instrumentation platform but with different MS and LC specifications. The LC gradient was 60 min long and a larger MS window was monitored for quantitative analyses. For in vivo MD samples, quantitation was also done with QuanLynx software. QuanLynx method files were created that would quantify multiple charge states of the NPs in C. borealis that have been previously found to change upon feeding or previously detected in C. borealis microdialysate. A similar method was followed by these QuanLynx files: generation of the XICs followed by integration. The values were not normalized 187 to TIC, however. The mass spectra were also checked to confirm mass error of less than 150 ppm from the predicted sequence. Those peaks that were present in very low abundance (area less than 5 counts) were not quantified. A peak had to be present in all time points with sufficient abundance for quantitation. Areas for detected peaks were divided by the area for bradykinin in that run. These values were then divided by the value at t=-15min to yield fold-changes. 4.4. Results and Discussion 4.4.1. In vitro recovery enhancement Due to previous work using column packing materials as AAs [34, 35, 38, 39, 43, 45, 49], initial experiments employed C18 silica microparticles as a generic, easily obtained AA (Figure 4.1). These experiments led to a modest increase in NP recovery, but problems in their implementation, including bead settling/clogging (only ~25% pass through the probe and tubing) and the need to use additives (BSA) to improve bead dispersion made them impractical for in vivo use. In order to obtain non-specific affinity enhancement of NPs, another type of particles that had C18 surface functionality but also magnetic cores for simplified sample handling and other surface modifications for increased water solubility were employed, C18 magnetic microparticles (C18MµP). These 1 µm diameter particles are commonly employed for removal of salts from biological samples prior to analyses that are sensitive to salt, such as mass spectrometry. Results obtained using C18MµP as affinity agents are presented in Fig. 4.2 and Table 4.2. The C18MµP significantly enhanced the recovery of 4 peptides—FLP I, FLP II, SP, and SMT. Recovery was at least doubled, with final RRs of several compounds at 50% or higher. However, settling and clogging were still observed due in part to the propensity of the particles to attract each other via magnetism. The settling observed was less than the C18 silica particles due to surface modifications of these particles for improved aqueous solubility. 188 Based on previous work employing antibody-coated microspheres [6, 7, 37, 39, 41-43] to improve the recovery of cytokines and neuropeptides, antibody-coated microparticles were also employed for AE-MD. Commercial magnetic immunoprecipitation (IP) kits and a commercially available anti-FMRFa antibody were used to create antibody-coated beads. Traditional agarose bead-based IP kits contain beads of ~140 µm diameter, which is unsuitable for passage through the probe and tubing. Magnetic IP kits employ smaller beads (diameters 1-5 µm), and have the advantage of simpler separation. Magnetic microparticles coated with protein G were linked to rabbit polyclonal anti-FMRFa as recommended by the manufacturer to create antibody-coated magnetic microparticles (AbMµP) and added to the perfusate. The relative recoveries obtained with and without AbMµP in the perfusate are enumerated in Fig. 4.3 and Table 4.2. No significant increases in RR were obtained for the NP standards, with the exception of SMT, whose RR was enhanced significantly (p=0.002) by about 2.5-fold. These results do not mirror findings of in vitro bead-binding assays (Figure 4.4), in which SMT bound poorly to AbMµP and other NPs had a high degree of binding. Bead settling and tube clogging were observed to a similar extent as observed with the magnetic C18 particles, and this could explain these contrary results—beads with bound NPs may have remained stuck in the tubing. Approximately half of the AbMµP passed through the probe and tubing. Magnetic nanoparticles of 100 nm diameter were the final affinity agent tested in this study. These particles are also coated with protein G and were conjugated to the same antiFMRFamide antibody (AbMnP). Particles of this size do not have permanent magnetic fields and thus are not attracted to each other in the absence of an external magnetic field [56]. This greatly reduces settling of the beads in the syringe and tubing. The smaller size of these particles also reduces settling, and it was observed that around 100% of the particles pass through the tubing. Two different concentrations of nanoparticles were used; one that was equal to that used for the 189 magnetic microparticles and C18 beads, and one that was 6 times concentrated (6x AbMnP), possible due to the reduced settling and lack of magnetic interaction between nanoparticles. RR enhancements are shown in Fig. 4.5 and Table 4.2. Statistically significant (p<0.05) recovery enhancements were obtained with AbMnP for 3 of the 6 NPs. When the concentration of nanobeads was increased, 4 out of 6 NPs had significantly enhanced RRs. FMRFa showed a strong trend toward enhanced recovery, but this trend did not meet the statistical significance threshold. The recovery of bradykinin was not enhanced by any of the affinity agents, due likely to its small, hydrophilic character and lack of an amidated C-terminus. From the data obtained here for several NPs with different sequences, it is fair to assume that the antibody used primarily recognizes C-terminal amidation, followed by hydrophobic and basic amino acids. As a side note, the promiscuity of this antibody provides additional support for the use of mass spectrometry as an unbiased technique for NP analysis. One concern that thus becomes important due to non-specific binding to antibodies is saturation of the beads’ binding capacity. High-concentration components of a complex biological sample (or fragments thereof), such as albumin in mammals and cyanin proteins in crustaceans, could fully occupy these sites in vivo, and thus binding of low-concentration biological molecules of interest to the beads will be non-linear and unreliable for accurate representation of in vivo concentration changes. Thus, antibody-linked beads in microdialysis perfusate should be used with caution when attempting to accurately determine the concentration changes of analytes in the extracellular environment. In addition, if a compound is to be delivered to an animal via MD, it may bind to the affinity agent and reduce the overall dose of the compound delivered. 4.4.2. In vivo recovery enhancement 190 Several proof-of-principle tests to determine the suitability of affinity-enhanced microdialysis for in vivo application were conducted. Jonah crabs (Cancer borealis) were implanted with microdialysis probes following a modified version of a published technique [14]. The probes of several crabs were perfused with crab saline or antibody-linked nanobeads in crab saline solution. Representative data for baseline NP content in microdialysis samples obtained with and without AbMnP in the perfusate is shown in Fig. 4.6. Here, extracted ion chromatograms (XICs) for two FMRFamide-like peptides in samples obtained under baseline conditions with and without AbMnP are displayed. These samples were obtained from the same crab on different days, with the AbMnP experiment conducted on the last day, so probe fouling is not a factor. They were also analyzed on the same day after storage under high acid conditions, which have been shown to stabilize MD samples [2]. XICs are plotted with the same y-axis scale after smoothing and baseline subtraction. FLP peaks from samples obtained without AbMnP have intensities that are 20 to 40% as intense as those obtained with AbMnP. Only very slight retention time shifts are observed, likely due to changes in ambient temperature. The UPLC column was kept at room temperature, which varies several degrees throughout the day. A similar qualitative enhancement of peak intensity was observed in two other AE-MD experiments, conducted on different crabs, the results of which are not presented here. Quantitative analysis also indicates that AbMnP improve NP recovery in vivo. Table 4.3 enumerates NPs previously detected in MD and whether these compounds were also detected in samples obtained during a feeding experiment with or without AbMnP in the perfusate. Compared with previous work in which only 35 NPs were detected in samples collected over 418 hours and concentrated ~100-fold prior to analysis [14], the detection of 31 NPs in a sample collected over only 30 min and analyzed without preconcentration is a great enhancement in 191 sensitivity. Microdialysate from the same crab obtained with simple crab saline as perfusate only contained an average of 25 NPs, with 17 of those detected in all three feeding trials. Therefore, the increased NP detection sensitivity is due not only to enhancements in UPLC-MS sensitivity that have occurred since the previous work was published, but also to the affinity-enhanced recovery by AbMnPs. For many trials without affinity agent in the perfusate, it was possible to detect NPs with reasonable reproducibility across trials, but their abundance was so low that reliable quantity changes could not be observed. In other words, the NPs were present at their lower limit of detection (LLOD), which is below their lower limit of quantification (LLOQ). Addition of affinity agents increased the concentration of the NPs for collection and detection to a higher level, and thus reliable quantitation could be conducted. While concerns about AA active site saturation should still be addressed by validation of observed NP trends by more sensitive techniques, affinity enhancement improves MD from a mostly qualitative technique to a quantitative technique by increasing the concentration of NPs to above their LLOQ. The LLOQ, LLOD, and linear range of this detection method when used with BK as an internal standard are illustrated in Fig. 4.7. For the LC-MS system employed, the LLOQ of these peptides is around 45 µM, and the LLOD is ~5 µM. Thus, NPs must be enriched for reliable quantitation. AE-MD was employed to enrich NPs collected in vivo. Results are indicated in Fig. 4.8, which describes relative concentration changes in seven identified crab NPs following feeding. All data points were obtained from the same crab; three feeding trials were conducted without AA, and one was conducted with AbMnP. The means and SEMs are plotted for the no AA trials. The NPs shown had low variance between the three no AA trials, and were detected in all time points. However, their concentrations do not appreciably change as they cannot be reliably quantified. Dynamic changes in these peptides are observed when AbMnPs are added to the 192 perfusate, increasing the concentration of the NPs to a level at which they can be quantitated. This result has been replicated in a separate C. borealis feeding trial (unpublished data). Most of the NPs appear to increase in concentration following feeding; reaching peak levels around 105 min after feeding. It is possible, therefore, that these NPs are neurochemicals involved in feeding. In the authors’ experience, crabs start feeding immediately after food is presented and stop 15-30 min. later, while still holding on to the food. They then recommence eating a few minutes later, around 60-90 min. after food is presented. This may occur as a result of the crab filling its stomach with food, allowing that food to be processed by the stomach and passed on to more distal parts of the digestive tract, and followed by repetition of the process. Since some of the NPs gradually increase before reaching peak concentration (RYLPT, SDRNFLRFa, YSFGLa, and pERPYSFGLa), these peptides might be continually released into the hemolymph while the animal is eating, and reaching a certain peak level could be an indicator of satiety. Other NPs that have a more dramatic spike in concentration levels (GPRNFLRFa, I/LNFTHKFa, and NFDEIDRSGFGFN) could be released after satiety is reached. Although these results are preliminary and further investigation will be required, they demonstrate how AE-MD can be used to observed dynamic changes in NPs that would otherwise appear unchanged in typical MD experiments, due to their presence below their LLOQ. 4.5. Conclusions and Future Work AE-MD has been conducted with a variety of AAs. AbMnP provided the greatest enhancement in neuropeptide recovery and could be used at higher concentrations than AbMµP due to their non-aggregation. This permitted statistically significant recovery enhancements for 4 out of 6 NP standards tested. The recovery enhancement was not specific to compounds with sequence similarity to FMRFamide, the compound against which the antibody was generated, and thus the mechanism of recovery enhancement is non-specific. This is likely due to the high 193 cross-reactivity of most NP antibodies, and although it could be detrimental if one desires to enrich a single NP for analysis due to specific interest in that compound, it will provide greater success in NP discovery and survey experiments, in which multiple and/or unknown NPs are of interest. These AbMnPs were used in experiments in the crab to determine the roles of circulatory NPs in feeding. They increased NP identification and made quantitation of NPs possible during a dynamic process. This will enable assignment of putative function for several NPs in feeding. AE-MD with AbMnP is a technique that has great potential to enrich NPs in microdialysate for correlation of function with molecular identity. 4.6. Respective Contributions and Acknowledgements CMS designed and carried out experiments. LL designed experiments and advised. CMS and LL wrote the paper. Andrew M. Kozicki is acknowledged for assistance with antibody beadbinding assays. Kevin T. Hayes is acknowledged for data processing assistance in the MD feeding study. 4.7. References 1. Lee, G. J.; Park, J. H.; Park, H. K. Microdialysis applications in neuroscience. Neurol Res. 2008, 30, 661-668. 2. Li, Q.; Zubieta, J. K.; Kennedy, R. T. Practical Aspects of in Vivo Detection of Neuropeptides by Microdialysis Coupled Off-Line to Capillary LC with Multistage MS. Anal Chem. 2009. 3. Kennedy, R. T.; Watson, C. J.; Haskins, W. E.; Powell, D. H.; Strecker, R. E. In vivo neurochemical monitoring by microdialysis and capillary separations. Curr Opin Chem Biol. 2002, 6, 659-665. 4. Perry, M.; Li, Q.; Kennedy, R. T. Review of recent advances in analytical techniques for the determination of neurotransmitters. Anal Chim Acta. 2009, 653, 1-22. 5. Torregrossa, M. M.; Kalivas, P. W. Microdialysis and the neurochemistry of addiction. Pharmacol Biochem Behav. 2008, 90, 261-272. 6. Ao, X.; Stenken, J. A. Microdialysis sampling of cytokines. Methods. 2006, 38, 331-341. 7. Duo, J.; Fletcher, H.; Stenken, J. A. Natural and synthetic affinity agents as microdialysis sampling mass transport enhancers: current progress and future perspectives. Biosens Bioelectron. 2006, 22, 449-457. 8. Kendrick, K. M. Microdialysis measurement of in vivo neuropeptide release. J Neurosci Methods. 1990, 34, 3546. 9. Wotjak, C. T.; Landgraf, R.; Engelmann, M. Listening to neuropeptides by microdialysis: echoes and new sounds? Pharmacol Biochem Behav. 2008, 90, 125-134. 10. Mabrouk, O. S.; Li, Q.; Song, P.; Kennedy, R. T. Microdialysis and mass spectrometric monitoring of dopamine and enkephalins in the globus pallidus reveal reciprocal interactions that regulate movement. J Neurochem. 2011, 118, 24-33. 11. Schmerberg, C. M.; Li, L. Function-Driven Discovery of Neuropeptides with Mass Spectrometry-Based Tools. Protein Pept Lett. 2012. 12. Van Eeckhaut, A.; Maes, K.; Aourz, N.; Smolders, I.; Michotte, Y. The absolute quantification of endogenous levels of brain neuropeptides in vivo using LC-MS/MS. Bioanalysis. 2011, 3, 1271-1285. 194 13. Lanckmans, K.; Sarre, S.; Smolders, I.; Michotte, Y. Quantitative liquid chromatography/mass spectrometry for the analysis of microdialysates. Talanta. 2008, 74, 458-469. 14. Behrens, H. L.; Chen, R.; Li, L. Combining microdialysis, NanoLC-MS, and MALDI-TOF/TOF to detect neuropeptides secreted in the crab, Cancer borealis. Anal Chem. 2008, 80, 6949-6958. 15. Behrens, H. L.; Li, L. Monitoring neuropeptides in vivo via microdialysis and mass spectrometry. Methods Mol Biol. 2010, 615, 57-73. 16. Haskins, W. E.; Watson, C. J.; Cellar, N. A.; Powell, D. H.; Kennedy, R. T. Discovery and neurochemical screening of peptides in brain extracellular fluid by chemical analysis of in vivo microdialysis samples. Anal Chem. 2004, 76, 5523-5533. 17. Myasein, K. T.; Pulido, J. S.; Hatfield, R. M.; McCannel, C. A.; Dundervill, R. F., 3rd; Shippy, S. A. Submicrolitre dialysis system to enable trace level peptide detection from volume-limited biological samples using MALDI-TOF-MS. Analyst. 2007, 132, 1046-1052. 18. Reed, B.; Bidlack, J. M.; Chait, B. T.; Kreek, M. J. Extracellular biotransformation of beta-endorphin in rat striatum and cerebrospinal fluid. J Neuroendocrinol. 2008, 20, 606-616. 19. Klintenberg, R.; Andren, P. E. Altered extracellular striatal in vivo biotransformation of the opioid neuropeptide dynorphin A(1-17) in the unilateral 6-OHDA rat model of Parkinson's disease. J Mass Spectrom. 2005, 40, 261-270. 20. Reed, B.; Zhang, Y.; Chait, B. T.; Kreek, M. J. Dynorphin A(1-17) biotransformation in striatum of freely moving rats using microdialysis and matrix-assisted laser desorption/ionization mass spectrometry. J Neurochem. 2003, 86, 815-823. 21. Zhang, H.; Stoeckli, M.; Andren, P. E.; Caprioli, R. M. Combining solid-phase preconcentration, capillary electrophoresis and off-line matrix-assisted laser desorption/ionization mass spectrometry: intracerebral metabolic processing of peptide E in vivo. J Mass Spectrom. 1999, 34, 377-383. 22. Prokai, L.; Kim, H. S.; Zharikova, A.; Roboz, J.; Ma, L.; Deng, L.; Simonsick, W. J., Jr. Electrospray ionization mass spectrometric and liquid chromatographic-mass spectrometric studies on the metabolism of synthetic dynorphin A peptides in brain tissue in vitro and in vivo. J Chromatogr A. 1998, 800, 59-68. 23. Osapay, G.; Prokai, L.; Kim, H. S.; Medzihradszky, K. F.; Coy, D. H.; Liapakis, G.; Reisine, T.; Melacini, G.; Zhu, Q.; Wang, S. H.; Mattern, R. H.; Goodman, M. Lanthionine-somatostatin analogs: synthesis, characterization, biological activity, and enzymatic stability studies. J Med Chem. 1997, 40, 2241-2251. 24. Haskins, W. E.; Wang, Z.; Watson, C. J.; Rostand, R. R.; Witowski, S. R.; Powell, D. H.; Kennedy, R. T. Capillary LC-MS2 at the attomole level for monitoring and discovering endogenous peptides in microdialysis samples collected in vivo. Anal Chem. 2001, 73, 5005-5014. 25. Baseski, H. M.; Watson, C. J.; Cellar, N. A.; Shackman, J. G.; Kennedy, R. T. Capillary liquid chromatography with MS3 for the determination of enkephalins in microdialysis samples from the striatum of anesthetized and freely-moving rats. J Mass Spectrom. 2005, 40, 146-153. 26. Lanckmans, K.; Stragier, B.; Sarre, S.; Smolders, I.; Michotte, Y. Nano-LC-MS/MS for the monitoring of angiotensin IV in rat brain microdialysates: limitations and possibilities. J Sep Sci. 2007, 30, 2217-2224. 27. Prokai, L.; Prokai-Tatrai, K.; Zharikova, A. D.; Nguyen, V.; Perjesi, P.; Stevens, S. M., Jr. Centrally acting and metabolically stable thyrotropin-releasing hormone analogues by replacement of histidine with substituted pyridinium. J Med Chem. 2004, 47, 6025-6033. 28. Prokai, L.; Zharikova, A.; Janaky, T.; Li, X.; Braddy, A. C.; Perjesi, P.; Matveeva, L.; Powell, D. H.; ProkaiTatrai, K. Integration of mass spectrometry into early-phase discovery and development of central nervous system agents. J Mass Spectrom. 2001, 36, 1211-1219. 29. Andren, P. E.; Caprioli, R. M. Determination of extracellular release of neurotensin in discrete rat brain regions utilizing in vivo microdialysis/electrospray mass spectrometry. Brain Res. 1999, 845, 123-129. 30. Emmett, M. R.; Andren, P. E.; Caprioli, R. M. Specific molecular mass detection of endogenously released neuropeptides using in vivo microdialysis/mass spectrometry. J Neurosci Methods. 1995, 62, 141-147. 31. Glass, J. D.; Guinn, J.; Kaur, G.; Francl, J. M. On the intrinsic regulation of neuropeptide Y release in the mammalian suprachiasmatic nucleus circadian clock. Eur J Neurosci. 2010, 31, 1117-1126. 32. Wilson, S. R.; Boix, F.; Holm, A.; Molander, P.; Lundanes, E.; Greibrokk, T. Determination of bradykinin and arg-bradykinin in rat muscle tissue by microdialysis and capillary column-switching liquid chromatography with mass spectrometric detection. J Sep Sci. 2005, 28, 1751-1758. 33. Nyitrai, G.; Kekesi, K. A.; Emri, Z.; Szarics, E.; Juhasz, G.; Kardos, J. GABA(B) receptor antagonist CGP36742 enhances somatostatin release in the rat hippocampus in vivo and in vitro. Eur J Pharmacol. 2003, 478, 111-119. 195 34. Pettersson, A.; Markides, K.; Bergquist, J. Enhanced microdialysis of neuropeptides. Acta Biochim Pol. 2001, 48, 1117-1120. 35. Pettersson, A.; Amirkhani, A.; Arvidsson, B.; Markides, K.; Bergquist, J. A feasibility study of solid supported enhanced microdialysis. Anal Chem. 2004, 76, 1678-1682. 36. Bernay, B.; Gaillard, M. C.; Guryca, V.; Emadali, A.; Kuhn, L.; Bertrand, A.; Detraz, I.; Carcenac, C.; Savasta, M.; Brouillet, E.; Garin, J.; Elalouf, J. M. Discovering new bioactive neuropeptides in the striatum secretome using in vivo microdialysis and versatile proteomics. Mol Cell Proteomics. 2009, 8, 946-958. 37. Ao, X.; Sellati, T. J.; Stenken, J. A. Enhanced microdialysis relative recovery of inflammatory cytokines using antibody-coated microspheres analyzed by flow cytometry. Anal Chem. 2004, 76, 3777-3784. 38. Ao, X.; Stenken, J. A. Water-soluble cyclodextrin polymers for enhanced relative recovery of hydrophobic analytes during microdialysis sampling. Analyst. 2003, 128, 1143-1149. 39. Ao, X.; Stenken, J. A. No delayed temporal response to sample concentration changes during enhanced microdialysis sampling using cyclodextrins and antibody-immobilized microspheres. Analyst. 2006, 131, 62-67. 40. Carneheim, C.; Stahle, L. Microdialysis of lipophilic compounds: a methodological study. Pharmacol Toxicol. 1991, 69, 378-380. 41. Duo, J.; Stenken, J. A. In vitro and in vivo affinity microdialysis sampling of cytokines using heparinimmobilized microspheres. Anal Bioanal Chem. 2010. 42. Duo, J.; Stenken, J. A. Heparin-immobilized microspheres for the capture of cytokines. Anal Bioanal Chem. 2010. 43. Fletcher, H. J.; Stenken, J. A. An in vitro comparison of microdialysis relative recovery of Met- and Leuenkephalin using cyclodextrins and antibodies as affinity agents. Anal Chim Acta. 2008, 620, 170-175. 44. Jensen, S. M.; Hansen, H. S.; Johansen, T.; Malmlof, K. In vivo and in vitro microdialysis sampling of free fatty acids. J Pharm Biomed Anal. 2007, 43, 1751-1756. 45. Khramov, A. N.; Stenken, J. A. Enhanced microdialysis recovery of some tricyclic antidepressants and structurally related drugs by cyclodextrin-mediated transport. Analyst. 1999, 124, 1027-1033. 46. Kurosaki, Y.; Nakamura, S.; Shiojiri, Y.; Kawasaki, H. Lipo-microdialysis: a new microdialysis method for studying the pharmacokinetics of lipophilic substances. Biol Pharm Bull. 1998, 21, 194-196. 47. Pich, E. M.; Koob, G. F.; Heilig, M.; Menzaghi, F.; Vale, W.; Weiss, F. Corticotropin-releasing factor release from the mediobasal hypothalamus of the rat as measured by microdialysis. Neuroscience. 1993, 55, 695707. 48. Stenken, J. A.; Church, M. K.; Gill, C. A.; Clough, G. F. How minimally invasive is microdialysis sampling? A cautionary note for cytokine collection in human skin and other clinical studies. AAPS J. 2010, 12, 73-78. 49. Sun, L.; Stenken, J. A. Improving microdialysis extraction efficiency of lipophilic eicosanoids. J Pharm Biomed Anal. 2003, 33, 1059-1071. 50. Trickler, W. J.; Miller, D. W. Use of osmotic agents in microdialysis studies to improve the recovery of macromolecules. J Pharm Sci. 2003, 92, 1419-1427. 51. Wang, Y.; Stenken, J. A. Affinity-based microdialysis sampling using heparin for in vitro collection of human cytokines. Anal Chim Acta. 2009, 651, 105-111. 52. Ward, K. W.; Medina, S. J.; Portelli, S. T.; Mahar Doan, K. M.; Spengler, M. D.; Ben, M. M.; Lundberg, D.; Levy, M. A.; Chen, E. P. Enhancement of in vitro and in vivo microdialysis recovery of SB-265123 using Intralipid and Encapsin as perfusates. Biopharm Drug Dispos. 2003, 24, 17-25. 53. Duo, J.; Stenken, J. A. Heparin-immobilized microspheres for the capture of cytokines. Anal Bioanal Chem. 2011, 399, 773-782. 54. Duo, J.; Stenken, J. A. In vitro and in vivo affinity microdialysis sampling of cytokines using heparinimmobilized microspheres. Anal Bioanal Chem. 2011, 399, 783-793. 55. Cooper, R.; Lee, L. Chips & Tips: Preventing suspension settling during injection. Lab on a Chip [serial on the Internet]. 2007: Available from: http://www.rsc.org/Publishing/Journals/lc/Chips_and_Tips/suspension_injection.asp#. 56. Safarik, I.; Safarikova, M. Magnetic techniques for the isolation and purification of proteins and peptides. BioMagnetic Research and Technology. 2004, 2, 7. 196 4.8. Figures 100.0% AE-MD with C18 Magnetic 80.0% Microparticles No AA 60.0% p=0.0093 Fold-change:1.29 C18SµP 40.0% 20.0% 0.0% FLP I Figure 4.1. Recovery enhancement of FLP I by adding C18 silica microspheres to the perfusate. The experiment was carried out 3 times and the mean recoveries are indicated with error bars showing the SEM. The p-value for a Student’s t-test and the fold change are indicated on the graph. Abbreviations are as follows: Homarus americanus FMRFamide-like peptide I (FLP I), C18 silica microparticles (C18SµP), and no affinity agent (No AA). 100.0% 80.0% AE-MD with C18 Magnetic Microparticles * * No AA 60.0% C18MµP * 40.0% * 20.0% 0.0% FMRFa FLP I FLP II SP SMT BK Figure 4.2. Relative recovery enhancement for six different NP standards by the addition of C18 magnetic microparticles to the perfusate. Values indicated are the means, with error bars showing the SEM. NP names are abbreviated as follows: Homarus americanus FMRFamide-like peptide I (FLP I), Homarus americanus FMRFamide-like peptide II (FLP II), substance P (SP), 197 somatostatin-14 (SMT), and bradykinin (BK). C18 magnetic microparticles are abbreviated C18MµP, and no affinity agent is written as No AA. Significant differences (p<0.05) from the No AA condition are indicated with an asterisk (*). 100.0% 80.0% 60.0% AE-MD with Antibody-Coated Microparticles No AA AbMµP * 40.0% 20.0% 0.0% FMRFa FLP I FLP II SP SMT BK Figure 4.3. Relative recovery enhancement for 6 NP standards by the addition of antibodycoated magnetic microparticles to the probe perfusate. Values indicated are the means, with error bars showing the SEM. NP abbreviations are indicated in the legend to Fig. 1. Antibody-coated magnetic microparticles is abbreviated AbMµP, and no affinity agent is written as No AA. Significant differences (p<0.05) from the No AA condition are indicated with an asterisk (*). 198 120.0% 90.0% Microbeads A. unbound B. Nanobeads 1:500 80.0% bound 100.0% unbound bound 70.0% 60.0% 80.0% 50.0% 60.0% 40.0% 40.0% 30.0% 20.0% 20.0% 10.0% 0.0% 0.0% bradykinin HoA FLP HoA FLP II I SMT FMRFa Nanobeads 1:50 120.0% 100.0% SP C. unbound bound 80.0% 60.0% 40.0% 20.0% 0.0% Figure 4.4 Bound and unbound percentages of NPs in bead binding assays for antibody-linked particles. A) Microscale beads. B) and C) Nanobeads prepared by incubating with antibody at a B) 1:500 or C) 1:50 dilution. Abbreviations in text. Each incubation was carried out 3 times and the average values for these are indicated with SEM as the error bars. 199 100.0% 80.0% 60.0% AE-MD with Antibody-Coated Nanoparticles *† *† No AA *† AbMnP 6x AbMnP * * * * 40.0% 20.0% 0.0% FMRFa FLP I FLP II SP SMT BK Figure 4.5. Relative recovery enhancement for 6 NP standards by the addition of antibodylinked magnetic nanoparticles at a concentration equivalent to those used with previous affinity agents and at a concentration six times higher. Values indicated are the means, with error bars showing the SEM. NP abbreviations are indicated in the legend to Fig. 1. Antibody-coated magnetic nanoparticles is abbreviated AbMnP, the higher concentration is noted as 6x AbMnP, and no affinity agent is written as No AA. Significant differences (p<0.05) from the No AA condition are indicated with an asterisk (*). Significant differences (p<0.05) from the AbMnP condition are indicated with a dagger (†). 200 A) B) Figure 4.6. Ultrahigh performance liquid chromatography-time-of-flight (UPLC-TOF) extracted ion chromatograms for two peptides of interest (A. GPRNFLRFamide, B. ENRNFLRFamide, in their +2 states) in microdialysis samples obtained from a single Cancer borealis under baseline conditions. The highest intensity peak (red) represents a sample collected with AE-MD, with antibody-linked magnetic nanoparticles (AbMnPs) in the perfusate, and the two lower intensity peaks (maroon and black) represent samples collected without affinity agent from the same crab. Samples were all run on UPLC-TOF on the same day with the same chromatographic and mass spectrometric conditions. Chromatograms are smoothed, baseline subtracted, and plotted with the same Y axis scale, which is represented as raw ion counts. Retention times are indicated at the top of the peak, and the masses selected for generation of the XIC are indicated in the top right corners. 201 FLP I FLP II 1.5 y = 0.0136x + 0.0423 R² = 0.9762 4 3 Area FLP II/BK Area FLP I/BK 5 2 1 0 0 100 200 300 y = 0.0049x - 0.1398 R² = 0.9902 1 0.5 0 0 400 -0.5 0.03 0.25 0.025 Area FLP II/BK Area FLP I/BK 0.3 0.15 y = 0.0038x + 0.1558 R² = 0.8726 0.1 0.05 0 200 300 400 Concentration of FLP II (uM) Concentration of FLP I (uM) 0.2 100 0.02 0.015 y = 0.0008x - 0.0103 R² = 0.5434 0.01 0.005 0 0 10 20 Concentration of FLP I (uM) 30 20 30 40 50 Concentration of FLP II (uM) Figure 4.7. Linear range for peptide quantitation in this system. Different concentrations of NP standards were spiked with 30 nM BK and the normalized areas were plotted against concentration. The bottom two panels show the range at which NPs are expected to be present endogenously. 202 GPRNFLRFa I/LNFTHKFa 12 Fold-change 6 8 6 4 4 2 2 0 0 100 200 Time after feeding (min) 0 0 100 200 Time after feeding (min) Fold-change Fold-change 6 4 2 2 No AA 1.5 1 0.5 0 100 200 Time after feeding (min) 0 100 AE-MD 200 Time after feeding (min) pERPYSFGLa 2 3 Fold-change Fold-change Time after feeding (min) 2.5 SDRNFLRFa No AA AEMD 1.5 2 1 0 200 3 4 0 100 YSFGLa NFDEIDRSGFGFN 8 0 0 6 5 4 3 2 1 0 0 -1 Fold-change 10 Fold-change RYLPT (Proctolin) 8 100 1 0.5 200 0 0 100 200 Time after feeding (min) Time after feeding (min) Figure 4.8. Affinity-enhanced microdialysis permits observation of changes in NP levels following feeding. The x-axis crosses the y-axis at a value of one, which would indicate no change from baseline conditions. Without affinity agent, the NPs are present at levels too low for changes to be observed. For No AA data, 3 feeding trials from the same crab are pictured as an average with standard error as the error bars. Some error bars are obscured by the size of the marker at each data point. The AE-MD data was obtained from the same crab in a feeding trial. AE-MD data shows dynamic changes in NPs after feeding for several peptides, whereas the No AA data shows little variation. 203 4.9. Tables Table 4.1. Sequences of several peptide standards used in bead-binding assays. C-terminal motifs are underlined. Details of the reverse phase separation method can be found in the text of the article, experimental section. Abbreviation: FMRFamide-like peptide (FLP). Table 4.1. Sequences, C-Terminal Motifs, and Reverse-Phase LC Retention Times of NP Standards Substance Sequence Retention Time (min) FMRFa FMRFa 18.25 Substance P RPKPQQFFGLMa 20.68 Homarus americanus FLP I SDRNFLRFa 19.17 H. americanus FLP II TNRNFLRFa 18.85 Bradykinin RPPGFSPFR 18.13 Somatostatin-14 AGCKNFFWKTFTSC 21.41 Table 4.2. Results of AE-MD experiments. Results from numerous in vitro microdialysis studies are summarized in this table. Technical replicates were averaged to yield one value per experiment, and these values were then combined to determine the average RR and standard error of the mean (SEM) of that value. The number of experiments is shown as n. Analysis of variance (ANOVA) tests indicated statistically significant differences in the RRs obtained across the different AAs, and post-hoc Tukey-Kramer honestly significant difference (HSD) tests yielded the p-values for significance indicated in the table. With one exception, only p-values of 0.05 or smaller are reported, as this was the significance threshold. Blank cells indicate nonsignificant p-values. Fold-changes were also calculated for the statistically significant values. Neuropeptide (NP) names are abbreviated as follows: FMRFamide (FMRFa), Homarus americanus FMRFamide-like peptide I (FLP I), Homarus americanus FMRFamide-like peptide II (FLP II), substance P (SP), somatostatin-14 (SMT), and bradykinin (BK). AAs are abbreviated as follows: C18 magnetic microparticles is abbreviated C18MµP, no affinity agent is written as No AA, antibody-coated magnetic microparticles is abbreviated AbMµP, antibody-coated 204 magnetic nanoparticles is abbreviated AbMnP, and the higher concentration is noted as 6x AbMnP. Table 4.2. Recovery Enhancement Caused by Addition of Affinity Agents Comparison to Comparison to No AA AbMnP Average NP AA n SE RR Foldp p Fold-Change Change FMRFa No AA 6 21.90% 2.52% C18MµP 5 23.00% 2.76% AbMµP 7 18.50% 2.34% AbMnP 4 23.00% 3.09% 6x AbMnP 3 34.80% 3.57% 0.0548 1.59 FLP I No AA 7 26.30% 3.46% C18MµP 6 73.10% 3.74% <0.0001 2.78 AbMµP 7 33.30% 3.46% AbMnP 4 48.30% 4.58% 0.0072 1.84 6x AbMnP 3 55.20% 5.29% 0.0013 2.1 FLP II No AA 6 26.00% 3.60% C18MµP 6 61.50% 3.60% <0.0001 2.36 AbMµP 8 34.60% 3.12% AbMnP 4 34.10% 4.41% 6x AbMnP 3 87.30% 5.09% <.0001 3.35 <.0001 2.56 SP No AA 5 2.21% 2.67% C18MµP 5 24.80% 2.67% 0.0002 11.2 AbMµP 5 7.82% 2.67% AbMnP 3 69.70% 3.45% <0.0001 31.6 6x AbMnP 3 92.10% 3.45% <0.0001 41.7 0.0024 1.32 SMT No AA 4 11.10% 4.55% C18MµP 5 45.10% 4.07% 0.0003 4.09 AbMµP 6 40.10% 3.71% 0.001 3.63 AbMnP 4 37.90% 4.55% 0.0051 3.43 6x AbMnP 3 82.90% 5.25% <0.0001 7.5 <.0001 2.19 BK No AA 6 51.60% 3.33% C18MµP 5 56.20% 3.64% AbMµP 7 47.70% 3.08% 6.16% AbMnP 5 40.11% % 6x AbMnP 3 45.20% 4.70% 205 Table 4.3. Comparison of NP complements detected with conventional and affinity-enhanced microdialysis. The number of NPs detected in a reference study [14] was compared to the results from this study. An x indicates a detected peptide. For the non-enhanced in vivo MD data obtained in this study, peptides that were present in all 3 feeding trials were determined to be detected. Table 4.3. Comparison of NP Complement Detected with Conventional and Enhanced In Vivo Microdialysis Sampling Method In vivo In vivo AEMD [14] MD 4-16 hrs 30 min Sampling Time Peptide Family Peptide sequence x A-type allatostatin ARPYSFGLa x x DPYAFGLa x ERPYSFGLa x PDMYAFGLa x pERPYSFGLa x x SPYAFGLa TNFAFSPRLa x x YAFGLa x x YSFGLa x x B-type allatostatin NNNWSKFQGSWa x x NWNKFQGSWa x STNWSSLRSAWa x TSWFKFQGSWa Allatostatin x x prohormone related DPYAFGLGKRPADL peptide DPYAFGLGKRPADLYEFGLa x x CabTRPs APSGFLGM(O)Ra x APSGFLGMRa x x APSGFQa x x GFLGMRa x Orcokinin NFDEIDRSGFa x NFDEIDRSGFG x x NFDEIDRSGFGF x NFDEIDRSGFGFH Affinity- In vivo MD 30 min x x x x x x x x x x 206 FLP RYamide YRamide SIFamide Other NP NFDEIDRSGFGFN NFDEIDRSGFGFV NFDEIDRSSFGFN NFDEIDRSSFGFV APQRNFLRFa APRNFLRFa DGGRNFLRFa DRNFLRFa GAHKNYLRFa GGRNFLRFa GPRNFLRFa RNFLRFa RSFLRFa SDRNFLRFa SENRNFLRFa SGRNFLRFa SMPSLRLRFa TNRNFLRFa FVGGSRYa HIGSLYRa GYRKPPFNGSIFa I/LNFTHKFa PFCNAFTGCa (CCAP) pQLNFSPGWa (RPCH) TNFAFSPRLa (CabPK-I) Total x x x x x x x x x x x x x x x x x x x x x 35 x x x x x x x x x x x x x x x x x x x x 31 17 207 Chapter 5: MS/MS Characterization of the Neuropeptidome of the Crayfish Orconectes rusticus Adapted from Schmerberg, C. M., Liang, Z., and Li, L. J. Proteome Res. 2012 [in preparation] 5.1. Abstract The rusty crayfish, Orconectes rusticus, is an important species in behavioral neuroscience and invasive species ecology. It presents several well-defined behaviors, permitting it to be both an ideal model species for studying social dominance and aggression, and a successful invasive species in the upper Midwest United States. Monoamine neurotransmitters (NTs), human drugs of abuse, and spatial learning have also been studied in O. rusticus. Despite its significance, the neuropeptides (NPs) of O. rusticus have not yet been cataloged, although NPs have been implicated in aggression, addiction, and adaptation to environmental change in other species. NP studies in other crayfish species have been limited in the techniques or organs used. In this work, the premier technique for NP identification— tandem mass spectrometry (MS/MS)—was used to determine the NP complements of the sinus gland (SG) and brain in Orconectes rusticus. Fractions from high performance liquid chromatography (HPLC) were enzymatically digested if necessary and injected onto ultra-high performance liquid chromatography (UPLC) coupled on-line with an electrospray ionization (ESI) quadrupole time-offlight (QTOF) mass spectrometer operating in data-dependent acquisition (DDA) mode. MS/MS interpretation was conducted by two different strategies: database search software (Mascot), and de novo sequencing software (PEAKS). These approaches were aided by the construction of a home-built crustacean neuropeptide database. Highly confident identifications were made for 98 previously identified neuropeptide sequences from 8 families. Over 47 additional putative neuropeptides were also identified from 12 families, including sequences from families already observed in Orconectes species (CPRPs, PDHs), and peptides not previously identified in crustaceans using mass spectrometry. These represent potentially novel neuropeptides in crustacean species. Due to overlap in 208 NP sequences, the minimum number of unique peptides that could generate the observed PSMs is 23. This catalogue of NPs will be vital for further research into the roles of NPs in behavior, environmental adaptation, and addiction in O. rusticus. 5.2. Introduction The rusty crayfish Orconectes rusticus is an important species in two major areas of biology. It is an important model species in behavioral neuroscience, studied in applications ranging from aggression to the reinforcing effects of human drugs of abuse. Studies of O. rusticus are also of vital importance in the field of invasive species ecology, as its invasion of several bodies of water in Northern Wisconsin has been documented in detail. 5.2.1 Significance of Orconectes rusticus to the Field of Neurobiology The contribution of the rusty crayfish to the field of behavioral neuroscience rests primarily on its expression of several well-described and stereotyped behaviors, as reviewed by several authors [1, 2]. O. rusticus and other crayfish typically lead solitary lives, without alliances or pair-bonds, and interaction is mostly agonistic, resulting from competition over resources (food, shelter, mates), but existing even when there is no contested resource, due probably to the high density at which they live in the wild. Social dominance is based mostly on size, with fighting determining hierarchies when the animals’ sizes are similar. Fights have a set progression from one stereotyped behavior to another; starting with ritualized visual displays, followed by progression to antennal whipping, claw locking, wrestling, and finally claw use [1, 2]. The ethology of aggression in O. rusticus is thus very well defined. Attempts have been made to understand the neurochemical basis of this behavior in the context of monoamine neurotransmitters (NTs), as reviewed in several papers [1, 2]. Studies in decapods including Orconectes rusticus are equivocal in the roles of amine NTs in agonistic posture and behavior. Thus, it appears that no single NT is a whole-body signal for aggression, but that state- 209 and/or dose-specific effects are important [1, 2]. The potential of O. rusticus to serve as a model species in the fields of addiction [3-7] and learning/memory [8-13] has also recently been demonstrated, although the neurochemistry of these behaviors has not been studied in as much detail. Although O. rusticus is a useful model animal in a number of behavioral neuroscience applications and a large amount of progress has been made in understanding the neurochemical basis of this aggression in this species, much remains to be studied. 5.2.2. Significance of Orconectes rusticus to the Field of Ecology Orconectes rusticus is also an important species in the field of invasive species ecology. Its invasion of the area in northern Wisconsin near the Michigan border—far from its native range in the Ohio River drainage—has been studied in detail, with numerous research and review articles written. A 2006 meta-review of data on crayfish collected since 1932 illustrates the displacement of several native crayfish species by O. rusticus, which was introduced starting in the 1960’s [14]. A recent review enumerated the factors that enable it to be a successful invader, and the negative ecological impact it can have [15]. The aggressiveness of O. rusticus is a major factor in its ability to kill or evict native crayfish. They decrease the diversity and amount of macrophytes, large aquatic plants that provide both food and shelter for fish and other invertebrates. Snails and other mollusks, fish eggs, and various types of litter are food sources for rusty crayfish, and these are reduced in number when O. rusticus are present [15]. The potential for O. rusticus to further invade lakes and rivers and displace native crayfish was modeled in 2011, and a 25% chance of this was determined for 115 lakes and 5,000 km of streams in Wisconsin [16]. From this and many other studies, it is clear that the rusty crayfish presents a significant threat to freshwater ecosystems. Its survival appears to be dependent on a threshold dissolved calcium content and pH of lake water of 2.5 mg/L and 5.5, respectively, as reviewed by Olden et al. [14]. Predation levels also influence the population of O. rusticus [17-19]. 210 5.2.3. Current Knowledge of Neuropeptide Content in Orconectes spp. and Significance of This Work It has previously been demonstrated that neuropeptides (NPs), an important class of neurotransmitters with additional neurohormonal function, are important in adaptation to environmental factors, aggression, and addiction. Although the neuropeptidomes of several other decapod crustacean species have been determined, including the NP content in the eyestalk of the crayfish Orconectes limosus [20], NPs in O. rusticus have not yet been described. In this work, the NP complements of both the brain and eyestalk of O. rusticus are characterized using multiple mass spectrometry (MS) approaches. Paramount among these is tandem MS (MS/MS), which is used to identify the sequence of a peptide directly from the ions it produces upon gas-phase fragmentation. This technique has become the gold standard for neuropeptide identification, as opposed to antibodybased techniques, genomics-based techniques, and sequential cleavage of amino acids (Edman degradation). Antibody-based techniques suffer from low specificity and do not provide sequence information. Gennomics-based techniques often cannot predict the full complexity of posttranslational modifications that NPs undergo. Edman degradation requires a large amount of highly purified sample. Neuropeptides are best characterized by tandem mass spectrometry (MS/MS), especially when no genome is available, due to the large degree of post-translational processing. This work comprises the first neuropeptide study in Orconectes rusticus. The NP contents of several ganglia in the related species Orconectes limosus have been reported in a number of different studies, identified by MSbased techniques and/or Edman degradation, but these reports have only yielded a total of 14 unique peptides from the following families: pigment dispersing hormones (PDH), molt-inhibiting hormones (MIH), crustacean hyperglycemic hormones (CHH), CHH precursor-related peptides (CPRPs), 211 orcokinins, orcomyotropin, and A-type allatostatins (A-AST) [20-25]. The most comprehensive one of the previously mentioned studies employed nanoscale liquid chromatography (nanoLC) coupled to tandem mass spectrometry (MS/MS) for de novo sequencing of NPs obtained from an extract of the Xorgan-sinus gland (XO-SG) complex [20], one of the main neurosecretory structures in crustacean species. This study identified a total of six peptides, with three additional truncated versions of the CPRP peptides also identified. Compared with more recent studies in which 122 NPs could be identified from tissues of the green crab Carcinus maenas [26], it is fair to assume that many Orconectes NPs remain undiscovered. Advances in MS and MS/MS instrumentation, combined with multistage separation techniques, permit such a study to provide a more comprehensive picture of the neuropeptidome of a given species. In addition, there have been improvements in software for de novo sequencing and MS/MS identification based on database search, and it has recently become possible to create a database of known crustacean NPs. In this work, the aforementioned strategies are employed to characterize the neuropeptidome of the Orconectes rusticus XO-SG and brain (supraoesophageal ganglion). A two-dimensional reversed phase (RP) liquid chromatography (LC) strategy was employed, with neutral and acidic mobile phases to provide orthognality. The second dimension nanoLC was coupled directly to a highly sensitive quadrupole time-of-flight (QTOF) mass spectrometer for MS/MS analysis. Software programs for database search interpretation of MS/MS spectra (used in this study: Mascot, Matrix Science) and de novo sequencing-based MS/MS interpretation (used in this study: PEAKS, Bioinformatics Solutions, Inc.) were employed and the results are compared. In all, 145 unique peptide-spectrum matches (PSMs) were obtained, mapping to 98 previously identified neuropeptide PSMs from 8 families and 47 putative neuropeptide PSMs from 12 families. This is the first investigation of NPs in O. rusticus and greatly increases the number of known NPs in Orconectes and other crustacean species. 212 5.3. Experimental Section All portions of this section can be found in Appendix A with added detail. 5.3.1. Animals Male and female Orconectes rusticus were kindly provided by Alex Laztka, a graduate student in the laboratory of Jake Vander Zanden, in the Department of Limnology at the University of Wisconsin-Madison, and co-workers. They were obtained from fresh water lakes in Northern Wisconsin centered around the Limnology Department’s Trout Lake Research Station, on Trout Lake in Boulder Junction, WI. Animals were sampled as part of a National Science Foundation (NSF)funded Long-Term Ecological Research (LTER) study, and were obtained and transported with the permission of the Wisconsin Department of Natural Resources. Permission for transport must be obtained due to the status of O. rusticus as an invasive species in Wisconsin. O. rusticus were obtained in modified minnow traps placed in lakes at a depth of approximately 1 m, in an area where the lake substrate is “cobble,” or rocks approximately 6-8 cm in diameter. The traps were modified by increasing the entrance hole to 5cm to accommodate mature male crayfish. Traps were baited with beef or chicken liver. This method has been shown to preferentially collect male crayfish, and although both genders were observed, around 75% of the animals were male. Traps were emptied at an interval of 20-24 hrs. O. rusticus was then separated from other catch by hand and kept in buckets in a refrigerator at 4ºC for approximately 12 hrs. The crayfish should not be stored in water, as the water will quickly become foul and deoxygenated. Storing animals in water in the refrigerator and/or during transport led to approximately 95% mortality in a batch of around 350 crayfish. Stored dry and cold, they enter a state similar to hibernation and can survive longer. They were then transferred to a carrying container that contains an ice pack and 213 transported to Madison, WI by personal vehicle (a trip of approximately 3.5 hrs). Species identity was verified prior to housing in Madison in freshwater aquaria. Aquaria contained fresh, dechlorinated water, and crushed gravel substrate. They also were equipped with Aquaclear 30 (Hagen) filtration units and conventional air bubblers. No more than 45 individuals were housed per 20 gallon tank. Care was taken to ensure a tight fit for the lid of the aquarium as the crayfish can escape through very small holes. Animals were not stored more than a day at a density of 45 animals/20 gal tank, and at lower densities, of about 20 per 20 gal tank, could be housed indefinitely. If kept for longer periods of time, O. rusticus were fed and monitored for molting, and regular water changes were conducted. Crayfish will cannibalize if they are not fed every 2-3 days, and fighting often occurs over food. They were fed white fish, as it does not foul the water and pelleted food designed for bottom-feeding fish from a pet store. Enough food was presented so that all animals had something to eat to prevent fighting. If O. rusticus molted, they were isolated with the old shell until their new shell hardens. Other individuals would attack and kill unprotected newly molted individuals. These newly molted individuals can and should consume the old shell for nutritional purposes. Hardening takes 1-2 days. Due to waste accumulation in the substrate and water, the tanks were cleaned weekly with a 20% water change, or more often if housed in high density. 5.3.1.1. Orconectes rusticus Dissection Brains and eyestalks were dissected from O. rusticus without the aid of a microscope. Whole eyestalk extracts were prepared instead of dissecting the sinus gland out of the eyestalk due to its miniscule size, which is difficult to locate even under a dissecting microscope. O. rusticus was put on ice until movement ceased (approximately 15-20 min). Appendages were removed and the tail nerve cut with dissecting scissors to prevent tail flipping. The carapace was cut along a line starting below 214 one eyestalk, continuing up toward the rostrum, and then down toward the other eyestalk (indicated in figures in Appendix A). The distal part of the body was then discarded and the brain was located inside the shell immediately behind the rostrum. It was removed with fine forceps and placed in a 1.5 mL tube containing 0.5 mL acidified methanol (90:9:1 methanol: water: acetic acid by volume) on ice. 25-30 brains were placed in a single tube. They were stored at -20°C or -80°C (if storage occurred for more than a month) until extraction. Entire eyestalks were removed by cutting the soft tissue where they were connected to the head with fine scissors. These were placed in a similar tube, with about 2025 pairs per tube. 5.3.2. Extraction and Sample Preparation Tissues were homogenized in acidified methanol using plastic pestles for soft tissues and a glass vial and pestle for whole eyestalks. Vigorous homogenization followed by sonication (5 min) was required for all tissues, in particular the eyestalks. Centrifugation was used to separate liquid and solid portions (5 min x 16,100 g), and the solids were re-extracted 3 times with the initial volume of acidified methanol, saving the liquid fraction each time. It was later determined via MALDI-TOF/TOF that the third extraction from O. rusticus brain tissue yielded mostly lipids and few peptides, so only the first two were combined for that tissue. For other tissues, all liquid portions were combined. The solvent was evaporated. Two brain and two eyestalk samples were prepared and analyzed, each containing tissues pooled from 25-30 individual crayfish. O. rusticus brain samples were subjected to a delipidation step. They were reconstituted in water and combined with an equal volume of a solution of chloroform: methanol (3:1) and a liquidliquid extraction was conducted. The aqueous layer was saved, and the lipid layer was re-extracted 3 times. All samples were reconstituted in 0.1% formic acid (FA) in water. 5.3.3. Liquid Chromatography-Mass Spectrometry 215 Samples were subjected to a first dimension of liquid chromatography as a pre-purification step. A neutral pH (7.4, solvent A: 25mM NH4HCO2, solvent B: 9:1 ACN: 25mM NH4HCO2) reversed-phase (Inertsil ODS-4 column, 3µm, 2.1 x 150 mm, GL Sciences, Tokyo, Japan) was used with an Alliance HPLC (Waters, Milford, MA) system. Fractions were collected every 3 min with a Rainin Dynamax Model FC-4 fraction collector. Early eluting fractions were combined with late eluting fractions to reduce the total number of samples while maintaining separation. Samples were injected onto a C18 reversed-phase column (BEH130 C18, 1.7 µm, 75 µm x 100 mm, Waters, Milford, MA) using a Waters nanoAcquity UPLC system (Waters, Milford, MA). The outlet of this column was connected to a fused silica capillary with a pulled tip of internal diameter 5 µm (Sutter Instrument Company, Novato, CA). This was used as the ESI inlet on a Waters Synapt G2 QTOF (Waters, Milford, MA). A 120 min reversed-phase run with solvent A as 0.1% FA in water and B as 0.1% FA in ACN was used. The instrument was operated in data-dependent MS/MS mode and Glu-fibrinopeptide was infused for lockspray calibration. 5.3.4. Databases A variety of approaches were used for database construction in this study. As there is no available genome for any decapod crustacean, various databases were assembled using a combination of in-house compiled information and publicly available data. The in-house component contained all known crab and lobster neuropeptides, compiled from published papers out of the Li lab and others, and theses from the Li lab. This database portion is referred to as LiDB and can be found in Appendix B and will be made publicly available shortly. The National Center for Biotechnology Information (NCBI) protein database (www.ncbi.nlm.nih.gov/protein) was also searched to obtain sets of relevant known protein sequences. One such list was created by bioinformatics mining by a previous lab member. This contained all NCBI crustacean NP sequences, several crustacean protein sequences, and 216 numerous other NP sequences from a variety of multicellular organisms. This list of sequences is referred to as WF, for the name of the previous lab member. The entire list of decapod sequences was also downloaded from NCBI (“decapod”), as was the entire list of crustacean sequences (“crustacean”). Finally, a database was acquired from NCBI by restricting the taxonomy to crustacea and searching for the keywords “peptide” or “hormone” (NCBI query: (peptide[All Fields] OR hormone[All Fields]) AND "Crustacea"[Organism]). This is referred to as crustaceanNP. Databases were compiled in FASTA format and edited for redundancy. Only results from crustaceanNP+LiDB are shown, as this was determined to be the most comprehensive database that did not also contain large numbers of erroneous sequences of compounds that were either not NPs or were not from related species. 5.3.5. Data Analysis A home-built computer (3.30 GHz AMD FX-6100 Six-Core Processor, 32.0 GB RAM, 64-bit) operating in Windows 7 Professional (64-bit) was used for the majority of analyses. Protein Lynx Global Server (PLGS, Waters, Milford, MA) was used to search a variety of home-built databases against the data obtained and to convert the data from .raw folders into .pkl files for MASCOT search. This software also conducted an internal mass calibration with the lock mass compound. Several databases and combinations of databases were used, as detailed in the results and discussion. For each search, the modifications C-terminal amidation, pyroglutamation, and methionine oxidation were always added. In all searches, a no enzyme search was conducted, a minimum of 3 fragment ion matches per peptide were required, and mass tolerance was set to default. The software’s built-in de novo sequencing tool was also employed with default mass tolerances and the same modifications. No results were obtained with either the database search or de novo functions. 217 The .pkl files created by PLGS were searched via an in-house Mascot server (Matrix Science, London, UK) using the Daemon batch uploader tool. For the Mascot server, only the LiDB+WF and LiDB+crustaceanNP databases were uploaded. For each search, the modifications C-terminal amidation, pyroglutamation, and methionine oxidation were selected. In all searches, a no enzyme search was conducted and mass tolerance was set to 0.5 Da for precursors and 0.25 Da for fragments. Peptide charges were set to 2+, 3+, and 4+. Files from all fractions were combined by Mascot into a single file for searching. These same .pkl files were analyzed using PEAKS studio 5.3 (Bioinformatics Solutions Inc., Waterloo, ON). Data was preprocessed to select only high-quality spectra. De novo was conducted with no enzyme and the modifications C-terminal amidation, pyroglutamation, and methionine oxidation. Mass tolerance was 0.5 Da for precursors and 0.25 Da for fragments. Following the de novo search, a comparison to database sequences was conducted. This was done with the LiDB+WF and LiDB+crustaceanNP databases, and all other parameters the same. 5.3.6. Results Curation Peptide spectrum matches (PSMs) obtained with PEAKS and Mascot were output in comma separated values (.csv) format for further analysis in Microsoft Excel. Although Mascot reports a “Mascot score,” this is their version of a -10logP value, which is also what PEAKS reports. All PSMs scoring less than 15 were removed from the results. The two brain and two eyestalk samples were analyzed in parallel, and the best score obtained was saved. False-discovery rates (FDRs) were not generated by PEAKS due to the small database size, and the FDRs generated by Mascot were for a score threshold of 40, not useful for our score cutoff of 15. Occasionally, PSMs with large numbers of repeating residues were disregarded as likely false positives. PSMs that were assigned to proteins or to unknown products of the Daphnia genome are listed separately in Appendix C. PSMs for peptides or 218 hormones were subjected to meta-analysis to compare the performance of Mascot and PEAKS. Venn diagrams were generated using software created at the Pacific Northwest National Laboratory. For final reporting of the neuropeptides identified, some overlapping PSMs were manually aligned and parsimony was used to determine the minimal number of precursors that could lead to those PSMs. Peptides are reported in Tables 5.1-5.3 and Appendix C. Several sequences determined by PEAKS de novo interpretation of MS/MS spectra were not found in databases, and these were subjected to further analysis. Those that appeared in more than one extract, or had sequence motifs common to NPs, were searched against the NCBI protein database using tblastn and restricting to Crustacea (taxon id 6657). Matches with the lowest E-value are reported. Where multiple proteins had the same E-value, either all are listed or they are described generally. This data is also reported in Appendix C. 5.4. Results and Discussion In total, 145 high-quality peptide-spectrum matches (PSMs) were made to MS/MS spectra. These mapped to 98 peptides previously observed in decapod crustaceans using mass spectrometry, from 8 different neuropeptide families. The additional spectra mapped to 47 putative neuropeptides from 15 families. These either represent peptides observed in non-decapod crustaceans, or peptides whose presence had previously only been identified by genetic techniques. Additional matches were made to non-neuropeptide entries in the database, including hemocyanin, actin, and currently unidentified products of the Daphnia genome. Although other databases were used, only data from crustaceanNP+LiDB is presented, as this combination contained the most NP sequences with the fewest non-NP or non-crustacean sequences. A smaller database is desired when searching for NPs, as searches must be done without using specific enzyme cleavage sites to capture the full complexity of NPs and potential novel proteolytic processing products. This smaller database, although it permitted 219 such searches to be carried out in a reasonable period of time (hours instead of days), led to a number of issues, mostly with determining the quality of a match. Reproducibility between samples of the same tissue type was moderate. Around one third of PSMs from Mascot and one fourth from PEAKS were observed in both pooled tissue samples (Fig. 5.1). This illustrates the necessity of analyzing multiple replicates. It is likely that this is due to the somewhat random nature of precursor selection for MS/MS, and that technical replicates might be sufficient. It is unlikely that the samples contained highly different NP contents, since they were pooled tissues from 25+ animals, all collected from the same geographical region around the same season. It is nevertheless possible that slight differences in the locations and days of collection led to NP variation between the samples. Another possible source of this variation between samples could be the inclusion of a number of random matches, which are not likely to occur the same way in two different samples. This concern, however, cannot be addressed in neuropeptide studies with the current statistical methods of assigning false discovery rates (FDRs) and the probabilities (P) of matches being correct. False discovery rates and match probability are determined based on statistical methods that take into account a number of factors, and should presumably return a good estimate of the accuracy of assigning a MS/MS spectrum to a given peptide sequence. All current methods to calculate this value take into account the number of fragment ion matches observed versus the number of possible matches. These formulas will provide a good estimation of the quality of a match when the peptide is large, but may not do so for smaller peptides with fewer possible matching ions. The gold standard for acceptability of a PSM remains visual verification, and in our experience, PSMs with lower Mascot scores still passed this test, while PSMs with similar scores from PEAKS did not. Although both software programs report a -10logP value, the exact scoring mechanisms to calculate that probability 220 (P) differ. Therefore, it may be appropriate to use different thresholds for score cutoff when interpreting results from each program. As indicated in Fig. 5.2, not all spectra with this score represent good matches (A), but some do (B). PEAKS results at this probability threshold could provide poor (C) or good (D) matches. For comparison of the programs in this study, however, a score cutoff of 15 was chosen. This value was chosen by observations as seen in Fig. 5.2, and a desire not to dismiss visually good matches that may have a low score. The use of crustaceanNP+LiDB for the database also led to complications because the LiDB sequences are short, representing just the active NP. Sequences in the crustaceanNP database are long preprohormones. Thus, matches to these longer proteins were scored better than similar matches to the shorter active NP sequences. It is clear from these results that the methods for assigning scores to PSMs used by both programs may not be very accurate in describing the quality of matches to neuropeptides, and additional by-hand analysis of lowscoring matches will still be necessary. An alternative approach that may be successful is appending decoy proteins to the database, and choosing a score threshold based on the acceptable false discovery rate (FDR) using matches to the decoy proteins. Mascot and PEAKS are both capable of estimating a FDR in this manner, but with small databases that contain both neuropeptide and prepropeptide sequences, this approach may not lead to an accurate determination of FDR. Indeed, when searching what was determined to be the “best” database, LiDB+crustaceanNP, PEAKS did not return FDR values, and Mascot only estimated FDR at a score of 40. The score cutoff for which Mascot returns a FDR is based on the quality of the database, and one cannot ask the program to return a FDR for any given score, such as the score empirically observed to produce acceptable matches (15 in this case). Therefore, another approach to determining the number of random positive identifications of PSMs should be employed, although no appropriate statistical method currently exists. 221 5.4.1. Previously Known Crustacean Neuropeptides Identified by MS/MS in O. rusticus Several PSMs from this study mapped to neuropeptides previously observed in other decapod crustacean species using MS/MS. These include one B-type allatostatin (AST-B), two FMRFamidelike peptides (FLPs), myosuppressin, 16 orcokinins, orcomyotropin, and one SIFamide (see Table 5.1). PDH and CPRPs will be discussed separately, as both previously observed NPs and novel gene products were observed. PSMs may represent 1) fragments of mature peptides that are produced as a result of degradation or fragmentation that occurs before or after sample collection, 2) unique mature peptides created by different processing pathways, or 3) incompletely processed peptides. Therefore, the multitude of orcokinin sequences, most of which contain homologous sequences, could all represent unique molecules present in the animal with unique activities. Alternatively, they could all represent fragments of 8 individual orcokinin peptides (See Fig. 5.3.). For this reason, all PSMs are reported. Most NPs are present in both tissues at equal abundance. This is in agreement with previous findings in other crustacean species [20, 26-30]. Many of the identified sequences match those previously observed by MS/MS or Edman sequencing in the closely related crayfish Orconectes limosus. Two of the orcokinins (NFDEIDRSGFGFN and NFDEIDRSGFGFV) were previously identified in Orconectes limosus [22, 25]. A different form of orcomyotropin, FDAFTTGFamide, was previously found in the hindgut of O. limosus [22]. Several other studies have used MS/MS or Edman sequencing techniques to map NPs in O. limosus, including molt-inhibiting hormone (MIH) [21], A-type allatostatins [23], and CHH [24]. These peptides were not identified in this study. It would be unlikely to identify CHH or MIH using these techniques, as they are large peptides (8-9 kDa) that require multi-scale MS approaches for complete sequencing [31]. A typical approach would be digestion with a protease such as trypsin, followed by MS/MS analysis of the fragments and assembling them into the full peptide, called 222 bottom-up sequencing. That method was not employed in this study. The absence of A-type ASTs is unusual and may be due to the short sequences of these peptides and the fact that many are only singly charged. Software programs for DDA acquisition and peptide sequencing preferentially pick multiply charged precursors, as singly charged molecules present in a LC-ESI analysis of tissue extracts are typically lipids or small molecules as opposed to peptides. Scoring functions for both Mascot and PEAKS are biased to produce low scores for short peptides even with good matches. These lowscoring matches may have been discarded by the software. Manual de novo sequencing of MS/MS spectra that did not yield hits according to the software programs may be able to identify some of these smaller peptides. It is not surprising that studies of the related crayfish O. limosus have yielded some of the same NPs as many sequences can be expected to be similar, and the NPs that have been seen in O. limosus but not in this study are likely missing due to different experimental or data processing approaches. B-type allatostatins (AST-Bs), FMRFamide-like peptides (FLPs), SIFamides, myosuppressin, and several of the orcokinins observed have not been previously identified in the studies of Orconectes limosus, but have been observed in many other decapod crustacean species, including other crayfish [32-37], lobsters [38], and crabs [39-43]. Although not every species has the same set of NPs, members of each of these families have been found in species across the order Decapoda, and some have been found in more distantly related species as well. Of particular note is the orcokinin DFDEIDRSGFA. This has previously been identified in one other species, Callinectes sapidus, and may represent the product of deamidation of the typical N-terminal N residue, either in vivo or during sample preparation and analysis [29]. One possible explanation is that deamidation of this residue is a modification that occurs in vivo to change this orcokinin’s signaling properties. This method of signaling modulation is not without precedent [44]. However, the binding efficiencies of 223 NFDEIDRSGFA vs. DFDEIDRSGFA at their receptor have not been established; in fact the orcokinin receptor has yet to be identified. In this study, the presence of PSMs mapping to AST-B, FLPs, SIFamide, myosuppressin, and orcokinins with sequence similarity to those previously found in other crustacean species expands our understanding of the conservation of these NPs across species, and the high degree of homology may indicate that these NP families are critical to neurotransmission and neuroendocrine regulation in many organisms. Other NPs observed in some of these species but not identified in Orconectes rusticus may still be present. In addition to A-type ASTs, CHH, and MIH mentioned previously, C-type ASTs, allatotropins, crustacean cardioactive peptide (CCAP), corazonin, kinins, YRamide, RYamides, proctolin, pyrokinin, red pigment concentrating hormone (RPCH), and tachykinin-related peptides have also been observed in decapod crustacean species. Some PSMs mapped to the known sequences of these peptides or their preprohormones, but these matches represented portions of the sequence not previously identified by MS/MS in these species, and they will be discussed in a separate section. It is probable that the peptides not identified in this study but expected to be present in this species are 1) found in neural organs other than the brain and eyestalk, 2) small peptides not well studied with MS/MS interpretation software, 3) large peptides that must be processed enzymatically prior to MS/MS analysis for identification, 4) present in very low quantities, and/or 5) subject to extensive post-mortem degradation. Other ganglia typically analyzed by MS/MS for NP quantitation include the thoracic and abdominal ganglia, the commissural organs, the stomatogastric ganglion, the pericardial organ, and several large nerves. These were not analyzed in this study due to more extensive dissection requirements. In addition, a multi-faceted mass spectrometric approach has been demonstrated to provide more complete coverage of a species’ neuropeptidome [29, 31, 32, 38, 39, 42, 45-49]. Although several neuropeptide families were not found in this study, it is possible that they do 224 exist in O. rusticus; other experimental approaches may be able to identify them. Variation in NP expression profiles among species is also a possible explanation, and the absence of some NPs in Orconectes rusticus may be important to its physiology or behavior. For instance, it is possible that other less aggressive species may have NPs involved in social bonding, similar to oxytocin in mammals. Not having an NP with this function may be one reason for increased aggressiveness in O. rusticus. 5.4.2. Identification of PDH and CPRP Isoforms in Orconectes rusticus Five isoforms of CPRP and 3 of PDH have been previously identified in the related crayfish species Orconectes limosus [20], and the sequences of these peptides are compared to the PSMs obtained in this study in Figures 5.4 and 5.5 and Table 5.2. Two groups of NPs that have high sequence homology across many crustacean species are the CPRPs and PDHs. Due to the high degree of sequence similarity and overlap between known crustacean CPRPs and PDHs, it is difficult to determine exactly how many mature peptides could be generated from the obtained PSMs in this study. PSMs for previously known NPs and novel products of the genes for these peptides were observed in this study. The CPRPs observed all align to previously described Orconectes limosus CPRPs A, A*, and B (Fig. 5.4.). These precursors could be responsible for all observed PSMs. Extensive coverage of the CPRPs was achieved, and most sequences came from eyestalk extracts (Table 5.2). This is expected, due to the localization of CHH primarily in the sinus gland of the eyestalk. The absence of any mutated residues indicates that CPRPs are highly conserved between these two Orconectes species. Comparison to known CPRP sequences from other crustaceans [20, 26, 29, 42, 47, 50-61] also suggests a great deal of homology. Sequence conservation throughout evolutionary time may indicate that this peptide has an important biological function, instead of being simply a byproduct of CHH synthesis. This study of Orconectes rusticus identified 31 PSMs, 225 primarily from the sinus glands, that map to three distinct CPRPs that were previously identified in O. limosus and that have a high degree of homology to other crustacean CPRPs. Sequences for the pigment dispersing hormone (PDH) sequences are longer and more varied between species, although they are also more commonly found in the eyestalk. Several PSMs were made to peptides of the PDH family, as indicated in Fig. 5.5 and Table 5.2. In Fig. 5.5A, alignments are made to the N-terminal portion of the peptide, which contains a precursor-related peptide only seen by MS in a single previous study (Callinectes sapidus, pQELHVPEREA [29]). This portion of the sequence has been characterized primarily by genetic techniques until now. The presence of these PDH-precursor related peptides may indicate analysis of incompletely processed PDH, or may suggest that these compounds are signaling molecules in their own right. The pyroglutamic acid residue at the N-terminus and amidated C-termini are common PTMs seen in authentic NPs. Currently, only one full PDH gene in Orconectes limosus has been sequenced, and several PSMs identified in this study do not match with portions of this gene. Indeed, two PSMs match only to the PDH gene found in the Cladoceran crustacean Daphnia magna. By parsimony, a minimum of 10 unique sequences would be needed to cover PSMs that map to the N-terminal portions of PDHs. As indicated in Fig. 5.5B, although only one PDH gene has been identified in Orconectes, multiple active PDH peptides have been identified. These peptides may thus be products of an additional PDH gene or genes. Also shown in Fig.5.5B, one PDH sequence must come from a non-Orconectes PDH sequence, PDH-I of Marsupenaeus japonicus. This study mapped 17 PSMs to the N-terminus of PDH, but the currently described Orconectes PDH gene is insufficient to describe the variety of sequences observed here. In addition, two PSMs mapped to this portion of the PDH gene from the distantly related Daphnia magna. Finally, 4 different C-terminal PDH sequences corresponding to the three previously known O. limosus PDHs plus PDH-I from M. japonicus were observed with a total of 11 PSMs. This study 226 has greatly increased the number of known PDH sequences and further suggests that the C-terminal portion of the prepro-PDH gene may constitute a unique NP. 5.4.2. Neuropeptides Not Previously Identified by MS/MS in Crustaceans An additional 47 PSMs were made to 43 unique identified neuropeptide sequences (some PSMs mapped to the same identified peptide) (Table 5.3). These matches were made to compounds previously only identified by genetic means, and often in the portions of the sequence not typically associated with an active peptide. PSMs were found in the non-peptide regions of crustacean cardioactive peptide (CCAP), FMRFamide-like peptides, red pigment concentrating hormone (RPCH), orcokinin, SIFamide, and tachykinin. These may indicate new precursor-related peptide products of the same genes responsible for producing those well-known neuropeptides. An alternative explanation is that these fragments are left over after production of the genuine NP. Several PSMs mapped to crustacean hyperglycemic hormone (CHH) fragments, although too few were obtained to determine a partial sequence for this peptide in O. rusticus. Additional CPRP isoforms could be suggested by several matches to CPRPs other than those previously found in O. limosus. Matches to bursicon-α were found for the first time using MS/MS (alignment in Fig. 5.6). The presence of this peptide in crustacean genomes has previously been noted, but had not been found using MS/MS techniques [26, 47]. Four of these matches appear in the signal peptide region, which would presumably be cleaved post-translationally. Although the coverage of the bursicon-α gene is not high, these results are encouraging for further analysis, particularly targeted bottom-up proteomics approaches. Two overlapping PSMs were found for a calcitonin-like diuretic hormone (CLDH) from Homarus americanus [62]. They map to a region of the gene immediately preceding the putative mature peptide, which could create a precursor-related peptide. Other matches include crustacean female hormone, eclosion hormone, eyestalk peptide, an insulin-like peptide, and an oxytocin-neurophysin-like peptide. 227 Many of these peptides have multiple PSMs from multiple organs and/or analysis techniques, thus increasing the confidence of their assignments. These matches are not sufficient to positively state the presence of these peptides in Orconectes rusticus¸ mostly because they do not provide full coverage of sequences of the hormones in question. Many of these are large hormones that would need additional treatment prior to MS/MS analysis to map their sequences. These additional PSMs are intriguing and suggest that additional precursor-related peptides may be encoded by the same genes that encode known NPs, and that several NPs not previously detected with MS/MS may be present in O. rusticus. 5.4.3. Comparison of Data Sets Results for PSMs mapping to proteins or unidentified transcripts of the Daphnia pulex genome are presented in Appendix C. The D. pulex genome has already been subjected to genome mining for neuropeptide sequences [63, 64], but it is possible that some of these still unidentified proteins produce NPs that were missed in the previous studies. A number of spectra were assigned sequences by de novo sequencing only with PEAKS. The identities of these were interrogated by BLAST, and it was determined that no neuropeptides or hormones were found in this data set, although several PSMs mapped to unidentified D. pulex proteins, which may yet contain undiscovered NP sequences. This list is also included in Appendix C. Both software programs performed equally in terms of numbers of identifications in both brain and eyestalk (Fig. 5.7.). The number of PSMs made in eyestalk, however, was much greater than in the brain (Fig. 5.8.), and very few NPs overlapped. This is likely due to the presence of large NPs in the eyestalk, such as CPRPs and PDHs. Smaller NPs typically found in the brain, such as ASTs and FLPs, were not identified well due to problems mentioned earlier. Many NPs have a tissue-specific pattern of distribution. Interestingly, only CPRPs and PDHs had a clear family trend in tissue distribution—most were present in the eyestalk. It is likely that the addition of these two groups to the eyestalk tally led to 228 the overall identification of more NP sequences in this tissue, as CPRPs and PDHs had many PSMs. The distribution of PSMs to neuropeptide families is indicated in Fig. 5.9. As mentioned previously, each PSM may indicate a unique neuropeptide with biological activity, or they may map to fragments of authentic NPs. For this reason, the PSMs were assembled into the lowest number of possible NPs that could produce the observed peptides. The least possible number of NP precursors that could explain the NPs observed to match the 8 NP families for which identifications were made to previously observed compounds was determined by parsimony and the resulting peptides are indicated in Table 5.4. Twenty-three NPs are sufficient to explain these PSMs, although it is expected that this number is a very low estimation. 5.5. Conclusions and Future Work In this work, the neuropeptides present in the crayfish Orconectes rusticus were catalogued using MS/MS. This species is of particular interest to neuroscience, for the behavioral studies conducted in it, and ecology, for its impact on the environment as an invasive species. This list of NPs will be of aid for further studies into this organism, which will be useful in understanding how an organism responds to changing environmental conditions via neuroendocrine regulation, among other things. Two MS/MS interpretation approaches were used: database search using the program Mascot and de novo sequencing using the program PEAKS. Determining a score cutoff was difficult, as the algorithms that these programs use to score the probability of a match are not optimal for small peptides, as many NPs are, and small databases, such as the ones employed in this study. The mixture of large preprohormones and small active NPs in the combination database used may also have contributed to difficulties obtaining scores that accurately represent the quality of a PSM. It is suggested that a different set of criteria be used for databases containing only short sequences of active NPs than for protein/preprohormone databases. A lower score threshold, or even a different scoring 229 mechanism could be used to ensure that all NPs present are fairly assigned. Lowering the score threshold will increase the manual work, as spectra will have to be verified by hand, but development of a new algorithm to score short peptide matches to spectra requires expertise in statistical methods. This study identified at minimum one B-type allatostatin, 3 CPRPs, 2 FMRFamide-like peptides, 1 myosuppressin, 8 orcokinins, 1 orcomyotropin, 6 PDHs, and one SIFamide from previously identified NPs in the extracts of brain and eyestalk. Potentially novel PDHs were discovered, which may represent a new class of PDH-precursor related signaling molecules, which were first suggested by Hui and colleagues [29]. A total of 98 peptide-spectrum matches were made to NPs previously observed in other decapod crustacean species. PSMs for signaling molecules not previously identified in decapod crustaceans were also made. Other PSMs map to previously unobserved (by MS) sequence regions of CHHs, bursicon, FMRFamides, a crustacean female hormone, insulin-like hormones, calcitonin-like diuretic hormones (CLDHs), eclosion hormone, eyestalk peptides, an oxytocin-neurophysin-like hormone, red pigment concentrating hormone (RPCH), and tachykinins. These peptides warrant further study, as many have homology to mammalian NPs involved in critical physiology. 5.6. Respective Contributions Statement and Acknowledgments CMS designed and carried out the research and wrote the paper. ZL aided in sample acquisition and preparation. LL advised and revised the paper. Nicole Woodards and Weifeng Cao of the Li Lab are acknowledged for assisting in database preparation. Alex Latzka of the Vander Zanden Lab and coworkers at the Trout Lake Research Station, Department of Limnology are acknowledged for crayfish collection. The School of Pharmacy Analytical Instrumentation Center is acknowledged for access to the Mascot software. 5.7. Works Cited 1. Kravitz, E. A.; Huber, R. Aggression in invertebrates. Curr Opin Neurobiol. 2003, 13, 736-743. 230 2. Panksepp, J. B.; Yue, Z.; Drerup, C.; Huber, R. Amine neurochemistry and aggression in crayfish. Microscopy Research and Technique. 2003, 60, 360-368. 3. Dziopa, L.; Imeh-Nathaniel, A.; Baier, D.; Kiel, M.; Sameera, S.; Brager, A.; Beatriz, V.; Nathaniel, T. I. Morphineconditioned cue alters c-Fos protein expression in the brain of crayfish. Brain Research Bulletin. 2011, 85, 385395. 4. Huber, R. Amines and motivated behaviors: a simpler systems approach to complex behavioral phenomena. Journal of Comparative Physiology a-Neuroethology Sensory Neural and Behavioral Physiology. 2005, 191, 231-239. 5. Nathaniel, T. I.; Huber, R.; Panksepp, J. Repeated cocaine treatments induce distinct locomotor effects in Crayfish. Brain Research Bulletin. 2012, 87, 328-333. 6. Nathaniel, T. I.; Panksepp, J.; Huber, R. Drug-seeking behavior in an invertebrate system: Evidence of morphineinduced reward, extinction and reinstatement in crayfish. Behavioural Brain Research. 2009, 197, 331-338. 7. Panksepp, J. B.; Huber, R. Ethological analyses of crayfish behavior: a new invertebrate system for measuring the rewarding properties of psychostimulants. Behavioural Brain Research. 2004, 153, 171-180. 8. Acquistapace, P.; Hazlett, B. A.; Gherardi, F. Unsuccessful predation and learning of predator cues by crayfish. J Crustacean Biol. 2003, 23, 364-370. 9. Tierney, A. J.; Lee, J. Spatial Learning in a T-Maze by the Crayfish Orconectes rusticus. J Comp Psychol. 2011, 125, 31-39. 10. Weisbord, C. D.; Callaghan, D. T.; Pyle, G. G. Associative learning in male rusty crayfish (Orconectes rusticus): conditioned behavioural response to an egg cue from walleye (Sander vitreus). Can J Zool. 2012, 90, 85-92. 11. Hazlett, B. A. Conditioned reinforcement in the crayfish Orconectes rusticus. Behaviour. 2007, 144, 847-859. 12. Hazlett, B. A.; Acquistapace, P.; Gherardi, F. Differences in memory capabilities in invasive and native crayfish. J Crustacean Biol. 2002, 22, 439-448. 13. Guo, K.; Ji, C.; Li, L. Stable-isotope dimethylation labeling combined with LC-ESI MS for quantification of aminecontaining metabolites in biological samples. Anal Chem. 2007, 79, 8631-8638. 14. Olden, J.; McCarthy, J.; Maxted, J.; Fetzer, W.; Vander Zanden, M. The rapid spread of rusty crayfish (Orconectes rusticus) with observations on native crayfish declines in Wisconsin (U.S.A.) over the past 130 years. Biol Invasions. 2006, 8, 1621-1628. 15. Strayer, D. L. Alien species in fresh waters: ecological effects, interactions with other stressors, and prospects for the future. Freshwater Biol. 2010, 55, 152-174. 16. Olden, J. D.; Vander Zanden, M. J.; Johnson, P. T. J. Assessing ecosystem vulnerability to invasive rusty crayfish (Orconectes rusticus). Ecol Appl. 2011, 21, 2587-2599. 17. Hein, C. L.; Roth, B. M.; Ives, A. R.; Vander Zanden, M. J. Fish predation and trapping for rusty crayfish (Orconectes rusticus) control: a whole-lake experiment. Can J Fish Aquat Sci. 2006, 63, 383-393. 18. Tetzlaff, J. C.; Roth, B. M.; Weidel, B. C.; Kitchell, J. F. Predation by native sunfishes (Centrarchidae) on the invasive crayfish Orconectes rusticus in four northern Wisconsin lakes. Ecol Freshw Fish. 2011, 20, 133-143. 19. Hein, C. L.; Vander Zanden, M. J.; Magnuson, J. J. Intensive trapping and increased fish predation cause massive population decline of an invasive crayfish. Freshwater Biol. 2007, 52, 1134-1146. 20. Bulau, P.; Meisen, I.; Schmitz, T.; Keller, R.; Peter-Katalinic, J. Identification of neuropeptides from the sinus gland of the crayfish Orconectes limosus using nanoscale on-line liquid chromatography tandem mass spectrometry. Mol Cell Proteomics. 2004, 3, 558-564. 21. Bulau, P.; Okuno, A.; Thome, E.; Schmitz, T.; Peter-Katalinic, J.; Keller, R. Characterization of a molt-inhibiting hormone (MIH) of the crayfish, Orconectes limosus, by cDNA cloning and mass spectrometric analysis. Peptides. 2005, 26, 2129-2136. 22. Dircksen, H.; Burdzik, S.; Sauter, A.; Keller, R. Two orcokinins and the novel octapeptide orcomyotropin in the hindgut of the crayfish Orconectes limosus: identified myostimulatory neuropeptides originating together in neurones of the terminal abdominal ganglion. J Exp Biol. 2000, 203, 2807-2818. 23. Dircksen, H.; Skiebe, P.; Abel, B.; Agricola, H.; Buchner, K.; Muren, J. E.; Nassel, D. R. Structure, distribution, and biological activity of novel members of the allatostatin family in the crayfish Orconectes limosus. Peptides. 1999, 20, 695-712. 24. Kegel, G.; Reichwein, B.; Tensen, C. P.; Keller, R. Amino acid sequence of crustacean hyperglycemic hormone (CHH) from the crayfish, Orconectes limosus: emergence of a novel neuropeptide family. Peptides. 1991, 12, 909-913. 25. Stangier, J.; Hilbich, C.; Burdzik, S.; Keller, R. Orcokinin: a novel myotropic peptide from the nervous system of the crayfish, Orconectes limosus. Peptides. 1992, 13, 859-864. 26. Ma, M.; Bors, E. K.; Dickinson, E. S.; Kwiatkowski, M. A.; Sousa, G. L.; Henry, R. P.; Smith, C. M.; Towle, D. W.; Christie, A. E.; Li, L. Characterization of the Carcinus maenas neuropeptidome by mass spectrometry and functional genomics. General and Comparative Endocrinology. 2009, 161, 320-334. 231 27. Fu, Q.; Goy, M. F.; Li, L. Identification of neuropeptides from the decapod crustacean sinus glands using nanoscale liquid chromatography tandem mass spectrometry. Biochem Biophys Res Commun. 2005, 337, 765-778. 28. Hsu, Y. W. A.; Messinger, D. I.; Chung, J. S.; Webster, S. G.; de la Iglesia, H. O.; Christie, A. E. Members of the crustacean hyperglycemic hormone (CHH) peptide family are differentially distributed both between and within the neuroendocrine organs of Cancer crabs: implications for differential release and pleiotropic function. Journal of Experimental Biology. 2006, 209, 3241-3256. 29. Hui, L.; Cunningham, R.; Zhang, Z.; Cao, W.; Jia, C.; Li, L. Discovery and characterization of the Crustacean hyperglycemic hormone precursor related peptides (CPRP) and orcokinin neuropeptides in the sinus glands of the blue crab Callinectes sapidus using multiple tandem mass spectrometry techniques. J Proteome Res. 2011, 10, 4219-4229. 30. Fu, Q.; Kutz, K. K.; Schmidt, J. J.; Hsu, Y. W.; Messinger, D. I.; Cain, S. D.; de la Iglesia, H. O.; Christie, A. E.; Li, L. Hormone complement of the Cancer productus sinus gland and pericardial organ: an anatomical and mass spectrometric investigation. J Comp Neurol. 2005, 493, 607-626. 31. Jia, C.; Hui, L.; Cao, W.; Lietz, C. B.; Jiang, X.; Chen, R.; Catherman, A. D.; Thomas, P. M.; Ge, Y.; Kelleher, N. L.; Li, L. High-definition De Novo Sequencing of Crustacean Hyperglycemic Hormone (CHH)-family Neuropeptides. Mol Cell Proteomics. 2012. 32. Dickinson, P. S.; Stemmler, E. A.; Barton, E. E.; Cashman, C. R.; Gardner, N. P.; Rus, S.; Brennan, H. R.; McClintock, T. S.; Christie, A. E. Molecular, mass spectral, and physiological analyses of orcokinins and orcokinin precursorrelated peptides in the lobster Homarus americanus and the crayfish Procambarus clarkii. Peptides. 2009, 30, 297317. 33. Skiebe, P. Neuropeptides in the crayfish stomatogastric nervous system. Microsc Res Tech. 2003, 60, 302-312. 34. Skiebe, P.; Dreger, M.; Borner, J.; Meseke, M.; Weckwerth, W. Immunocytochemical and molecular data guide peptide identification by mass spectrometry: orcokinin and orcomyotropin-related peptides in the stomatogastric nervous system of several crustacean species. Cell Mol Biol (Noisy-le-grand). 2003, 49, 851-871. 35. Skiebe, P.; Dreger, M.; Meseke, M.; Evers, J. F.; Hucho, F. Identification of orcokinins in single neurons in the stomatogastric nervous system of the crayfish, Cherax destructor. J Comp Neurol. 2002, 444, 245-259. 36. Yasuda, A.; Yasuda-Kamatani, Y.; Nozaki, M.; Nakajima, T. Identification of GYRKPPFNGSIFamide (crustaceanSIFamide) in the crayfish Procambarus clarkii by topological mass spectrometry analysis. Gen Comp Endocrinol. 2004, 135, 391-400. 37. Yasuda-Kamatani, Y.; Yasuda, A. Identification of orcokinin gene-related peptides in the brain of the crayfish Procambarus clarkii by the combination of MALDI-TOF and on-line capillary HPLC/Q-Tof mass spectrometries and molecular cloning. Gen Comp Endocrinol. 2000, 118, 161-172. 38. Ma, M.; Chen, R.; Sousa, G. L.; Bors, E. K.; Kwiatkowski, M. A.; Goiney, C. C.; Goy, M. F.; Christie, A. E.; Li, L. Mass spectral characterization of peptide transmitters/hormones in the nervous system and neuroendocrine organs of the American lobster Homarus americanus. Gen Comp Endocrinol. 2008, 156, 395-409. 39. Hui, L.; Xiang, F.; Zhang, Y.; Li, L. Mass spectrometric elucidation of the neuropeptidome of a crustacean neuroendocrine organ. Peptides. 2012, 36, 230-239. 40. Ma, M.; Bors, E. K.; Dickinson, E. S.; Kwiatkowski, M. A.; Sousa, G. L.; Henry, R. P.; Smith, C. M.; Towle, D. W.; Christie, A. E.; Li, L. Characterization of the Carcinus maenas neuropeptidome by mass spectrometry and functional genomics. Gen Comp Endocrinol. 2009, 161, 320-334. 41. Ma, M.; Sturm, R. M.; Kutz-Naber, K. K.; Fu, Q.; Li, L. Immunoaffinity-based mass spectrometric characterization of the FMRFamide-related peptide family in the pericardial organ of Cancer borealis. Biochem Biophys Res Commun. 2009, 390, 325-330. 42. Ma, M.; Wang, J.; Chen, R.; Li, L. Expanding the Crustacean neuropeptidome using a multifaceted mass spectrometric approach. J Proteome Res. 2009, 8, 2426-2437. 43. Szabo, T. M.; Chen, R.; Goeritz, M. L.; Maloney, R. T.; Tang, L. S.; Li, L.; Marder, E. Distribution and physiological effects of B-type allatostatins (myoinhibitory peptides, MIPs) in the stomatogastric nervous system of the crab Cancer borealis. J Comp Neurol. 2011, 519, 2658-2676. 44. Lorand, L.; Graham, R. M. Transglutaminases: crosslinking enzymes with pleiotropic functions. Nat Rev Mol Cell Biol. 2003, 4, 140-156. 45. Chen, R.; Hui, L.; Cape, S. S.; Wang, J.; Li, L. Comparative Neuropeptidomic Analysis of Food Intake via a Multifaceted Mass Spectrometric Approach. ACS Chem Neurosci. 2010, 1, 204-214. 46. Chen, R.; Jiang, X.; Conaway, M. C.; Mohtashemi, I.; Hui, L.; Viner, R.; Li, L. Mass spectral analysis of neuropeptide expression and distribution in the nervous system of the lobster Homarus americanus. J Proteome Res. 2010, 9, 818-832. 232 47. Ma, M.; Gard, A. L.; Xiang, F.; Wang, J.; Davoodian, N.; Lenz, P. H.; Malecha, S. R.; Christie, A. E.; Li, L. Combining in silico transcriptome mining and biological mass spectrometry for neuropeptide discovery in the Pacific white shrimp Litopenaeus vannamei. Peptides. 2010, 31, 27-43. 48. Wang, J.; Jiang, X.; Sturm, R. M.; Li, L. Combining tissue extraction and off-line capillary electrophoresis matrixassisted laser desorption/ionization Fourier transform mass spectrometry for neuropeptide analysis in individual neuronal organs using 2,5-dihydroxybenzoic acid as a multi-functional agent. J Chromatogr A. 2009, 1216, 82838288. 49. Wang, J.; Zhang, Y.; Xiang, F.; Zhang, Z.; Li, L. Combining capillary electrophoresis matrix-assisted laser desorption/ionization mass spectrometry and stable isotopic labeling techniques for comparative crustacean peptidomics. J Chromatogr A. 2010, 1217, 4463-4470. 50. Choi, C. Y.; Zheng, J.; Watson, R. D. Molecular cloning of a cDNA encoding a crustacean hyperglycemic hormone from eyestalk ganglia of the blue crab, Callinectes sapidus. Gen Comp Endocrinol. 2006, 148, 383-387. 51. Davey, M. L.; Hall, M. R.; Willis, R. H.; Oliver, R. W.; Thurn, M. J.; Wilson, K. J. Five Crustacean Hyperglycemic Family Hormones of Penaeus monodon: Complementary DNA Sequence and Identification in Single Sinus Glands by Electrospray Ionization-Fourier Transform Mass Spectrometry. Mar Biotechnol (NY). 2000, 2, 80-91. 52. Dircksen, H.; Bocking, D.; Heyn, U.; Mandel, C.; Chung, J. S.; Baggerman, G.; Verhaert, P.; Daufeldt, S.; Plosch, T.; Jaros, P. P.; Waelkens, E.; Keller, R.; Webster, S. G. Crustacean hyperglycaemic hormone (CHH)-like peptides and CHH-precursor-related peptides from pericardial organ neurosecretory cells in the shore crab, Carcinus maenas, are putatively spliced and modified products of multiple genes. Biochem J. 2001, 356, 159-170. 53. Fu, Q.; Christie, A. E.; Li, L. Mass spectrometric characterization of crustacean hyperglycemic hormone precursorrelated peptides (CPRPs) from the sinus gland of the crab, Cancer productus. Peptides. 2005, 26, 2137-2150. 54. Hsu, Y. W.; Messinger, D. I.; Chung, J. S.; Webster, S. G.; de la Iglesia, H. O.; Christie, A. E. Members of the crustacean hyperglycemic hormone (CHH) peptide family are differentially distributed both between and within the neuroendocrine organs of Cancer crabs: implications for differential release and pleiotropic function. J Exp Biol. 2006, 209, 3241-3256. 55. Hsu, Y. W.; Weller, J. R.; Christie, A. E.; de la Iglesia, H. O. Molecular cloning of four cDNAs encoding preprocrustacean hyperglycemic hormone (CHH) from the eyestalk of the red rock crab Cancer productus: identification of two genetically encoded CHH isoforms and two putative post-translationally derived CHH variants. Gen Comp Endocrinol. 2008, 155, 517-525. 56. Ma, M.; Chen, R.; Ge, Y.; He, H.; Marshall, A. G.; Li, L. Combining bottom-up and top-down mass spectrometric strategies for de novo sequencing of the crustacean hyperglycemic hormone from Cancer borealis. Anal Chem. 2009, 81, 240-247. 57. Marco, H. G.; Hansen, I. A.; Scheller, K.; Gade, G. Molecular cloning and localization of a cDNA encoding a crustacean hyperglycemic hormone from the South African spiny lobster, Jasus lalandii. Peptides. 2003, 24, 845851. 58. Stemmler, E. A.; Hsu, Y. W.; Cashman, C. R.; Messinger, D. I.; de la Iglesia, H. O.; Dickinson, P. S.; Christie, A. E. Direct tissue MALDI-FTMS profiling of individual Cancer productus sinus glands reveals that one of three distinct combinations of crustacean hyperglycemic hormone precursor-related peptide (CPRP) isoforms are present in individual crabs. Gen Comp Endocrinol. 2007, 154, 184-192. 59. Toullec, J. Y.; Serrano, L.; Lopez, P.; Soyez, D.; Spanings-Pierrot, C. The crustacean hyperglycemic hormones from an euryhaline crab Pachygrapsus marmoratus and a fresh water crab Potamon ibericum: eyestalk and pericardial isoforms. Peptides. 2006, 27, 1269-1280. 60. Tsai, K. W.; Chang, S. J.; Wu, H. J.; Shih, H. Y.; Chen, C. H.; Lee, C. Y. Molecular cloning and differential expression pattern of two structural variants of the crustacean hyperglycemic hormone family from the mud crab Scylla olivacea. Gen Comp Endocrinol. 2008, 159, 16-25. 61. Wilcockson, D. C.; Chung, S. J.; Webster, S. G. Is crustacean hyperglycaemic hormone precursor-related peptide a circulating neurohormone in crabs? Cell Tissue Res. 2002, 307, 129-138. 62. Christie, A. E.; Stevens, J. S.; Bowers, M. R.; Chapline, M. C.; Jensen, D. A.; Schegg, K. M.; Goldwaser, J.; Kwiatkowski, M. A.; Pleasant, T. K.; Shoenfeld, L.; Tempest, L. K.; Williams, C. R.; Wiwatpanit, T.; Smith, C. M.; Beale, K. M.; Towle, D. W.; Schooley, D. A.; Dickinson, P. S. Identification of a calcitonin-like diuretic hormone that functions as an intrinsic modulator of the American lobster, Homarus americanus, cardiac neuromuscular system. The Journal of Experimental Biology. 2010, 213, 118-127. 63. Christie, A. E.; McCoole, M. D.; Harmon, S. M.; Baer, K. N.; Lenz, P. H. Genomic analyses of the Daphnia pulex peptidome. Gen Comp Endocrinol. 2011, 171, 131-150. 233 64. Dircksen, H.; Neupert, S.; Predel, R.; Verleyen, P.; Huybrechts, J.; Strauss, J.; Hauser, F.; Stafflinger, E.; Schneider, M.; Pauwels, K.; Schoofs, L.; Grimmelikhuijzen, C. J. Genomics, transcriptomics, and peptidomics of Daphnia pulex neuropeptides and protein hormones. J Proteome Res. 2011, 10, 4478-4504. 5.8. Figures Figure 5.1. Reproducibility of PSMs in A) Mascot and B) PEAKS. Incomplete overlap is likely a result of instrumental variability in the selection of precursors for MS/MS, although it could be due to biological variation and false-positive PSMs. A) Mascot Eyestalk 1 33 Both 19 Eyestalk 2 17 B)PEAKS Eyestalk 1 36 Brain 1 9 Both 11 Brain 2 12 P Both 15 Eyestalk 2 27 Brain 1 21 Both 9 Brain 2 16 Figure 5.2. Comparison of peptide spectrum matches (PSMs) from Mascot and PEAKS. A) A PSM obtained with Mascot with a score of 18 that shows a poor degree of peak-fragment correlation. B) A PSM obtained with Mascot with a score of 15 showing good correlation between MS/MS peaks and peptide fragments. C) A PSM from PEAKS with a score of 15.07 that matches poorly. D) A PSM from PEAKS with a score of 16.85 that matches well. Some additional high-intensity peaks without assignments map to neutral losses 234 A) B) C) D) 235 Figure 5.3. Alignment of previously identified orcokinin sequences. PSMs for orcokinin peptides that have previously been observed via MS/MS in decapods are aligned. Peptides are marked with a “u” in the Unique column cannot be explained as fragments of the other peptides. By parsimony, these 16 PSMs can thus be mapped to 8 unique peptides. PSM DFDEIDRSGFA NFDEIDR NFDEIDRS NFDEIDRSGFA NFDEIDRSGFG NFDEIDRSGFGF NFDEIDRSGFGFA NFDEIDRSGFGFN FDEIDRSGFGFN EIDRSGFGFN NFDEIDRSGFGFV NFDEIDRSSFG NFDEIDRTGFamide NFDEIDRTGFG NFDEIDRTGFGF NFDEIDRTGFGFH Unique u u u u u u u u 236 Figure 5.4. Alignment of CPRP PSMs. A lowercase m indicates an oxidized methionine. Orconectes limosus CPRP sequences are taken from reference and compared to sequences obtained in this study. Red indicates the residue that distinguishes CPRP A* from A, and green indicates the residue that distinguishes B from the others. The column to the far right shows which CPRP each fragment maps to. For those fragments that occur in multiple CPRPs, it is left blank. Olim CPRP A Olim CPRP A* Olim CPRP B RSVEGSSRMERLLSSGSSSSEPLSFLSQDHSVS RSVEGSSRMERLLSSGSSSSEPLSFLSQDQSVS RSVEGSSRMERLLSSGSSSSEPLSFLSQDQSVN RSVEGSSRMERLLSSGSSSSEPLSFLSQDQSVS RSVEGSSRmERLLSSGSSSSEPLSFLSQDQSVS RSVEGSSRMERLLSSGSSSSEPLSFLSQDQSVN RSVEGSSRMERLLSSGSSSSEPLSFLSQDQSVNa mide RSVEGSSRMERL RSVEGSSRMERLL GSSRMERLL MERLLSSGSSSSEPLSFLSQDQSVS RLLSSGSSSSEPLSFLSQDQSVS RLLSSGSSSSEPLSFL RLLSSGSSSSEPLSF RLLSSGSSSSEPL RLLSSGSSSSEPLS LLSSGSSSSEPLS LLSSGSSSSEPLSF LLSSGSSSSEPLSFL LLSSGSSSSEPLSFLSQ LLSSGSSSSEPLSFLSQDQSVS LLSSGSSSSEPLSFLSQDQSVN LSSGSSSSEPLSFL LSSGSSSSEPLSFLSQDQSVS SSGSSSSEPLSFLSQDHSV SSGSSSSEPLSFLSQDQSVS SGSSSSEPLSFL SGSSSSEPLSFLSQDQSVS SGSSSSEPLSFLSQDQSVN SSSSEPLSFLSQDQSVS SSSEPLSFLSQDQSVS SSEPLSFLSQDQSVS SEPLSFLSQDQSVS PLSFLSQDQSVS CPRP CPRP CPRP CPRP A* A* B B CPRP A* CPRP A* CPRP A* CPRP B CPRP A* CPRP A CPRP A* CPRP CPRP CPRP CPRP CPRP CPRP CPRP A* B A* A* A* A* A* 237 Figure 5.5. Alignment of PDH sequences. A lowercase m indicates an oxidized methionine. A lowercase l in a PSM sequence indicates a L or I residue, since they cannot be distinguished using MS. A lowercase p in front of a Q indicates the pyroglutamic acid form of Q. A) Alignment of sequences to the N-terminal portion of the precursor. Active peptides formed from this portion are sometimes called PDH precursor-related peptides. Reference sequences are given first, followed by several rows of PSMs from the data. Underlined sequences differ from the literature sequence for O. limosus. Peptides that are unique, in that they cannot be contained in any other PSM, are indicated with a letter u in the right-most column. In the Daphnia magna sequence, the peptides that are matched are highlighted in bold. B) Alignment to the C-terminal portion. This is the active PDH peptide. Sequences for PDH A, B, and C from O. limosus are indicated as Olim PDH-A, -B, -C. Sequence for Marsupenaeus japonicus PDH I is indicated as Marj PDH-I. Red indicates residues that differ between Olim PDH-A and –B. Green indicates the residue that differs between Olim PDH-B and C. Blue indicates the residues that differ between Olim PDH-A and Marj PDH-I. PSMs follow the reference sequences, with modified residues indicated by colors, and the precursor that PSM requires indicated in the righthand column. For PSMs where multiple precursors are suggested, this column is left blank. 238 A. N-Terminus Orconectes limosus PDH precursor (S59496.1) 1-56 MRSAMVVLVLVAMVAVFTRAQE LKYPEREVVAELAAQIYGWPGSLGTMAGGPHKRNSELINSILGLPKVMNEAGRR MVAmSlQlT u pQElKYPEREVVAEl pQElKYPEREVVAElA pQElKYPEREVVAElAA pQElKYPEREVVAElAAQ u SSlKYFEREVVSElAAQll SSlKYFEREVVSElAAQllR SSlKYFEREVVSElAAQllRV u EAVATlAAHllKVVCAPlEGAGGlPHKamide u VV lAAVlTQG u AAQllRVAQGPSAFVAGPHK u VAQGPSAFVAGPHK AQGPSAFVAGPHK lGTmAGGPHKamide u AGGlPHKRNamide u EYllKFamide u DlNPTEKamide u Daphnia magna PDH precursor (CAA72409.1) MHQLSAKLSHFSIALLVLFVSLTMDVESAPAATSSINRPEAQLSIQEMEKFLEGLTRYLHRQRLEQQRMH QPQAKSLMSEEVTYDPDAIDRAILGEMAAPLDAERSSSVELANNSMSHARPPTNNKWPWSLSNFERIEDQ NVKQRQPYGK SlTmDVES SlmSEEVamide B. C-Terminus Olim PDH-A NSELINSILGLPKVMNEAamide Olim PDH-B NSELINAILGSPTLFGEVamide Olim PDH-C NSELINAILGSPTLMGEVamide Marj PDH-I NSELINSLLGIPKVMTDAamide NSEllNSllGlPKVMNEAamide NSEllNSllGl NSEllNSllGlP NSEllNSllGlPK SEllNSllGlPamide SllGlPKVm SllGlPKVMNEAa llGlPKVMNEAa llGlPKVmT NSEllNAllGSPTlM NSEllNAllGSPTl Olim PDH-A Marj PDH-I Olim PDH-C 239 Figure 5.6. Alignment of PSMs with bursicon alpha sequences. PSMs are indicated by color with where they align on the bursicon alpha gene. Two overlapping PSMs are both indicated with red, and a third with some overlap is indicated in blue. Where those overlap, the sequence is colored purple. The underlined portion of the gene sequences is the signal peptide. A lowercase M indicates an oxidized methionine. Bα-Cs ACG50067.1 bursicon hormone alpha subunit [Callinectes sapidus] Bα-Cm ABX55995.1 bursicon alpha [Carcinus maenas] Bα-Hg ADI86242.1 bursicon alpha subunit [Homarus gammarus] Bα-Cs MNSNLTWAMVGAAVTVLVVIGVDVARADECSLRPVIHILSYPGCTSKPIPSFACQGRCTSYVQVSGSKLW Bα-Cm MMSSLPWTVVGAAVTVLVVIGVGVAQADECSLRPVIHILSYPGCTSKPIPSFACQGRCTSYVQVSGSKLW Bα-Hg MGGLSWVLMVLGVATVVWSDECSLTPVIHILSYPGCVSKPIPSFACQGRCTSYVQVSGSKLW AmVGAAVT mVGAAVTV VTVLVVIG TVVWSDECSLT Bα-Cs QTERSCMCCQESGEREAAITLNCPKPRPGEPKEKKVLTRAPIDCMCRPCTDVEEGTVLAQEIANFIQDSP Bα-Cm QTERSCMCCQESGEREAAITLNCPKPRPGEPKEKKVLTRAPIDCMCRPCTDVEEGTVLAQKIANFIQDSP Bα-Hg QTERSCMCCQESGEREASVVLNCPKVRKGEPTRRKILTRAPIDCMCRPCTDVEEGTVLAQEIANFIHDSP VLTRAPIDCMCRPCT Bα-Cs MDSVPFLK Bα-Cm MDSVPFLK Bα-Hg MGNVPFLK Figure 5.7. Overlap of the two software programs in PSM identifications. B. Brain A. Eyestalk PEAKS 28 Both 45 Mascot 23 PEAKS 26 Both 20 Mascot 12 240 Figure 5.8. Overlap in identifications between tissues. Mascot PEAKS Eyestalk 65 Both 8 Eyestalk 64 Brain 38 Both 4 Brain 28 Figure 5.9. PSMs mapped to each NP family in the brain and eyestalk tissues. Abbreviations in text. Total PSMs in eyestalk: 96. Total PSMs in brain: 58. Eyestalk Brain CPRP PDH Orcokinin CHH SIFamide Bursicon FMRFamide Myosuppressin AST-B Crustacean female hormone Insulin-like Orcomyotropin Calcitonin CCAP EH Eyestalk peptide Oxytocin-neurophysin RPCH Tachykinin Orcomyotropin SIFamide FDAFTTGFGHS GYRKPPFNGSIFa YAIAGRPRFamide pQDLDHVFLRFamide FMRFamide Myosuppressin Orcokinin TNWNKFQGSWamide EPLDHVFLRFamide NP Matching Sequence AST-B FMRFamide NP Family and Mascot scores are both -10logP. TNWNKFQGSWa EPLDHVFLR EPLDHVFLRF EPLDHVFLRFa YAIAGRPR pQDLDHVFLR pQDLDHVFLRF pQDLDHVFLRFa DFDEIDRSGFA EIDRSGFGFN FDEIDRSGFGFN NFDEIDR NFDEIDRS NFDEIDRSGFA NFDEIDRSGFG NFDEIDRSGFGF NFDEIDRSGFGFA NFDEIDRSGFGFN NFDEIDRSGFGFV NFDEIDRSSFG NFDEIDRTGFa NFDEIDRTGFG NFDEIDRTGFGF NFDEIDRTGFGFH FDAFTTGFGHS GYRKPPFN Sequence of PSM Eyestalk Brain PEAKS Score Mascot Score PEAKS Score Mascot Score 32.36 35.44 16.55 51.99 32.87 33.24 35.44 29.98 16.55 53.35 48.09 29.92 45.87 47.52 30.71 20.73 19.91 18.8 23.52 33.66 22.13 49.52 51.82 46.25 58.31 75.5 46.55 53.36 41.7 48.84 45.69 46.01 76.44 101.5 75.71 64.87 69.98 58.89 16.85 21.59 28.28 36.02 37.43 34.47 27.52 21.27 64.5 63.15 38.98 43.02 19.25 19.96 Table 5.1. PSMs that map to putative neuropeptides previously identified in decapod crustaceans. Abbreviations in text. PEAKS 241 GYRKPPFNG GYRKPPFNGSIF GYRKPPFNGSIFa KPPFNGSIF KPPFNGSIFa RKPPF RKPPFN RKPPFNG RKPPFNGSI RKPPFNGSIF RKPPFNGSIFa 49.1 53.8 36.15 17.94 27.45 21.18 23.42 30.71 56.9 64.97 21.04 20.16 30.91 39.46 38.42 23.7 33.66 21.41 17.23 242 NP Family CPRP GSSRMERLL LLSSGSSSSEPLS LLSSGSSSSEPLSF LLSSGSSSSEPLSFL LLSSGSSSSEPLSFLSQ LLSSGSSSSEPLSFLSQDQSVN LLSSGSSSSEPLSFLSQDQSVS LSSGSSSSEPLSFL LSSGSSSSEPLSFLSQDQSVS MERLLSSGSSSSEPLSFLSQDQSVS PLSFLSQDQSVS RLLSSGSSSSEPL RLLSSGSSSSEPLS RLLSSGSSSSEPLSF RLLSSGSSSSEPLSFL RLLSSGSSSSEPLSFLSQDQSVS RSVEGSSRM(O)ERLLSSGSSSSEPLSFLSQDQSVS RSVEGSSRMERL RSVEGSSRMERLL RSVEGSSRMERLLSSGSSSSEPLSFLSQDQSVN RSVEGSSRMERLLSSGSSSSEPLSFLSQDQSVNamide RSVEGSSRMERLLSSGSSSSEPLSFLSQDQSVS SEPLSFLSQDQSVS SGSSSSEPLSFL SGSSSSEPLSFLSQDQSVN SGSSSSEPLSFLSQDQSVS SSEPLSFLSQDQSVS Sequence of PSM Eyestalk PEAKS Score Mascot Score 16.43 25.83 33.61 43.97 24.56 64.89 34.29 51.49 58.65 83.14 51.41 109.36 110.22 21.4 45.31 33.81 116.9 151.69 64.49 82.22 40.65 46.53 63.06 94.94 70.99 92.77 54.37 59.16 92.46 100.88 47.02 28.97 15.38 81.38 71.45 129.83 38.04 16.35 21.71 24.69 87.6 73.48 28.58 20.08 Brain PEAKS Score Mascot Score Lowercase l indicates either a I or L residue, as these are isobaric. PEAKS and Mascot scores are both -10logP. Table 5.2. PSMs for CPRP and PDH. Abbreviations in text. M(O) indicates an oxidized methionine. pQ indicates pyroglutamate. 243 PDH SSGSSSSEPLSFLSQDHSV SSGSSSSEPLSFLSQDQSVS SSSEPLSFLSQDQSVS SSSSEPLSFLSQDQSVS AAQllRVAQGPSAFVAGPHK AGGlPHKRNamide AQGPSAFVAGPHK DlNPTEKamide EAVATlAAHllKVVCAPlEGAGGlPHKamide EYllKFamide lGTM(O)AGGPHKamide llGlPKVM(O)T llGlPKVMNEAamide MVAM(O)SlQlT NSEllNAllGSPTl NSEllNAllGSPTlM NSEllNSllGl NSEllNSllGlP NSEllNSllGlPK NSEllNSllGlPKVMNEAamide pQElKYPEREVVAEl pQElKYPEREVVAElA pQElKYPEREVVAElAA pQElKYPEREVVAElAAQ SEllNSllGlPamide SllGlPKVM(O) SllGlPKVMNEAamide SlM(O)SEEVamide SlTM(O)DVES SSlKYFEREVVSElAAQll SSlKYFEREVVSElAAQllR SSlKYFEREVVSElAAQllRV VAQGPSAFVAGPHK 28.04 18.82 30.95 15.39 34.24 24.73 33.91 25.29 31.32 25.71 38.13 41.45 65.89 76.48 102.29 19.06 19.79 48.58 36.65 69.16 45.91 21.47 32.27 50.76 82.63 67 16.23 16.66 26.61 29.27 24.71 34.91 86.67 35.58 38.6 15.74 19.11 47.45 21.08 55.2 51.55 17 21.09 16.58 29.83 19.03 36.46 37.37 16.36 25.71 244 19.05 Peptide Family Bursicon Bursicon Bursicon Bursicon Bursicon Calcitonin Calcitonin CCAP CCAP CCAP CHH CHH CHH CHH Accession Number ABX55995.1 ACG50067.1 ACG50067.1 ACG50067.1 ADI86242.1 ACX46386.1 ACX46386.1 ABB46292.1 BAF34909.1 BAF34909.1 AAD45236.1 ACN65120.1 ACS35346.1 AER27833.1 Bursicon hormone alpha subunit [Callinectes sapidus] Bursicon hormone alpha subunit [Callinectes sapidus] Bursicon hormone alpha subunit [Callinectes sapidus] Bursicon alpha subunit [Homarus gammarus] Prepro-calcitonin-like diuretic hormone [Homarus americanus] Prepro-calcitonin-like diuretic hormone [Homarus americanus] Crustacean cardioactive peptide [Homarus gammarus] Crustacean cardioactive peptide [Procambarus clarkii] Crustacean cardioactive peptide [Procambarus clarkii] Hyperglycemic hormone-like [Metapenaeus ensis] Crustacean hyperglycemic hormone [Panulirus homarus] Crustacean hyperglycemic hormone isoform 1 [Rimicaris kairei] Crustacean hyperglycaemic hormone [Ptychognathus pusillus] Bursicon alpha [Carcinus maenas] Description starting with LL are from the in-house database LiDB. PAGHPLEKRQamide HPSGLAALTASH SPPPAGRTATVIGCSVNVSTTYamide pEAASPGASSPWVEHR KLWEQLQ AGPLAKRDIG TPHTQPR TRLGHSIIRANELEKF LTRLGHSIIRANELEKFVRSSGSA TVVWSDECSLT VLTRAPIDCMCRPCT M(O)VGAAVTV AM(O)VGAAVT VTVLVVIG PSM Sequence 15.04 22.2 17.91 15.02 19.46 19.38 21.51 15.38 18.99 PEAKS PEAKS 37.72 Brain Eyestalk Mascot 18.17 18.3 58.81 28.86 Mascot known NP genes. M(O) indicates an oxidized methionine. Pyroglutamate residues are indicated as pE or pQ. Accession numbers Table 5.3. PSMs for NPs and hormones not previously identified with MS/MS in decapods. Some map to precursor regions of VVlAAVlTQG 245 CHH CHH CHH CHH CHH CHH CPRP CPRP CPRP CPRP Crustacean female hormone Crustacean female hormone Crustacean female hormone EH Eyestalk peptide Eyestalk peptide FMRFamide FMRFamide Insulin-like AFD28272.1 AFG16934.1 AFM29133.1 LL206 O15981.1 O97384.1 AAM21927.1 AAS46643.1 ABS01332.1 LL343 ADO00266.1 AF112986_1 EFX80896.1 ADA67878.1 BAE06263.1 AF112986_1 AFK81936.1 ADO00266.1 ADO00266.1 CHH AFD28272.1 FLRFamide precursor protein B [Procambarus clarkii] Putative sulfakinin-like peptide [Daphnia pulex] Insulin-like androgenic gland hormone precursor [Penaeus Eyestalk peptide [Jasus edwardsii] Hyperglycemic hormone [Pandalopsis japonica] Crustacean hyperglycmic hormone [Portunus pelagicus] Crustacean hyperglycemic hormone Hoa-CHH-A (pCHH-A[pQ61V132amide]) [Homarus amerianus] Crustacean hyperglycemic hormone 5; Pej-SGP-V [Marsupenaeus japonicus] Crustacean hyperglycemic hormone 2; Pm-SGP-II [Penaeus monodon] Crustacean hyperglycemic hormone precursor [Pachygrapsus marmoratus] Crustacean hyperglycemic hormone precursor [Macrobrachium rosenbergii] Prepro-crustacean hyperglycemic hormone [Galathea strigosa] CHH precursor-related peptide Capr CPRP II [13-38] [Cancer productus] Crustacean female hormone [Callinectes sapidus] Crustacean female hormone [Callinectes sapidus] Crustacean female hormone [Callinectes sapidus] Eclosion hormone [Amphibalanus amphitrite] Eyestalk peptide [Jasus edwardsii] CHH protein [Scylla paramamosain] CHH protein [Scylla paramamosain] VGMLMVLSLT PVELGSSNPamide PAEYSRNVR TPARQR DVRQDGQTNamide PGLASLGGSTST QGPLPIQPVamide ERQIQGP DGGYIDLSENamide GDLPGGLVHamide LSFMPEHP RIEKLLST DRSLFGKL SSPVASLIRG LVAGASSAGT LDDLLLSDV APM(O)QGYGTEamide EEHVDIR pEGAAHPLEKRamide LVACIAMATLPQTQamide 20.02 20.84 17.36 15.07 21.73 15.63 19.2 21.37 30.15 28.32 16.77 18.55 17.93 15.44 19.84 18.19 19.29 16.15 18.05 22.81 246 Insulin-like Insulin-like Orcokinin Orcokinin Orcokinin Oxytocinneurophysin RPCH RPCH SIFamiderelated peptide Tachykinin Tachykinin Tachykinin EFX76240.1 EFX76240.1 Q9NL83.1 Q9NL83.1 Q9NL83.2 ADD24046.1 ADQ73633.1 Q867W1.1 BAC82426.1 BAC82426.1 BAC82426.1 ADN95181.1 Insulin-like EFX70023.1 Insulin/IGF/relaxin-like peptide 2 [Daphnia pulex] Insulin/IGF/relaxin-like peptide 1 [Daphnia pulex] Insulin/IGF/relaxin-like peptide 1 [Daphnia pulex] Orcomyotropin/orcokinin precursorlike peptide [Procambarus clarkii] Orcomyotropin/orcokinin precursorlike peptide [Procambarus clarkii] Orcomyotropin/orcokinin precursorlike peptide [Procambarus clarkii] Oxytocin-neurophysin 1 [Lepeophtheirus salmonis] Red pigment concentrating hormone precursor [Scylla paramamosain] Red pigment concentrating hormone precursor [Scylla olivacea] GYRKPPFNGSIF-amide; FRP1_PROCL [Procambarus clarkii] Preprotachykinin [Procambarus clarkii] Preprotachykinin [Procambarus clarkii] Preprotachykinin [Procambarus clarkii] monodon] HFDDESEIDAYIQAL HFDDESEIDAY AGMDSELETLL AGGDSLYEPGK PPGSSSGDSCGP SVGGAPGGVVPSSPGSSSGDSamide QYKQCSSCGP VYVPRYIANLY VYVPRYIAN MTAQM(O)FTIALLLSLS VTPAEP M(O)IIPSTVGRCWM(O) SLGLTTTAATPNamide 51.8 17.89 77.24 38.47 38.23 18.39 21.23 45.72 22.85 18.49 18.54 18.11 87.4 37.25 16.8 45.89 29.9 22.28 22.03 247 2 1 8 1 6 Myosuppressin Orcokinin Orcomyotropin PDH Least PossibleLowest Number of Identified NPs that Can Explain Observed PSMs 1 3 FMRFamide-like peptides AST-B CPRP NP Family TNWNKFQGSWamide RSVEGSSRMERLLSSGSSSSEPLSFLSQDHSVS RSVEGSSRMERLLSSGSSSSEPLSFLSQDQSVS RSVEGSSRMERLLSSGSSSSEPLSFLSQDQSVN EPLDHVFLRFamide YAIAGRPR pQDLDHVFLRFamide DFDEIDRSGFA NFDEIDRSGFA NFDEIDRSGFGFA NFDEIDRSGFGFN NFDEIDRSGFGFV NFDEIDRSSFG NFDEIDRTGFamide NFDEIDRTGFGFH FDAFTTGFGHS MVAmSlQlT pQElKYPEREVVAElAAQ SSlKYFEREVVSElAAQllRV EAVATlAAHllKVVCAPlEGAGGlPHKamide VVlAAVlTQG Sequences methionine. Lowercase l’s indicate either I or L residues. Pyroglutamic acid is indicated as pQ or pE. required due to sequence variations being present at different parts of the overall sequence. A lowercase m indicates an oxidized Table 5.4. NPs required to explain all observed PSMs, and sequences of unique PSMs. There are more unique sequences than NPs are 248 SIFamide 1 AAQllRVAQGPSAFVAGPHK lGTmAGGPHKamide AGGlPHKRNamide EYllKFamide DlNPTEKamide SlTmDVES SlmSEEVamide NSELINSILGLPKVMNEAamide NSELINAILGSPTLFGEVamide NSELINAILGSPTLMGEVamide NSELINSLLGIPKVMTDAamide GYRKPPFNGSIFamide 249 250 Chapter 6: Development of Methods for Neuropeptide Quantitation with DataIndependent MS/MS Modified from Schmerberg, C. M. and Li, L. Rapid Commun. Mass Spec. [in preparation] 6.1. Abstract Quantitation using mass spectrometry (MS) and tandem MS (MS/MS) is not always straightforward due to a number of factors, most notably run-to-run variability and variability of some aspects of MS (primarily in ionization efficiency) based on molecular properties. Specialized methods for highly accurate quantitation using MS and MS/MS have been developed, but these typically require isotopic labeling and/or prior method optimization for the analytes of interest. Therefore, most experimental strategies for samples of unknown composition employ two steps and two separate instrument runs, one for identification of the components of the sample, followed by one for quantification of these identified components [1]. A new method of MS/MS data acquisition, called data-independent acquisition (DIA), has been developed to allow simultaneous identification and quantification of unknown samples. This method differs from typical MS/MS in that it fragments all ions concurrently, instead of selecting single ions for fragmentation. The precursor and fragment ions can be aligned to each other by tracking their retention times, charge states, and masses. The fragment ions can be quantified, which yields quantitative information about their precursors, in a scheme similar to selectedreaction-monitoring (SRM) methods [2]. In this chapter, an open-source, vendor-neutral software designed for SRM quantitation (Skyline) [3] is applied to highly multiplex reaction monitoring (all reactions monitoring, ARM) quantitation of neuropeptides (NPs) using DIA MS/MS data. This is the first work demonstrating use of this software for DIA MS/MS quantitation, and the first to quantify endogenous peptides from DIA data as opposed to larger proteins with multiple 251 peptide fragments. After establishment of the quantitation method, proof-of-principle was established by quantifying NPs in neuroendocrine tissues of the blue crab Callinectes sapidus after feeding. 6.2. Introduction Tandem mass spectrometry (MS/MS) is a highly useful analytical tool both for identification of molecules and their quantification. However, it typically cannot conduct both identification and quantification simultaneously, because different mass spectrometer (MS) parameters are typically required for each type of experiment. Notable exceptions exist, such as spectral counting and tandem mass tags, but those methods have disadvantages that will be detailed in following sections. There are several reasons different parameters would be required for identification and quantification, but unifying feature is that mass filters are typically used for both approaches, albeit in different ways. These mass filters either prevent monitoring multiple fragments in MRM-type experiments, which is required for identification, or prevent enough measurements of fragment ion intensities to be obtained across a given peak (i.e. too few data points/peak), which is required for quantitation. Data-independent analysis (DIA) MS employs no mass filter but produces both precursor and fragment ions. This increases the complexity of data interpretation, but it is possible to quantitate the product ions of specific precursors in a pseudo-multiple reaction monitoring (pMRM), or all-reactions monitoring (ARM), manner, as well as to determine identity in a single run. For further information in MS-based quantitation, a review is available [1]. 6.2.1. Methods for Quantitation by MS/MS The “gold standard” of quantitative MS/MS, in terms both of quantitation accuracy and sensitivity, is selected-reaction-monitoring (SRM). This is typically conducted in a liquid 252 chromatography-triple quadrupole mass spectrometer setup. The robustness of SRM lies in that it is essentially counting the number of ions with a given precursor mass and a given fragment mass that travel through the instrument to the detector, by using the first and last quadrupoles as mass filters. The first quadrupole permits only the precursor molecule to enter the second quadrupole, where collision/fragmentation occurs, and the third quadrupole permits only a given fragment produced by that fragmentation reaction to pass through to the detector (See Fig. 6.1). Multiple compounds can be quantified by cycling the quadrupoles through a given set of precursor and fragment masses over time, which is called multiple reaction monitoring (MRM), and if the retention times of analytes of interest are also known, the instrument can carry out a scheduled SRM at that time, as another method to permit multiplexed quantitation. The behavior of a given fragment during and between runs in this environment is highly reproducible, and the number of transitions monitored at any given time is limited so that enough data can be collected to obtain a Gaussian peak profile (usually >6 points), when ion intensity is plotted against time. The intensity of the analyte(s) of interest is usually tracked against that of an internal standard, for relative or absolute (with calibration curves) quantitation. A vendor-neutral, open-source software for MRM experiments has been developed by researchers at the University of Washington (Skyline) [3]. 6.2.2. Methods for Identification by MS/MS In identification, the selection of precursors for fragmentation is typically carried out in a scheme called data-dependent acquisition (DDA). MS1 scans are conducted, permitting all analytes to go to the detector, and from that data, several high-intensity precursors are selected for fragmentation. Each precursor is then sequentially selected by the first mass analyzer and fragmented, followed by detection of all of its fragment ions. The number of precursors that will 253 be monitored is dependent on the experimental parameters and the speed and resolution of the instrument. Using a high resolution time-of-flight (TOF) analyzer for the MS2 scan, only a handful of precursors can be monitored at a time because the separation of ions is relatively slow compared to the speed at which precursors elute from an ultrahigh performance liquid chromatography (UPLC) column, typically used prior to MS ionization. The MS2 spectrum that results and its associated precursor can be interpreted to determine the identity of that precursor. Typically, only high-intensity ions are selected as precursors, to provide higher-quality MS2 spectra. In addition, only a few scans of each precursor-fragment ion combination are acquired, because with high-quality MS2 spectra, only a few spectra are required to provide sequence information. Therefore, not enough data points are obtained for SRM-style quantitation. Occasionally, the intensity of an ion in the MS1 spectrum over time can be determined from DDA MS/MS experiments, but that is only possible with faster-scanning lower-resolution instruments such as ion traps, and the specificity is reduced compared to MS2 ion monitoring. This can be combined with MS1-level quantification strategies including isotope-coded affinity tags (ICAT), chemical modification with isotopic reagents, and incorporation of heavy amino acids, as well as label-free quantitation employing internal standards [1]. Although these can be used for quantitation based on the MS1 scan concurrent with identification based on MS2 scans, the quantitation is not as accurate. 6.2.3. Methods for Simultaneous Quantitation and Identification by MS/MS Alternative methods exist for identification and quantitation in a single run. For MS/MS experiments with high scan rates, the number of times an ion is selected for MS/MS analysis can be used as a measure of its abundance, called its spectral count [1]. This method is most robust for quantification at the protein level based on multiple peptides identified, and is not applicable 254 for endogenous neuropeptide (NP) study. A NP-compatible approach employs isotopic MS2 fragment ions produced by tags that are isobaric at the MS1 level. One example of this is DiLeu labels developed by the Li laboratory [4]. These tags use isotopes to generate characteristic fragment ions of given masses, and the relative intensities of these ions can be used for quantification. Finally, several transitions can be monitored with full fragment scans in some high-speed instruments, allowing for simultaneous identification and quantification of those predefined transitions with Skyline SRM software [5]. Although several methods for simultaneous quantification and identification exist, they are not optimal for experiments attempting to discover and quantify endogenous peptides, such as NPs. A new method of MS data acquisition, termed data-independent acquisition (DIA), has been developed for simultaneous identification and quantification of molecules (Fig. 6.2). This method is fundamentally different from other MS/MS methods in that mass filtering is not employed, either at the MS1 level as in DDA MS/MS, or at both MS1 and MS2 levels as in MRM. By eliminating mass filtering at the MS1 level (or using large isolation windows), multiple precursors are fragmented simultaneously and their fragments enter the second mass analyzer simultaneously. This results in a highly complex MS/MS spectrum as the fragments cannot initially be assigned to precursors for sequence identification. However, precursors and fragments can be aligned by software using their retention times and mass to charge ratios (m/z). By eliminating filtering at the MS2 level, the intensities of all of these fragments over time can be determined for quantification. By matching precursors and fragments, then quantifying the fragments of a given precursor, all fragmentation reactions are monitored (all reactions monitoring, ARM). Every peptide with several transitions observed can thus be quantified. 255 Several instrument vendors have incorporated this technique into new instruments with slight variations, and the one described and used here is Waters MSE. This useful technique for simultaneous identification and quantification has been conducted in a variety of samples at the protein level [2, 6-9], with each protein having multiple peptides detected. However quantification has not previously been done with analytes at the peptide level. In addition, the SRM software Skyline has not previously been demonstrated in quantitation of DIA MS/MS data. In this work, the MSE strategy for DIA MS/MS is employed to collect data for quantitation and identification of peptides, using a protein digest as internal standard and Skyline software for data analysis. This technique is found to be linear for several model NP standards, and proof-of-principle experiments establish its ability to quantify NPs in tissue extracts from the blue crab Callinectes sapidus after feeding. This tool is highly useful for function-driven discovery neuropeptidomics experiments, in which an experimental manipulation is employed to drive expression of NPs, whose identity may not already be known. These NPs are then identified and given a putative function based on the manipulation. 6.3. Materials and Methods More detailed information for this section is found in Appendix A. 6.3.1. Sample Preparation and Instrumental Analysis A set of standards to test the linearity of the method was prepared using myoglobin digest and peptide standards obtained from American Peptide Co. (Sunnyvale, CA). Equine skeletal myoglobin (ERA, Colden, CO) was digested with trypsin (bovine pancreas, Sigma-Aldrich, St. Louis, MO) following a published procedure [10]. Myoglobin was dissolved in 100 mM NH4HCO3 at 0.5 mg/mL. Trypsin was dissolved in the same solution at 1 µg/mL and added to the myoglobin solution at an enzyme: substrate ratio of 1:10. This was then diluted 1:1 with 256 methanol and placed in a water bath at 37°C for 45 min. The reaction was stopped by adding icecold acetic acid to a final concentration of 5%. The sample was then spun at 15,100 x g for 5 min. The peptide standards used were α-melanocyte stimulating hormone (MSH), bradykinin (BK), crustacean cardioactive peptide (CCAP), Homarus americanus FMRFamide-like peptide (FLP) I, H. americanus FLP II, somatostatin-14 (SMT), and substance P (SP). These were spiked into a solution of 1x diluted crab saline (220 mM NaCl; 5.5 mM KCl; 6.5 mM CaCl2; 13 mM MgCl2; 5 mM HEPES, pH 7.4) with 0.05% formic acid and 1.88 µM myoglobin digest at the following concentrations: 100, 50, 37.5, 25, 12.5, 6.3, and 2.5 nM. These samples were not subjected to pre-injection purification. 6.3.2. Animals and Feeding Experiments Female blue crabs (Callinectes sapidus), were obtained from a local market. Details of their housing and dissection are detailed in Appendix A. For animals from the fed condition, they were presented with raw fish 1hr prior to dissection. It was confirmed that they had eaten by the presence of food in their stomachs. Unfed animals were exposed to food but did not eat, also evidenced by stomach contents. Four animals were used per condition. Animals were weighed following anesthesia immediately prior to dissection. Sinus glands (SGs), brains, and pericardial organs (POs) were isolated and stored in tubes containing 50 µL of acidified methanol (90:9:1 MeOH: H2O: acetic acid) on ice. Tissues were kept at -20°C until extraction. Following extraction (pellet extracted 3 x 100 µL acidified methanol: grind, sonicate, spin 5 min at 16,100 x g) and solvent evaporation (Savant SC10 Speedvac, Thermo Scientific), PO samples were using C18 Ziptips (EMD Millipore, Billerica, MA), eluted in 15 µL of 50/50/0.1 (ACN/H2O/FA), diluted with 5 µL.0.1% FA, and added to 2 µL of 56.4 µM myoglobin digest in LC vials. SG 257 and brain samples were resuspended in 30 µL of 0.1% FA in H2O and placed in LC vials, along with 2 µL of 56.4 µM myoglobin digest internal standard. 6.3.3. Instrumental Analysis Ultrahigh performance liquid chromatography (UPLC)-MSE analysis was conducted on a Waters nanoAcquity UPLC coupled to a Waters Synapt G2 mass spectrometer (Waters, Milford, MA). Samples (2 µL) were trapped on a preconcentration column and desalted on-line. This column was then put in line with a C18 reversed-phase column (BEH130 C18, 1.7 µm, 75 µm x 100 mm, Waters, Milford, MA). The outlet of this column was connected to a fused silica capillary with a pulled tip of internal diameter ~5 µm (Sutter Instrument Company, Novato, CA). This was used as the ESI inlet. A 75 min reversed-phase run with solvent A as 0.1% FA in water and B as 0.1% FA in ACN was used. The instrument was operated in data-independent MS/MS mode with the high energy scan having a voltage ramp from 25 to 65 V and Glu-fibrinopeptide was infused for lockspray calibration. 6.3.4. Data Analysis 6.3.4.1. Linearity Experiment Data Analysis The sequences of the neuropeptide standards (CCAP: PFC(-H)NAFTGC(-H)amide, FLP I: SDRNFLRFamide, FLP II: TNRNFLRFamide, α-MSH: acetyl-SYSMEHFRWGKPVamide, somatostatin-14: AGC(-H)KNFFWKTFTSC(-H)amide, bradykinin: RPPGFSPFR, and substance P: RPKPQQFGGLMamide, with (-H) indicating a C in a disulfide bond that loses a hydrogen) and 9 myoglobin tryptic peptides (FDKFK, FKHLK, TEAEMK, TEAEM(Ox)K, TEAEMKASEDLK, ALEFLR, ALEFRNDIAAK, NDIAAK, and ELGFQG, with M(O) indicating an oxidized methionine) were input into the Skyline SRM software. Modifications were added to peptides manually. A non-specific enzyme type was used, with cleavage at every 258 amino acid, and 9 missed cleavages permitted, the most allowed. Skyline does not have a “no enzyme” option built in and this is the closest approximation possible. Most NPs are shorter than 9 amino acids, so this should be acceptable. In addition, Skyline will quantify peptides that do not meet its filter criteria if they are input manually and the exception to the filters is noted by the user. The iRT function with retention time predictor was used, calibrated off of results from Mascot (Matrix Science, London, UK) identifications of equine myoglobin tryptic peptides in a previous LC run of the myoglobin digest alone under the same conditions. The 2+ and 3+ precursors were monitored to the 4 most abundant 1+ and 2+ b and y ions, and in some cases (CCAP, somatostatin-14) the precursor ion itself was also monitored. MS/MS filtering parameters were set to DIA with MSe isolation, and 10,000 resolving power. Peak integration was checked manually for a good match to the most abundant transitions and agreement with the predicted retention times. Between runs, which were all conducted sequentially on the same day, variation of less than 0.2 min was observed in retention time. Peak areas were exported to Microsoft Excel. All transitions for each peptide were summed, and the 9 myoglobin peptides were summed together. Peptide peak areas were normalized to the total myoglobin internal standard peak area. Data was plotted and linear regressions were conducted. 6.3.4.2. Feeding Data Analysis Data files were converted to .pkl files by Protein Lynx Global Server (PLGS, Waters, Milford, MA). They were then searched against the database LiDB+crustaceanNP as defined in Ch. 5 with Mascot (Matrix Science, London, UK). The resulting data files were used to generate a list of observed NPs that had match scores greater than 15. A Skyline SRM method was created that quantified all identified crab NPs from a previously published study on crab NP changes in feeding [11]. The sequences for these were pasted in from a Microsoft Excel spreadsheet and the 259 proper post-translational modifications (PTMs) were added manually. Myoglobin tryptic peptides quantified in the linearity experiments were also quantified in this document. Peaks identified using Mascot were also added to the list of NPs to be quantified, with the appropriate PTMs. The same retention time prediction was used, and again peaks were checked manually for proper integration. However, all predicted b and y ions were monitored instead of just 4. If an ion was observed to have a large amount of interference in its channel, it was removed. Peak areas were exported to Microsoft Excel. Peptide peak areas were compiled and normalized to the myoglobin peak area and the crab’s weight. From 8 crabs, the average weight ±standard error of the mean was 190.0 ±10.3 g with the largest crab at 248.2 g and the smallest at 162.2 g. There was no difference in weight between the fed and unfed groups, using a paired Student’s t test (p=0.832). These normalized peak areas were compared using a paired Student’s t test and graphed in Microsoft Excel. 6.5. Results and Discussion 6.5.1. Linearity for Quantification of Neuropeptide Standards First, it should be noted that poor or no signal was observed for somatostatin-14 in any sample, likely due to degradation of the standard over years. No results are presented for this NP. All other standards were present in sufficient quantity for the peak to be integrated. Plotting the normalized peak areas for the NP standards versus the concentrations and performing linear regressions yielded fits with a high degree of linearity (Fig. 6.3). Values for the goodness of fit for these regressions range from 0.93 to 0.989. These results indicate that although each analyte is a single peptide, accurate measurement is possible by making enough measurements of its intensity by analyzing several transitions for each of two charge states per peptide. The inclusion of an internal standard (ITSD) with multiple peptides that can be monitored allows for even more 260 measurements of its intensity to be made, and thus this value is more reproducivle. It could also be possible to use several peptide standards spiked in to the sample as an ITSD in a similar manner. These results suggest that absolute quantitation of known NPs following generation of calibration curves is also possible, although that is not carried out in this experiment. The margins of error obtained are sufficient for relative quantitation in function-driven discovery assays. It should also be noted that these linearity measurements were conducted on a single set of samples and a single technical replicate. For this to produce such highly linear results with no outliers, the quantitation method must be very rugged. This is similar to the type of run one might conduct in a function-driven discovery assay, where sample volume is limited and multiple technical replicates are too consumptive of precious sample and instrument time. Although only one experiment from a single day is shown here, an additional linearity study was conducted using mammalian NP standards as ITSD and crustacean NP standards as analytes, and a high degree of linearity was observed in this experiment as well. Therefore, day-to-day reproducibility and proof of the concept that multiple peptides (whether a digest or mixture of standards) can serve as an ITSD for reliable label-free quantitation of NPs in DIA MS/MS are established. 6.5.2. Quantitation of Neuropeptides in Fed and Unfed Callinectes sapidus The scheme used above with peptide standards was also used to quantify NPs from tissue extracts of fed and unfed blue crabs to demonstrate its applicability in a function-driven NP discovery experiment. Feeding is an important biological process known to be regulated by NPs, and previous work in the Jonah crab, Cancer borealis, has demonstrated changes in NP levels in the pericardial organ (PO) and brain using isotopic labeling with formaldehyde and deuterium 261 formaldehyde [11]. In the previous study, several members of the RFamide, RYamide, and tachykinin-related peptide (TRP) families, along with the peptides proctolin (RYLPT) and YRamide increased significantly in the brain following feeding. One peptide from the pyrokinin family decreased in the brain following feeding. In the PO, NPs were observed to decrease significantly, including many RFamides and one RYamide. A model in which NPs are secreted from the PO, a known neurosecretory organ, into the hemolymph, from where they diffuse to many target sites including the brain was one hypothetical explanation for the observed results. Replication of these results and expansion of the list of peptides differentially regulated by feeding was the goal of this portion of the study. Callinectes sapidus is a commercially important crab species whose neuropeptidome has recently been described [12, 13]. It was chosen for study due to the high degree of NP homology and its enthusiastic feeding behavior compared to C. borealis. The NPs previously quantified with respect to feeding mentioned above were quantified from the MSE data, along with 18 crab NPs identified by a Mascot search with the data against a crab neuropeptide database. These additional peptides were quantified to demonstrate the power of this technique to quantify any NP of interest and conduct untargeted methods. In turn, this same data could be re-analyzed for a NP found in a later study to potentially be of interest in feeding. The peak areas were normalized to a myoglobin digest ITSD as done previously and normalized to the weight of the crab to account for larger crabs typically having larger tissues. The results obtained for the NPs studied previously in feeding are indicated in Fig. 6.4. Some NPs are not detected, or are only detected at very low levels. In addition, different trends and numbers of significant values are observed than were previously reported from feeding studies in Cancer borealis [11]. Although the actual results are different, it is difficult to determine the 262 relevance of this finding, as different species of crabs and methods of MS were used. What can be determined is that most of the NPs were detected and quantified, and values obtained do not have a high degree of error. Statistically significant p<0.05) increases were seen in the SG for an orcokinin NFDEIDRSGFamide and a pyrokinin SGGFAFSPRLamide. Results obtained for quantification of the peptides identified by Mascot are shown in Fig. 6.5. Several of these NPs had statistically significant changes, and many were quantified with low error. Several of the NPs are modified forms of each other, and those that have similar sequences also show similar trends. Significant changes were observed for 11 NPs in the PO. This is likely due to better identification of a putative NP from this tissue because of its rich NP content. Proof-of-principle experiments demonstrate that this method can be used to quantify NPs from the crab resulting from experimental manipulation in an untargeted manner. 6.6. Conclusions and Future Work In this chapter, a method for quantitation of DIA MS/MS data in a pseudo-MRM manner was developed. It used an open-source, vendor-neutral software for accurate identification of precursors and fragments and integration of the resulting peaks. This quantitation method shows linearity for mixtures of peptide standards over the range 2.5-100 nM using a myoglobin digest as an internal standard. A proof-of-principle experiment was also conducted to quantify 28 NPs previously associated with feeding and 18 crab peptide sequences identified from the data by a database searching method. It is thus possible to take samples from any species for which a database of the proteins or peptides of interest exists and quantify compounds at the peptide level using this method. If a database does not exist, a homologous species can be used. This method will be of great use in targeted and untargeted functional analysis studies of NPs. 6.7. Works Cited 263 1. Bantscheff, M.; Lemeer, S.; Savitski, M. M.; Kuster, B. Quantitative mass spectrometry in proteomics: critical review update from 2007 to the present. Anal. Bioanal. Chem. 2012, 404, 939-965. 2. Gillet, L. C.; Navarro, P.; Tate, S.; Rost, H.; Selevsek, N.; Reiter, L.; Bonner, R.; Aebersold, R. Targeted Data Extraction of the MS/MS Spectra Generated by Data-independent Acquisition: A New Concept for Consistent and Accurate Proteome Analysis. Molecular & Cellular Proteomics. 2012, 11. 3. MacLean, B.; Tomazela, D. M.; Shulman, N.; Chambers, M.; Finney, G. L.; Frewen, B.; Kern, R.; Tabb, D. L.; Liebler, D. C.; MacCoss, M. J. Skyline: an open source document editor for creating and analyzing targeted proteomics experiments. Bioinformatics. 2010, 26, 966-968. 4. Xiang, F.; Ye, H.; Chen, R.; Fu, Q.; Li, L. N,N-dimethyl leucines as novel isobaric tandem mass tags for quantitative proteomics and peptidomics. Anal Chem. 2010, 82, 2817-2825. 5. Sherrod, S. D.; Myers, M. V.; Li, M.; Myers, J. S.; Carpenter, K. L.; MacLean, B.; MacCoss, M. J.; Liebler, D. C.; Ham, A.-J. L. Label-Free Quantitation of Protein Modifications by Pseudo Selected Reaction Monitoring with Internal Reference Peptides. Journal of Proteome Research. 2012, 11, 3467-3479. 6. Geromanos, S. J.; Vissers, J. P. C.; Silva, J. C.; Dorschel, C. A.; Li, G.-Z.; Gorenstein, M. V.; Bateman, R. H.; Langridge, J. I. The detection, correlation, and comparison of peptide precursor and product ions from data independent LC-MS with data dependant LC-MS/MS. Proteomics. 2009, 9, 1683-1695. 7. Levin, Y.; Hradetzky, E.; Bahn, S. Quantification of proteins using data-independent analysis (MSE) in simple andcomplex samples: a systematic evaluation. Proteomics. 2011, 11, 3273-3287. 8. Martins-de-Souza, D.; Guest, P. C.; Guest, F. L.; Bauder, C.; Rahmoune, H.; Pietsch, S.; Roeber, S.; Kretzschmar, H.; Mann, D.; Baborie, A.; Bahn, S. Characterization of the human primary visual cortex and cerebellum proteomes using shotgun mass spectrometry-data-independent analyses. Proteomics. 2012, 12, 500-504. 9. Mbeunkui, F.; Goshe, M. B. Investigation of solubilization and digestion methods for microsomal membrane proteome analysis using data-independent LC-MSE. Proteomics. 2011, 11, 898-911. 10. Li, F.; Schmerberg, C. M.; Ji, Q. C. Accelerated tryptic digestion of proteins in plasma for absolute quantitation using a protein internal standard by liquid chromatography/tandem mass spectrometry. Rapid Commun Mass Spectrom. 2009, 23, 729-732. 11. Chen, R.; Hui, L.; Cape, S. S.; Wang, J.; Li, L. Comparative Neuropeptidomic Analysis of Food Intake via a Multi-faceted Mass Spectrometric Approach. ACS Chem Neurosci. 2010, 1, 204-214. 12. Hui, L.; Cunningham, R.; Zhang, Z.; Cao, W.; Jia, C.; Li, L. Discovery and characterization of the Crustacean hyperglycemic hormone precursor related peptides (CPRP) and orcokinin neuropeptides in the sinus glands of the blue crab Callinectes sapidus using multiple tandem mass spectrometry techniques. J Proteome Res. 2011, 10, 4219-4229. 13. Hui, L.; Xiang, F.; Zhang, Y.; Li, L. Mass spectrometric elucidation of the neuropeptidome of a crustacean neuroendocrine organ. Peptides. 2012, 36, 230-239. 264 6.7. Figures Figure 6.1. Multiple reaction monitoring (MRM) MS/MS. In the first MS, the precursors are detected and a single one is selected by mass filters to enter the collision cell. There, it collides with gas and undergoes collision-induced dissociation (CID). A single fragment ion of interest is then sent to the second MS analyzer, where it is detected. Setting the mass filters to the appropriate m/z ratios leads to highly efficient and reproducible transfer of ions from MS 1 all the way to the detector. In the duty cycle of the instrument, the masses chosen in MS1 and MS2 can be rapidly changed (on the order of msec) to allow a different precursor and fragment to be analyzed. 265 Figure 6.2. Data-Independent Analysis (DIA) MS/MS. DIA MS/MS, called MSE on Waters QTOF instruments, has two spectral acquisition modes alternating throughout the entire acquisition. A low energy spectrum is generated by allowing just precursors to be analyzed. In a high energy spectrum, all precursors are subjected to CID simultaneously, and then all fragments are analyzed. Precursors and fragments are then aligned to each other based on a number of parameters, most notably their LC retention time. 266 Figure 6.3. Linearity of normalized peptide standard responses. The linearity of quantitation using multiple myoglobin tryptic peptides as internal standards was determined for 6 NP standards (abbreviations in text). Good linearity was observed in the 2.5-100 nM range (5 to 200 fmol on column) with ITSD at 1.88 µM. 0.04 Area/Area ITSD Area/Area ITSD RPPGFSPFRamide PFC(-H)NAFTGC(-H)amide 0.05 0.03 0.02 y = 0.0004x + 0.0081 R² = 0.9558 0.01 0 0 50 100 Concentration (nM) 150 0.07 0.06 0.05 0.04 0.03 0.02 0.01 0 y = 0.0006x + 0.0038 R² = 0.9625 0 150 Ac-SYSMEHFRWGKPVamide SDRNFLRFamide Area/Area ITSD Area/Area ITSD 0.05 0.03 0.025 0.02 0.015 0.01 0.005 0 0.04 0.03 0.02 y = 0.0002x + 0.0033 R² = 0.9644 0 50 100 Concentration (nM) y = 0.0004x + 0.0014 R² = 0.989 0.01 0 150 0 50 100 Concentration (nM) RPKPQQFFGLMamide 150 TNRNFLRFamide 0.1 Area/Area ITSD 0.05 Area/Area ITSD 50 100 Concentration (nM) 0.04 0.03 0.02 y = 0.0003x + 0.0145 R² = 0.93 0.01 0.08 0.06 0.04 y = 0.0008x + 0.0024 R² = 0.977 0.02 0 0 0 0 50 100 Concentration (nM) 150 50 100 Concentration (nM) 150 267 Figure 6.4. Changes in NP levels following feeding in three tissues. NPs quantified here were chosen based on their inclusion in a previous feeding study in the crab [11]. Means and standard errors of the means are indicated. Statistically significant changes between the two conditions are indicated with an asterisk (*p<0.05). These are on NFDEIDRSGFamide SGGFAFSPRFamide for the sinus gland. Four animals were used in each condition. and 268 0.0012 A) Pericardial Organ Fed Unfed 0.001 0.0008 0.0006 0.0004 0.0002 0 0.0012 B) Brain 0.001 Fed Unfed 0.0008 0.0006 0.0004 0.0002 0 0.006 C) Sinus Gland 0.005 Fed 0.004 Unfed * 0.003 * 0.002 0.001 0 -0.001 269 Figure 6.5. Changes in NP levels following feeding in three tissues. NPs quantified here were chosen based on an untargeted approach. Data was searched against a crab NP database and the resulting NPs with scores>15 that were not very large were quantified. Means and standard errors of the means are indicated. Statistically significant values are indicated with an asterisk (* p<0.05). Four animals were used for each condition. 270 0.007 A) Pericardial Organ * 0.006 * 0.005 Fed 0.004 Unfed 0.003 0.002 0.001 * * * * * * * * 0 0.02 0.018 0.016 0.014 0.012 0.01 0.008 0.006 0.004 0.002 0 * B) Brain Fed Unfed 271 0.05 0.045 C) Sinus Gland 0.04 0.035 0.03 0.025 0.02 0.015 0.01 0.005 0 Fed Unfed 272 Chapter 7: Application of Microdialysis and Data-Independent MS/MS Quantitation for Determination of Feeding-Related Neuropeptides Adapted from Schmerberg, C. M. and Li, L. ACS Chem. Neurosci. [in preparation] 7.1. Abstract Food consumption is an important behavior for sustaining the life of an animal that is regulated by an intricate array of neuropeptides (NPs) in many species. Although much is known about the identities of these NPs in mammals, the exact mechanisms of how these multiple chemical entities act on several feeding-related neuronal circuits to generate feeding behavior is currently unknown and difficult to study systematically in mammals. In order to better understand dynamic changes in NPs during feeding behavior, a decapod crustacean model system was employed in conjunction with novel microdialysis (MD) and data-independent acquisition (DIA) tandem mass spectrometry (MS/MS) quantitation and identification methods. The simpler neurochemistry and neural circuitry of the decapod crustacean make it a potentially useful model for elucidating the effects of some of these NPs, as well as a successful model animal for analytical chemistry method development. MD samples were collected from the Jonah crab, Cancer borealis, throughout the time course of feeding and analyzed for NP content in an untargeted quantitative MS/MS method that also provides peptide sequence information. A total of 28 NPs previously identified as potentially related to feeding in previous studies were quantified, and 5 of those showed statistically significant fold-changes: APQRNFLRFamide, GAHKNYLRFamide, GPRNFLRFamide, SENRNFLRFamide, and HIGSLYRamide. Some general trends were observed, particularly an immediate release into the hemolymph in the first 30 min after feeding, followed by an overshoot and return to baseline. A variety of NP level changes were observed in the time period much later after feeding, indicating that the initial roles 273 for these NPs may be similar, but the long-term roles may be different. This work demonstrates the success of this technique for quantifying NPs in a behavioral neuroscience experiment and establishes putative roles for several NPs in feeding behavior. 7.2 Introduction Neuropeptides (NPs) are known to play an important role in regulation of feeding behavior in a variety of species. Several NPs have been identified as modulating food consumption on a number of different levels in mammalian systems, including galanin, ghrelin, neuropeptide Y, melanin-concentrating hormone (MCH), alpha-melanocyte stimulating hormone (α-MSH), orexins, oxytocin, insulin, leptin, cholecystokinin (CCK), agouti-related protein (AgRP), neurotensin (NT), neuropeptide W, neuropeptide YY (PYY) and cocaine and amphetamine regulated transcript (CART) [1-3]. These NPs can act locally as neurotransmitters or at a distance in an endocrine manner [4, 5]. Some NPs are implicated in feeding-related modulation of the brain’s endogenous reward pathways, including circulating hormones and locally acting neuropeptides [1]. Several of the circulating hormones’ levels are controlled primarily by other organs, such as insulin from the pancreas, leptin from fat cells, and ghrelin from the stomach, and thus they and other NPs can serve as a way for the body to monitor energy levels and stores and alter feeding behavior accordingly [6, 7]. Many NPs have been identified in mammals as important in feeding behavior through a number of pathways. Although much progress has been made in understanding the effects of NPs on consumption behavior, the mechanisms by which a chemical signal acts to produce a given behavior are not well understood. One challenge lies in understanding behavior generation in the complex brains of traditional animal models; another lies in difficulties to accurately characterize NP changes on a physiologically relevant time scale. A number of diverse signals converge on 274 feeding behavior, including reward circuitry, stress signaling, metabolic state signals, somatosensation, and multiple hypothalamic subnuclei [2]. Put simply, the decision to eat is influenced not only by how “hungry” one is (stomach fullness and/or circulating glucose levels), but also how much one derives pleasure from food, environmental effects (stress, circadian), the type of food, whether other rewarding stimuli are present, etc. A major concern in this area of research is dissecting the pathways of feeding for energy maintenance and feeding for pleasure (homeostatic vs. hedonic), and it appears NPs are involved in both pathways in mammals [3]. Secondly, NPs can be difficult to quantify accurately due to the presence of alternative splice forms and antibody cross-reactivity, and accessing them on a physiologically relevant time scale can also be a challenge [8]. With a simple nervous system and more limited suite of behaviors, decapod crustaceans are potentially an attractive model for ingestive behavior. Food is highly motivating for crustaceans, and they can even be conditioned to press a lever to receive a food reward [9]. The crustacean stomatogastric ganglion (STG) and other ganglia are part of well-defined neural circuits responsible mainly for rhythmic movements of the gut in the crab. This well-established model for understanding rhythmic neuron firing and the effects of neuromodulators on activity has been extensively studied and can be modeled using computational neuroscience [10, 11]. Thus, putative NPs can be applied in this system experimentally, and models can be used to fit the experimental evidence. This, in turn, can provide information about the components of the circuit by which the NP acts, for instance, changing the current flux through one particular type of ion channel. Certain firing patterns in this circuit are associated with chewing or filtering of food, and thus can be correlated directly to feeding behavior. In addition to possessing this set of useful tools for understanding NP action on the circuit and ion channel level, the decapod 275 crustacean contains several organs that are rich in NPs that can be analyzed using well-developed mass spectrometry (MS)-based techniques [8]. Mass spectrometry-based techniques for NP analysis are more specific than antibodybased methods due to the ability to characterize an analyte based on its mass to charge ratio (m/z) and its fragments’ m/z. MS-based techniques can also be highly accurate and multiplexed, as evidenced in Chapter 6. Microdialysis (MD) sampling of NPs allows for collection of NPs concurrent with performance of normal behaviors, so that a close correlation can be made between changes in NP levels and behavioral changes. These techniques have been developed extensively in the decapod crustacean due to the reduced complexity of its neurochemical environment and neuronal circuits [12, 13]. In this chapter, techniques developed in previous chapters were applied to study NP changes in crabs during feeding behavior. Microdialysis was used to collect NPs from the hemolymph of Jonah crabs, Cancer borealis, before, during, and up to 3 hrs after feeding. These samples were then subjected to data-independent analysis (DIA) tandem MS (MS/MS) to enable determination and/or verification of NP sequences and highly specific quantity changes in the same instrument run. The resulting data was analyzed to quantify 28 NPs previously studied with regards to feeding in C. borealis [14]. The method was successful in identifying statistically significant changes in abundance at 30-min interval time points for 5 of the NPs quantified. These included the RFamide-like peptides APQRNFLRFamide, GAHKNYLRFamide, GPRNFLRFamide, and SENRNFLRFamide; and the YRamide HIGSLYRamide. Overall profiles for the NPs in hemolymph appear to have initial release events, followed by a return to baseline levels, although there are notable differences in some NPs. These changes suggest putative roles for these NPs in the neuroendocrine control of feeding behavior and physiological changes associated with ingestion. These findings are 276 consistent with previous tissue extract studies of feeding-related NPs in crabs. This is the first quantitative assessment of NPs in dialysates from the crab and demonstrates the utility of MD and MS/MS-based tools for behavioral neuroscience studies of NP function. 7.3. Methods Several of the methods have been described previously in Chapters 4 and 6, and are presented in detail in Appendix A. Only where the methods differ from previous chapters will they be discussed here. 7.3.1. In Vivo Microdialysis PAES (20kDa MWCO) probes (CMA Microdialysis) were implanted in Cancer borealis as indicated in Appendix A and Chapter 4. Following implantation, crabs with probes were kept separate from other animals with the aid of plexiglass dividers that the water could flow through and around. The outlet of the probe was connected to an automated refrigerated fraction collector. Animals were allowed to recover a minimum of 24 hrs prior to collecting samples that were used for analysis. Crabs typically would not eat until a minimum of 48 hrs had elapsed after surgery, so feeding experiments were conducted beginning the 2nd day after surgery. Crab saline was infused through the system at a flow rate of 0.5 µL/min, and the probe was allowed to run at this flow rate for a minimum of 30 min before any samples were collected to permit equilibration. In many cases, the experiment was started after the perfusate had been running at that rate for several hours and equilibration was already complete. If the rate had to be changed or the syringe refilled, 30 min of equilibration time immediately preceded collection of samples for the feeding experiment. The dead volume of the probe system from the tip of the probe to the collection tube was calculated based on information from the manufacturers of the probe, tubing, and fraction collector, and this was taken into account for timing collections correctly with 277 feeding. Crabs were fed white-fleshed raw fish (cod, tilapia, etc.), sold for human consumption, thawed and cut into small pieces immediately prior to use. Most crabs took and started eating the food immediately. If it took more than 2-3 min for the crab to start eating, the experiment was abandoned for that day. For crabs that did eat, excess food was always present so that the animal could feed until satiety was reached. Crabs typically stopped eating after 45-60 min. At t = 180 min, any remaining food was removed from the tank, even if the crab had to be moved to retrieve it. A second feeding experiment was conducted with each crab (three crabs total) a minimum of 24 hrs after the start of the first one. Crabs were not otherwise disturbed at any point during this process, and the room was consistently kept under red light to simulate night conditions. For every experiment, two baseline samples were taken from t = -60min to t = -30 min and t = -30min to t = 0 min. Collection tubes were changed every 30 min during the experiment by the refrigerated fraction collector until 8 samples were obtained, for a final time of 180 min post-feeding. Samples are graphed according to the time midway through their collection time. The resulting 15 µL samples were immediately acidified by adding 1.5 µL pure formic acid (FA) to them, bringing their acid content to approximately 10%, as this has been shown to reduce NP degradation [15]. They were then kept at -20°C until they could be analyzed. The feeding experiment was conducted twice for each crab with three crabs for a total of 6 replicates (3 biological x 2 experimental). 7.3.2. Instrumental Analysis Samples were placed in LC vials and an internal standard (1 µL of 56.4 µM myoglobin digest solution prepared as in Protocol A and Chapter 6) was added. Immediately prior to analysis on UPLC-MS/MS, 1 µL of a 0.91 M NH4HCO3 solution was added to increase the sample’s pH so as not to damage the UPLC column. Samples were analyzed as in Chapter 6 and 278 Protocol A, with a Waters nanoAcquity UPLC coupled via electrospray ionization (ESI) to a Waters Synapt G2 QTOF operating in MSE mode. Reversed-phase LC runs were 75 min long for each run. 7.3.3. Data Analysis A previously developed method for quantification of MSE data (Chapter 6) using Skyline software (University of Washington [16]) was used. A list of 28 NPs previously observed in feeding experiments (enumerated in Table 7.1.) [14] along with 9 myoglobin tryptic peptides (FDKFK, FKHLK, TEAEMK, TEAEM(O)K, TEAEMKASEDLK, ALEFLR, ALEFRNDIAAK, NDIAAK, and ELGFQG, with M(O) indicating an oxidized methionine) was selected for data extraction and integration. The iRT function with retention time predictor was used, calibrated off of results from Mascot (Matrix Science, London, UK) identifications of equine myoglobin tryptic peptides in a previous LC run of the myoglobin digest alone under the same UPLC conditions. The 2+ and 3+ precursors were monitored for their transitions to the most abundant 1+ and 2+ b and y ions. MS/MS filtering parameters were set to DIA with MSe isolation, and 10,000 resolving power. Peak integration was checked manually for a good match to the most abundant transitions and agreement with the predicted retention times. Variation was greater between sets of samples run on the instrument on different days than within a single day. All samples from a given MD experiment were run on the instrument on the same day, and analyzed in parallel. Peak areas were exported to Microsoft Excel. All transitions for each peptide were summed, and the 9 myoglobin peptides were summed together. Peptide peak areas were normalized to the total myoglobin internal standard peak area. The two baseline values were averaged for each experiment, and all values were normalized to this to correct for variability between animals and between different days. Thus, data represents fold-changes from baseline. 279 Results were plotted using Microsoft Excel and JMP statistical software (Version 9.0.2 SAS Institute, Inc., Cary, NC, USA). A few outlier data points were observed, likely due to signal interference in UPLC-MS/MS or improper peak picking, so data points of >5-fold or <0.2fold compared to baseline were removed. The most data points removed from any given peptide for any time point was 2, and no two experimental replicates were removed for a single crab, so there were always 3 biological replicates at every data point. The validity of repeated measures was determined using multivariate analysis in JMP. Briefly, a multiple analysis of variance (MANOVA) and multivariate repeated-measures analysis were conducted, and the validity of the measurements made was assessed based on these tests. A one way analysis of variance (ANOVA) comparison was conducted, followed by a Tukey-Kramer Honestly Significant Differences (HSD) all pairs test at an alpha value of 0.05. p-Values that were less than 0.05 were considered significant. Means and standard errors of the means (SEM) for all NPs were graphed in Microsoft Excel, with p-values from the Tukey-Kramer HSD test superimposed on the graph for significant values as indicated. 7.4. Results and Discussion The method was successful at generating reproducible measurements of NPs in multiple samples from 2 experimental replicates with each of 3 different animals. The distribution of outliers is illustrated by Fig. 7.1.A, which a scatterplot of all data points before outliers representing a 5-fold change were removed. Neuropeptide values that had such a high foldchange typically represented chromatograms where either the incorrect peak was integrated, or the peak was not observed at all, and as such are due to integration error. A total of 22 outlying points out of 1344 total, or 1.34%, were removed by this criterion, and the resulting scatterplot is shown in Fig. 7.1.B. The outliers were likely a result of poor peak detection due to interference 280 or integration of the incorrect peak. Removal of outliers did not reduce the total number of biological replicates, and although variability is still observed after removal of the egregious outliers, it is greatly reduced. To determine the validity of making multiple repeated measures in each experimental replicate, a multivariate repeated-measures analysis was conducted in JMP on a multiple analysis of variance test (MANOVA). Between experiments, no significant difference was found in NP profiles (prob>F 0.5688). Within experiments, sphericity was met, so multivariate F tests and adjusted univariate F tests were appropriate. These determined that overall, all interactions within subjects and of time*peptide were non-significant (F-values>0.75 for adjusted univariate F tests). However, the time factor did cause significant differences (p<0.0001 for the multivariate F test and univariate Greenhouse-Geisser- and Huynh-Feldt-adjusted F tests). These tests verify that there was no bias introduced by quantifying multiple peptides over time. The NP profiles were not significantly different between experiments, and overall interactions between NPs were non-significant. The observed variation between NP profiles was not dependent on experimental replicate and was instead related to time. Therefore, it is valid to quantify changes in these NPs over time and suggest that observed changes are a result of changing internal conditions in the animals over time. Of the 28 NPs quantified from 9 different families, significantly different values were obtained for time points in 5 of them, from the RFamide and YRamide families. Although all 14 of the 28 were observed to change in tissue extract studies, a more limited number of biological replicates were conducted for this experiment due to the difficulty of experimental procedures, expense of materials (MD probes), and availability of MS/MS instrument time (each experiment takes 12 hours of instrument time to run). Therefore, more variability is observed and fewer NPs 281 have large, significant changes. The repetition of this experiment should provide additional information about each data point, perhaps yielding more significantly changed values. For this reason, overall trends in NP concentration profile will also be discussed, even for values where statistical significance at the p<0.05 level has not been reached. No baseline values were statistically significant from each other. Detailed results and graphs for each peptide are included in this report. The peptides will be grouped and discussed by family. 7.4.1. RFamide-Like Peptides Peptides of the RFamide-like peptide family were altered the most. Of 10 peptides analyzed from this family, 4 had statistically significant changes. RFamide peptides are one of the largest groups of NPs in crustaceans, with 41 individual peptides of this family described in Cancer borealis (Chapter 2, [17, 18]). There are several known subgroups of RFamides, including the neuropeptide F (NPF)/short neuropeptide F peptides (sNPF), which carry a Cterminal motif of RXRFamide; the myosuppressins, which have HVFLRFamide at the Cterminus, and the sulfakinins, which have a sulfated tyrosine and the C-terminal sequence HMRFamide. Additional C-terminal sequences observed include FLRFamide, YLRFamide, and FVRFamide [19]. For the RFamide-like peptides, the hemolymph concentrations following feeding have a profile that can be largely described as release, an overshoot, and then eventual return to baseline. One peptide, GAHKNYLRFamide, had several significantly different values, summarized in Table 7.3. This peptide was first identified in 2006 from the POs and stomatogastric nervous systems of several Cancer species (C. borealis, C. productus, and C. magister) [20]. The value obtained in the first 30 min after feeding was greater than the values obtained 90-120 min, 120-150 min, and 150-180 min after feeding. The concentration at 60-90 282 min post-feeding was also different from the value 150-180 min after feeding. This is illustrated in Fig. 7.2. There are two clear phases in the hemolymph concentration of this peptide. Shortly after feeding, levels become higher than baseline by 20-25%. Much later, they drop back down to baseline or 10-15% less than baseline. This suggests that GAHKNYLRFamide is released during active feeding behavior and is returned to baseline levels shortly after feeding has stopped. Although the only statistical difference is between the time points indicated, the overall profile potentially shows an initial release that is stopped after 30 min (the lower value for 45min). A second release event may occur around 75 min, and after another 30 min period, the levels are low again, perhaps even lower than they were before feeding. The potentially reduced level at 45 min and the clearly reduced level starting at 105 min may indicate a depletion of PO stores of this peptide, supported by previous studies finding a statistically significant 43% decrease in its levels in this tissue 30-45 min after feeding [14]. In the same study, this peptide was also observed to increase in brain tissue during this time period, so it may be being bound and internalized with receptors as part of a desensitization mechanism in this tissue. This nonsignificant profile matches with a two-stage feeding behavior that is typically observed in this and other crab species, although it has not been studied systematically. Crabs typically start eating immediately and stop after 30 min, then wait another 15-25 min and start eating again in a shorter feeding period. This could mirror the two observed increases in GAHKNYLRFamide as they occur around the times that each of these feeding periods begins. In any case, the hemolymph levels remain elevated for a while after feeding begins, so its degradation must be slower than its release for a long period of time. This also suggests that GAHKNYLRFamide is not initiating feeding, but may be sustaining the behavior or serving as a whole-body signal that food is being taken in, as a mechanism of coordinating multiple organ systems. 283 Another RFamide with statistically significant changes in its levels after feeding is SENRNFLRFamide (Fig. 7.3, Table 7.4). This peptide has been found in many crustacean species, but was first identified in 2005 from the PO of the crab Cancer productus [21]. SENRNFLRFamide is more concentrated in hemolymph during the time 0-30 min after feeding compared to 90-120 min and 120-150 min after feeding. Therefore, a reduction in this peptide appears after the animal has stopped eating. The non-significant trend observed is a rapid increase immediately after feeding that drops back to baseline levels or lower after more than 90 min following feeding. This peptide could have similar functions as described for GAHKNYLRFamide. Reduced levels in the hemolymph at later times could also be due to depletion of stores, similar to observations in PO tissue extract studies [14]. This peptide did not have a significant increase in brain concentration after feeding in those studies, so the brain may not be its primary site of action, or it may be degraded by extracellular peptidases instead of internalized while bound to its receptor as part of desensitization. A third RFamide, GPRNFLRFamide, was significantly higher in concentration immediately after feeding when compared to more than 2 hrs after feeding (Fig. 7.4, Table 7.5). This peptide was initially identified in Cancer borealis after immunoaffinity enrichment of extracts from its POs [17]. The difference in values was around 80% of the baseline value, so this change is large in magnitude. In general, it appears that this peptide is released immediately after feeding and then may rapidly return to baseline or sub-baseline levels. Thus, it may be an indicator of the beginning of the feeding process that could trigger release of other compounds with more gradual action, or it could be active in initiating feeding behaviors. This peptide was detected in tissue studies, and showed a strong but not statistically significant trend toward a small decrease in PO extract, but was not significantly changed in either tissue [14]. Thus, it may 284 be released by other organs and act outside of the brain. Its degradation or sequestration from the hemolymph appears to be quite rapid. The final RFamide-like peptide with significant changes in concentration is APQRNFLRFamide, also known as Procambarus clarkii RFamide-Related Peptide 6 (PrcFaRP 6). It was identified first in Cancer borealis in 2003 [22], but received this name in 2006 after it was found in P. clarkii in 2006 [23]. The results for this peptide are illustrated in Table 7.6 and Fig. 7.5. This peptide’s concentration is significantly changed between the samples taken 0 to 30 min after feeding and 90 min to 120 min after feeding, a profile very similar to GPRNFLRFamide. The overall concentration profile of APQRNFLRFamide is not similar, however, as it appears that this peptide is released immediately after feeding, decreases below baseline after 2 hrs, and then returns to a baseline value. As with SENRNFLRFamide and GPRNFLRFamide, this profile indicates that the peptide may be important in initiating feeding behavior but not necessarily for sustaining it, unlike GAHKNYLRFamide. It is interesting to note that 3 of the peptides with significant changes have the extended N-terminal motif of – RNFLRFamide, which has not previously been suggested as a functionally relevant feature. However, additional peptides with this sequence at the C-terminus were quantified in this study and did not show significant changes, so this may be coincidental. For the remaining 5 FMRFamide-related peptides quantified in this study, overall trends may be discussed, but no statistically significant values were identified. These are illustrated in Fig. 7.6. The same general trend is observed with these peptides—an immediate release followed by an overshoot period and a recovery to baseline values. Although most peptides of this family have the same general concentration profile, there are slight individual variations in timing of the start of the initial spike, in how long the initial spike in concentration lasts, how low the 285 concentration reaches in the post-spike period, and how much the baseline value is recovered after the 180 min experiment. Slight differences in these may be important for understanding the functions of RFamides in feeding. For instance, GYSKNYLRFamide and NRNFLRFamide appear to have a slightly more sustained release when compared to AYNRSFLRFamide and GNRFLRFamide. Rapid decreases in hemolymph concentration could be due to active degradation or endocytosis, and other peptides might be present longer to either act in a similar way to prolong the signal, or to act in a different way for a slight change in behavior, i.e. feeding initiation vs. sustaining. What is clear, however, is that the RFamide family peptides change dramatically after feeding, including 4 that were observed to have statistically significant changes in this study. These peptides have been shown to alter neuronal and muscle firing [19], and may make contractions of stomach muscles more easily initiated by motor neuron signals. The function of many RFamides is still unknown, and additional putative roles include rewardlike function for those similar to neuropeptide F, which is analogous to neuropeptide Y in mammals and has been shown to be important in relaying a natural reward signal in insects [24]. 7.5.2. HIGSLYRamide The peptide HIGSLYRamide was first identified in Cancer productus in 2005 [21], but has since been detected in other crab species. No putative function for this peptide exists, and no other YRamide peptides have been identified. This peptide was significantly increased in hemolymph in the time period 0-30 min after feeding compared to both the first time period (-60 to -30 min relative to feeding) and a later time period (120 to 150 min after feeding) (Fig. 7.7 and Table 7.7). This indicates that this one time period, immediately after feeding, is a time in which this peptide changes dramatically. The increase observed is approximately 100%. Although the graph shows that this peptide likely returns to baseline levels quickly, a statistically different 286 value for its concentration is not achieved until the 120 to 150 min time point. This may indicate some variability between animals in the rate at which this peptide’s concentration decreases following the initial spike. This finding is consistent with a large increase in the brain seen in the previous feeding study analyzing tissue extracts [14]. It thus appears that a large amount of this peptide goes into circulation immediately following feeding, and it may end up in the brain via receptor binding. Since the level is unchanged in PO, it is possible that another source releases this peptide. The rapid return to baseline levels may indicate that this peptide is involved in the initial stages of feeding and is less important later. This dramatic and statistically significant change in HIGSLYRamide may indicate that this peptide is important immediately after feeding in the crab. 7.5.3. Orcokinins Orcokinins are a large group of hindgut-stimulating peptides [19]. In this study, none of the 7 quantified were observed to change significantly after feeding (Fig, 7.8). In general, their levels were steady over time, with a light possible increase in the first 30 min sample. In previous studies, they have been found to increase slightly or be unchanged in the brain [14]. The ability to detect and quantify these peptides in microdialysates is a great advancement, however, over previous techniques. Analysis of hemolymph led to the detection of just one orcokinin [25], and previous analysis of microdialysates collected over long time periods (4-18 hrs) and heavily concentrated prior to analysis led to the detection of 6 orcokinins, 3 of which were also analyzed here. The observation of 4 different orcokinins and the ability to quantify them reproducibly in samples collected over only 30 min demonstrates the power of the methodology used in this study. These peptides do not appear to be involved in feeding behavior, however, from the 287 results of this and previous studies. This is surprising due to their known activity on the hindgut of some crustaceans. 7.5.4. Tachykinin-Related Peptides (TRPs) The tachykinin-related peptides are well-known and highly conserved NPs [19]. These peptides have structural homology to mammalian tachykinins, from whence they derive their name. Three are known to exist in Cancer borealis, and two are quantified here. Cancer borealis tachykinin-related peptide (CabTRP) Ia (APSGFLGMRamide) and CabTRP II (TPSGFLGMRamide) do not change significantly throughout the course of feeding (Fig. 7.9). However, an interesting trend is observed that may be significant upon further study. APSGFLGMRamide appears to have its peak value in the 0 to 30 min time point, whereas the peak for TPSGFLGMRamide occurs in the 30 to 60 min time point. It also appears to return to baseline more gradually. This may indicate a sequential release of these related peptides, with roles in feeding that must be carried out in a particular order. Both peptides increase dramatically in the brain after feeding [14]. They have been shown to be released at the synapse, from neuroendocrine organs, and from midgut epithelial cells, so a feeding-related increase in the hemolymph and brain may suggest that these sites are releasing large amounts of stored TRPs into the circulation. From there, they can act on the brain and perhaps on other target sites. Evidence in insects discussed in Chapter 2 suggests a pro-feeding role for tachykinins in the hemolymph, including stimulation of feeding in the cockroach [26]. The different profiles of TRP levels in hemolymph for the two peptides quantified, added to previous evidence of feeding-related increases in the brain and endocrine release of these peptides, suggest that TRPs may be important communication molecules between the gut and/or neurosecretory organs and the brain. 288 7.5.5. RYamides The RYamides quantified in this study, FVGGSRYamide and SGFYANRYamide, did not significantly change throughout the course of feeding (Fig. 7.10). However, they do have a trend similar to other peptides in this study: a rapid increase immediately after feeding, followed by a return to baseline. The reduction in these peptides later after feeding appears to be more gradual than for other peptides, and they have a larger fold-change. In previous studies, their decrease in the PO was not significant, but an increase in the brain was [14]. As the function of RYamides in crustaceans is as yet unknown, the significance of these findings is unclear. However, there is a non-significant suggestion from this work that these peptides are released into the hemolymph following feeding in large amounts and are eliminated from circulation more gradually than other peptides. One of the sites where they might act after this release is the brain. 7.5.6. B-Type Allatostatins Two B-type allatostatins, STNWSSLRSAWamide and VPNDWAHFRGSWamide, were characterized in this work. They were not observed to change significantly throughout the course of feeding (Fig. 7.11). Although they have been observed in tissue extract studies of feeding, they were not observed to change significantly [14]. As little is known about the function of these peptides, these results provide information only that they are not likely to be involved in feeding regulation. 7.5.7. Other Crustacean Neuropeptides Two other peptides that are commonly found in many crustaceans were quantified (Fig. 7.12): crustacean cardioactive peptide (CCAP, PFC(-H)NAFTGC(-H)amide, (-H) indicates a Cys in a disulfide bond) and proctolin (RYLPT). Neither had statistically significant changes in 289 concentration after feeding. CCAP followed a concentration profile similar to that observed for GAHNYLRFamide, and proctolin was unique among the peptides quantified in that it did not return to baseline levels in later time points. Proctolin’s action in defined neural circuits has been studied in detail (See Chapter 2, [19]), and in general it increases the activity of excitable cells. CCAP has been implicated in feeding, particularly in insects (See Chapter 2), and has an interesting bi-modal effect on crustacean neurons and muscles. At low hemolymph concentrations, it affects muscle activity, and at higher concentrations, it alters neuronal activity. Both peptides were observed to increase in the brain in a previous feeding study, but the increase was not significant [14]. Proctolin’s concentration profile suggests long-lasting changes related to feeding, and as such it may be acting more as an indicator of the animal’s satiety or available energy store level. 7.5.8. Other Insect Neuropeptides Pyrokinin (SGGFAFSPRLamide) and a SIFamide (GYRKPPNGSIFamide) were also quantified (Fig. 7.13). No significant changes were observed, but the overall trend of an immediate increase followed by an overshoot of the baseline and/or return to baseline also holds for these two peptides. Both have been studied in detail in insects. Pyrokinins are primarily regulators of reproductive function in insects, and they were the only NP that decreased in the brain of C. borealis following feeding [14]. It is thus likely that pyrokinins are released, potentially from neurons with cell bodies in the brain, into the hemolymph following feeding. As most NPs previously discussed seem to act as signals from other parts of the body to the brain, the presence of an NP that potentially signals in the other direction is of note. SIFamide is released by midgut epithelial endocrine cells after their depolarization [27] and activates the pyloric rhythm [28]. It was not significantly changed in the brain after feeding, so the site of 290 action for SIFamide may be outside of the brain. These results suggest a possible role for SIFamide as a communicator from the midgut to organs of the stomatogastric nervous system, to increase stomach contractions. 7.6. Conclusions and Future Directions The quantities of 28 neuropeptides in the hemolymph of the crab Cancer borealis were monitored in 30-min time segments following feeding using microdialysis and novel MS/MS tools. The measures of changes in NP profiles were determined to be statistically relevant and not caused by multiple repeated sampling. Of these NPs, 5 were observed to change significantly at times after feeding. Most NPs followed a pattern of release, overshoot, and recovery within a 3 hr time period after feeding. Orcokinins and AST-B do not appear to change greatly following feeding. Proctolin is released more gradually and does not return to baseline, even after 3 hrs. Slight differences in NPs from the same families may point to different roles, or sequential release to sustain the signal even in the presence of receptor desensitization. The RFamides were the most significantly altered family of NPs, with 4 changed significantly. They appear to be involved in the active phases of feeding behavior and GAHKNYKRFamide levels may even mirror two distinct episodes of feeding that are frequently observed in the crab; one immediately after the food is present, followed by a short lull in feeding activity that precedes a second feeding period. This pattern is not statistically significant and feeding behavior in crabs has not been described quantitatively, nor have the physiological changes during feeding been determined. For instance, is the two-stage feeding behavior related to filling the stomach with food, followed by a period of digestion that removes some ingested material out of the stomach to make room for more food, which is then consumed in the second feeding period? If so, it is possible that this peptide is involved in the initiation of feeding 291 behavior, or “reminds” the crab to eat again once its stomach has been partially emptied. This is a potentially important function as it informs the animal not only about the state of its stomach (empty or full) but indicates to the crab that more food than can fit in the stomach is desired. From experiments in which blue crabs, Callinectes sapidus, were allowed to eat ad libitum for an hour and then dissected, food was present in the stomach during dissection (Chapter 6). Further investigation of stomach filling and emptying in the crab would be of use for interpretation of the data obtained here. The other RFamides and a YRamide that had significant changes followed the same pattern as most other NPs mentioned previously. This pattern suggests that release of the peptide occurs after feeding, but it is removed from circulation rather rapidly, perhaps to tell the animal to stop eating. This active response indicates that these peptides are likely correlated with the behavior of feeding itself and are not signals of the body’s energy stores. This work demonstrates the application of cutting-edge analytical chemistry tools to the study of NPs in a dynamic physical process, and information potentially relevant to understanding neuroendocrine control of feeding behavior was obtained. Additional replicates would improve the quality of the measurements and perhaps increase the number of statistically significant changes. As it has been demonstrated that the changes here are valid measures, increasing the number of experimental replicates to 5 or 6 individual crabs may make a great impact in the significance of these findings. The same data could be processed again, looking for other NPs. Although 28 were chosen for quantification, an untargeted approach could be done similar to that described in Chapter 6. Another untargeted approach, that will be described in Chapter 8, in which all known small- to medium-sized crab NPs (~500) are quantified in some of the samples to provide a shorter list for quantification in all samples, could also be applied to this 292 same dataset. This process is challenging and time-consuming, but could yield new NPs not previously detected in feeding studies of the crab. It is likely that several important NPs were not quantified based on the choice to focus on the 28 previously studied; a different MS method was used in the previous study and it is biased toward better detection of certain types of NPs, and detects only the most abundant compounds. Finally, the time resolution of the experiment could be improved by employing affinity-enhanced MD (Chapter 4), using different sample handling techniques, and/or using a different type of MS detection that requires less sample. This work provides a useful experimental framework for combining analytical chemistry techniques into a behavioral neuroscience experiment, and indicates some putative feedingrelated neuropeptides in the crab Cancer borealis. Further refinement of the feeding experiment, additional replicates, and re-analysis of additional peptides may be useful in providing more confident assignment of functions for these NPs. Application of this technique to other behavioral experiments on a similar time scale are likely to be successful in quantifying changes in NPs for assessment of function. 7.7. Respective Contributions Statement and Acknowledgements CMS designed and carried out experiments and wrote the paper. LL was responsible for the initial ideas and theory, and revised the paper. Heidi L. Behrens is acknowledged for initially developing the crab microdialysis technique [12, 13] and training. Zhidan Liang is acknowledged for conducting the surgical implantation of an MD probe for the crab used as biological replicate #3. Kirk J. Grubbs is acknowledged for assistance in application of statistical methods. 7.8. Works Cited 1. Fulton, S. Appetite and reward. Frontiers in Neuroendocrinology. 2010, 31, 85-103. 2. Parker, J. A.; Bloom, S. R. Hypothalamic neuropeptides and the regulation of appetite. Neuropharmacology. 2012, 63, 18-30. 293 3. Menzies, J. W.; Skibicka, K.; Egecioglu, E.; Leng, G.; Dickson, S. Peripheral Signals Modifying Food Reward. In: Joost H-G, editor. Appetite Control: Springer Berlin Heidelberg; 2012. p. 131-158. 4. Skibicka, K. P.; Dickson, S. L. Ghrelin and food reward: The story of potential underlying substrates. Peptides. 2011, 32, 2265-2273. 5. Thompson, J. L.; Borgland, S. L. A role for hypocretin/orexin in motivation. Behavioural Brain Research. 2011, 217, 446-453. 6. Popovic, V.; Duntas, L. H. Brain somatic cross-talk: ghrelin, leptin and ultimate challengers of obesity. Nutr Neurosci. 2005, 8, 1-5. 7. Moran, T. H.; Dailey, M. J. Intestinal feedback signaling and satiety. Physiol Behav. 2011, 105, 77-81. 8. Li, L.; Sweedler, J. V. Peptides in the brain: mass spectrometry-based measurement approaches and challenges. Annu Rev Anal Chem (Palo Alto Calif). 2008, 1, 451-483. 9. Abramson, C. I.; Feinman, R. D. Lever-press conditioning in the crab. Physiol Behav. 1990, 48, 267-272. 10. Stein, W. Modulation of stomatogastric rhythms. J Comp Physiol A Neuroethol Sens Neural Behav Physiol. 2009, 195, 989-1009. 11. Marder, E.; Bucher, D. Understanding Circuit Dynamics Using the Stomatogastric Nervous System of Lobsters and Crabs. Annual Review of Physiology. 2007, 69, 291-316. 12. Behrens, H. L.; Chen, R.; Li, L. Combining microdialysis, NanoLC-MS, and MALDI-TOF/TOF to detect neuropeptides secreted in the crab, Cancer borealis. Anal Chem. 2008, 80, 6949-6958. 13. Behrens, H. L.; Li, L. Monitoring neuropeptides in vivo via microdialysis and mass spectrometry. Methods Mol Biol. 2010, 615, 57-73. 14. Chen, R.; Hui, L.; Cape, S. S.; Wang, J.; Li, L. Comparative Neuropeptidomic Analysis of Food Intake via a Multi-faceted Mass Spectrometric Approach. ACS Chem Neurosci. 2010, 1, 204-214. 15. Li, Q.; Zubieta, J. K.; Kennedy, R. T. Practical aspects of in vivo detection of neuropeptides by microdialysis coupled off-line to capillary LC with multistage MS. Anal Chem. 2009, 81, 2242-2250. 16. MacLean, B.; Tomazela, D. M.; Shulman, N.; Chambers, M.; Finney, G. L.; Frewen, B.; Kern, R.; Tabb, D. L.; Liebler, D. C.; MacCoss, M. J. Skyline: an open source document editor for creating and analyzing targeted proteomics experiments. Bioinformatics. 2010, 26, 966-968. 17. Ma, M.; Sturm, R. M.; Kutz-Naber, K. K.; Fu, Q.; Li, L. Immunoaffinity-based mass spectrometric characterization of the FMRFamide-related peptide family in the pericardial organ of Cancer borealis. Biochemical and Biophysical Research Communications. 2009, 390, 325-330. 18. Ma, M.; Wang, J.; Chen, R.; Li, L. Expanding the Crustacean neuropeptidome using a multifaceted mass spectrometric approach. J Proteome Res. 2009, 8, 2426-2437. 19. Christie, A. E.; Stemmler, E. A.; Dickinson, P. S. Crustacean neuropeptides. Cell. Mol. Life Sci. 2010, 67, 41354169. 20. Cruz-Bermúdez, N. D.; Fu, Q.; Kutz-Naber, K. K.; Christie, A. E.; Li, L.; Marder, E. Mass spectrometric characterization and physiological actions of GAHKNYLRFamide, a novel FMRFamide-like peptide from crabs of the genus Cancer. Journal of Neurochemistry. 2006, 97, 784-799. 21. Fu, Q.; Kutz, K. K.; Schmidt, J. J.; Hsu, Y.-W. A.; Messinger, D. I.; Cain, S. D.; de la Iglesia, H. O.; Christie, A. E.; Li, L. Hormone complement of the Cancer productus sinus gland and pericardial organ: An anatomical and mass spectrometric investigation. The Journal of Comparative Neurology. 2005, 493, 607-626. 22. Huybrechts, J.; Nusbaum, M. P.; Bosch, L. V.; Baggerman, G.; Loof, A. D.; Schoofs, L. Neuropeptidomic analysis of the brain and thoracic ganglion from the Jonah crab, Cancer borealis. Biochemical and Biophysical Research Communications. 2003, 308, 535-544. 23. Yasuda-Kamatani, Y.; Yasuda, A. Characteristic expression patterns of allatostatin-like peptide, FMRFamiderelated peptide, orcokinin, tachykinin-related peptide, and SIFamide in the olfactory system of crayfish Procambarus clarkii. The Journal of Comparative Neurology. 2006, 496, 135-147. 24. Shohat-Ophir, G.; Kaun, K. R.; Azanchi, R.; Mohammed, H.; Heberlein, U. Sexual deprivation increases ethanol intake in Drosophila. Science. 2012, 335, 1351-1355. 25. Chen, R.; Ma, M.; Hui, L.; Zhang, J.; Li, L. Measurement of neuropeptides in crustacean hemolymph via MALDI mass spectrometry. J Am Soc Mass Spectrom. 2009, 20, 708-718. 26. Audsley, N.; Weaver, R. J. Neuropeptides associated with the regulation of feeding in insects. General and Comparative Endocrinology. 2009, 162, 93-104. 27. Christie, A. E.; Kutz-Naber, K. K.; Stemmler, E. A.; Klein, A.; Messinger, D. I.; Goiney, C. C.; Conterato, A. J.; Bruns, E. A.; Hsu, Y.-W. A.; Li, L.; Dickinson, P. S. Midgut epithelial endocrine cells are a rich source of the neuropeptides APSGFLGMRamide (Cancer borealis tachykinin-related peptide Ia) and 294 GYRKPPFNGSIFamide (Gly1-SIFamide) in the crabs Cancer borealis, Cancer magister and Cancer productus. J Exp Biol. 2007, 210, 699-714. 28. Christie, A. E.; Stemmler, E. A.; Peguero, B.; Messinger, D. I.; Provencher, H. L.; Scheerlinck, P.; Hsu, Y. W.; Guiney, M. E.; de la Iglesia, H. O.; Dickinson, P. S. Identification, physiological actions, and distribution of VYRKPPFNGSIFamide (Val1)-SIFamide) in the stomatogastric nervous system of the American lobster Homarus americanus. J Comp Neurol. 2006, 496, 406-421. 295 7.9. Figures Figure 7.1. Scatterplot of all data A) before and B) after removal of outliers that represent more than a 5-fold change from baseline. A total of 22 data points out of 1344, or 1.34%, were removed. Data points removed likely represent errors in chromatogram integration. 60 A) 50 40 30 20 10 0 0 5 10 15 20 25 5 10 15 20 25 30 B) 4.5 4 3.5 3 2.5 2 1.5 1 0.5 0 0 30 296 Figure 7.2. Changes in the RFamide peptide GAHKNYLRFamide after feeding. Data are charted at the time in the middle of their collection period. Values plotted are means and SEM for 6 experiments on 3 separate crabs. Brackets connect levels significantly different from each other by a Tukey-Kramer test, and the p-values are indicated. This peptide changes significantly at several time points after feeding and is likely to be a very important feeding-related peptide. GAHKNYLRFamide p=0.0497 1.600 p=0.0314 1.400 Fold-Change 1.200 1.000 0.800 0.600 0.400 0.200 0.000 -45 -15 15 45 75 105 Time After Feeding (min) p=0.0285 p=0.0068 135 165 297 Figure 7.3. Changes in the RFamide peptide SENRNFLRFamide after feeding. Data are charted at the time in the middle of their collection period. Values plotted are means and SEM for 6 experiments on 3 separate crabs. Brackets connect levels significantly different from each other by a Tukey-Kramer test, and the p-values are indicated. This peptide has a very large increase after feeding followed by a return to baseline around 2 hrs later. SENRNFLRFamide 1.800 1.600 p=0.0208 1.400 Fold-Change 1.200 1.000 0.800 0.600 0.400 0.200 0.000 -45 -15 15 45 75 105 Time After Feeding (min) p=0.0234 135 165 298 Figure 7.4. Changes in the RFamide peptide GPRNFLRFamide after feeding. Data are charted at the time in the middle of their collection period. Values plotted are means and SEM for 6 experiments on 3 separate crabs. Brackets connect levels significantly different from each other by a Tukey-Kramer test, and the p-value is indicated. This peptide increases after feeding and returns to baseline levels around 2hrs later. 2.000 GPRNFLRFamide 1.800 1.600 Fold-Change 1.400 1.200 1.000 0.800 0.600 0.400 0.200 0.000 -45 -15 15 45 75 105 Time after Feeding (min) p=0.0404 135 165 299 Figure 7.5. Changes in the RFamide peptide APQRNFLRFamide after feeding. Data are charted at the time in the middle of their collection period. Values plotted are means and SEM for 6 experiments on 3 separate crabs. Brackets connect levels significantly different from each other by a Tukey-Kramer test, and the p-value is indicated. This peptide increases after feeding and may drop to levels below baseline around 2hrs after feeding. 1.600 APQRNFLRFamide 1.400 Fold-Change 1.200 1.000 0.800 0.600 0.400 0.200 0.000 -45 -15 15 45 75 105 Time After Feeding (min) p=0.0373 135 165 300 Figure 7.6. Overall trends of non-significantly changed NPs in the RFamide family. The overall trend for peptides in this family is plotted as a solid line with A) AYNRSFLRFamide, GNRFLRFamide, and GYSKNYLRFamide, and B) NRNFLRFamide, QDLDHVFLRFamide, and SMPSLRLRFamide. Peptides are shown on two graphs for clarity. Means and SEMs are plotted. The overall trend for RFamide peptides is an immediate increase after feeding, followed by an overshoot, and finally a return to baseline values. These results are not statistically significant. A) RFamides-1 2.500 AYNRSFLRFamide GNRFLRFamide 2.000 GYSKNYLRFamide Fold-Change Family 1.500 1.000 0.500 0.000 -45 -15 15 45 75 105 Time After Feeding (min) 135 165 301 B) RFamides-2 1.800 QDLDHVFLRFamide 1.600 SMPSLRLRFamide 1.400 Family 1.200 Fold-Change NRNFLRFamide 1.000 0.800 0.600 0.400 0.200 0.000 -45 -15 15 45 75 105 Time After Feeding (min) 135 165 302 Figure 7.7. Changes in HIGSLYRamide after feeding. Data are charted at the time in the middle of their collection period. Values plotted are means and SEMs for 6 experiments on 3 separate crabs. Brackets connect levels significantly different from each other by a Tukey-Kramer test, and the p-values are indicated. This peptide is significantly increased immediately after feeding and drops to baseline levels significantly after 2hrs post-feeding. HIGSLYRamide 3.000 2.500 Fold-Change 2.000 1.500 1.000 0.500 0.000 -45 -15 p=0.0301 15 75 105 Time After Feeding (min) p=0.0117 135 165 303 Figure 7.8. Overall trends of non-significantly changed NPs in the orcokinin family. The overall trend for peptides in this family is plotted as a solid line with A) NFDEIDRSGFamide, NFDEIDRSGFA, NFDEIDRSGFGFA, and NFDEIDRSGFGFV, and B) NFDEIDRSSFGFN, NFDEIDRSSFGFV, and NFDEIDRTGFGH. Peptides are shown on two graphs for clarity. Means and SEMs are plotted. The overall trend for orcokinins may be a non-feeding-related slight oscillation in their hemolymph levels. These results are not statistically significant. A) Orcokinins-1 NFDEIDRSGFamide NFDEIDRSGFA NFDEIDRSGFGFA 1.600 NFDEIDRSGFGFV 1.400 Family Fold-Change 1.200 1.000 0.800 0.600 0.400 0.200 0.000 -45 -15 15 45 75 105 Time After Feeding (min) 135 165 304 B) Orcokinins-2 NFDEIDRSSFGFN NFDEIDRSSFGFV 1.800 NFDEIDRTGFGFH 1.600 Family Fold-Change 1.400 1.200 1.000 0.800 0.600 0.400 0.200 0.000 -45 -15 15 45 75 105 Time After Feeding (min) 135 165 305 Figure 7.9. Overall trends of tachykinin-related peptides (TRPs) APSFLGMRamide (CabTRP Ia) and TPSFLGMRamide (CabTRP II). Means and SEMs are plotted. Other abbreviations are in the text. Tachykinins appear to increase after feeding, with CabTRP Ia increasing more slowly than CabTRP II. They then return to baseline. These results are not statistically significant. Tachykinins APSGFLGMRamide 2.500 TPSGFLGMRamide Fold-Change 2.000 1.500 1.000 0.500 0.000 -45 -15 15 45 75 105 Time After Feeding (min) 135 165 306 Figure 7.10. Overall trends of RYamides FVGGSRYamide and SGFYANRYamide. Means and SEMs are plotted. They appear to increase after feeding with a more gradual return to baseline than some other peptides. These results are not statistically significant. RYamides FVGGSRYamide 2.500 SGFYANRYamide Fold-Change 2.000 1.500 1.000 0.500 0.000 -45 -15 15 45 75 105 Time After Feeding (min) 135 165 307 Figure 7.11. Overall trends of B-type allatostatins STNWSSLRSAWamide and VPNDWAHFRGSWamide. Means and SEMs are plotted. Slight increases may be observed after feeding, or oscillations in their hemolymph concentrations that are unrelated to feeding may be occurring. These results are not statistically significant. B-Type Allatostatins 1.800 STNWSSLRSAWamide 1.600 VPNDWAHFRGSWamide 1.400 Fold-Change 1.200 1.000 0.800 0.600 0.400 0.200 0.000 -45 -15 15 45 75 105 Time After Feeding (min) 135 165 308 Figure 7.12. Overall trends of crustacean cardioactive peptide (CCAP, PFC(-H)NAFTGC(H)amide, (-H) indicates a Cys in a disulfide bond) and proctolin (RYLPT). Means and SEMs are plotted. CCAP follows a trend similar to many other peptides with changes in their profiles throughout feeding. Proctolin increases after feeding and continues to stay elevated up to 3hrs after feeding. These results are not statistically significant. Other Crustacean NPs 2.500 PFC(-H)NAFTGC(-H)amide RYLPT Fold-Change 2.000 1.500 1.000 0.500 0.000 -45 -15 15 45 75 105 Time After Feeding (min) 135 165 309 Figure 7.13. Overall trends of a pyrokinin (SGGFAFSPRLamide) and a SIFamide (GYRKPPNGSIFamide). Means and SEMs are plotted. These peptides again follow the trend of increase after feeding, followed by a return to baseline. These results are not statistically significant. Other Insect NPs 2.000 SGGFAFSPRLamide 1.800 GYRKPPNGSIFamide 1.600 Fold-Change 1.400 1.200 1.000 0.800 0.600 0.400 0.200 0.000 -45 -15 15 45 75 105 Time After Feeding (min) 135 165 310 7.10. Tables Table 7.1. Peptides quantified in this study and criteria for inclusion of their data in this report. An ANOVA test and Tukey-Kramer HSD test were conducted to determine if each time point was significantly different from any of the others at α=0.05. p-Values <0.05 were considered significant. The bottom row lists the total number of families or peptides in that category. Family Peptide Quantified RFamide RFamide RYamide RFamide RFamide RFamide SIFamide RFamide YRamide Orcokinin Orcokinin Orcokinin Orcokinin Orcokinin Orcokinin Orcokinin RFamide CCAP RFamide Proctolin RFamide RYamide Pyrokinin RFamide AST-B TRP TRP AST-B 9 APQRNFLRFamide AYNRSFLRFamide FVGGSRYamide GAHKNYLRFamide GNRFLRFamide GPRNFLRFamide GYRKPPNGSIFamide GYSKNYLRFamide HIGSLYRamide NFDEIDRSGFamide NFDEIDRSGFA NFDEIDRSGFGFA NFDEIDRSGFGFV NFDEIDRSSFGFN NFDEIDRSSFGFV NFDEIDRTGFGFH NRNFLRFamide PFC(-H)NAFTGC(-H)amide QDLDHVFLRFamide RYLPT SENRNFLRFamide SGFYANRYamide SGGFAFSPRLamide SMPSLRLRFamide STNWSSLRSAWamide APSGFLGMRamide TPSGFLGMRamide VPNDWAHFRGSWamide 28 Significantly Different x x x x x 5 311 Table 7.2. Complete summary of data for this chapter. At each time point for each peptide, the average fold-change, standard error, and number of observations are reported. Abbreviations in text. Orcokinin Orcokinin Orcokinin Orcokinin Orcokinin Orcokinin Orcokinin Proctolin Pyrokinin RFamide RFamide RFamide RFamide RFamide RFamide RFamide RFamide RFamide RFamide RFamide RYamide RYamide SIFamide Tachykinin YRamide AST-B AST-B CCAP Table 7.2 Family STNWSSLRSAWamide VPNDWAHFRGSWamide PFC(-H)NAFTGC(H)amide NFDEIDRSGFamide NFDEIDRSGFA NFDEIDRSGFGFA NFDEIDRSGFGFV NFDEIDRSSFGFN NFDEIDRSSFGFV NFDEIDRTGFGFH RYLPT SGGFAFSPRLamide APQRNFLRFamide APSGFLGMRamide AYNRSFLRFamide GAHKNYLRFamide GNRFLRFamide GPRNFLRFamide GYSKNYLRFamide NRNFLRFamide QDLDHVFLRFamide SENRNFLRFamide SMPSLRLRFamide FVGGSRYamide SGFYANRYamide GYRKPPNGSIFamide TPSGFLGMRamide HIGSLYRamide Time Point (min) Peptide 1.017 0.948 0.879 0.752 0.916 0.913 1.003 0.965 0.926 0.960 0.954 0.947 0.990 0.942 1.017 0.932 0.964 0.948 0.960 0.974 0.986 0.899 1.059 0.925 0.954 1.017 0.963 0.941 Average 0.049 0.042 0.069 0.106 0.038 0.053 0.022 0.054 0.043 0.042 0.050 0.034 0.038 0.013 0.020 0.025 0.024 0.034 0.026 0.015 0.028 0.021 0.120 0.024 0.053 0.026 0.066 0.051 -45 Standard Error 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 n 0.983 1.052 1.121 1.248 1.084 1.087 0.997 1.035 1.074 1.040 1.046 1.053 1.010 1.058 0.983 1.068 1.036 1.052 1.040 1.026 1.014 1.101 0.941 1.075 1.046 0.983 1.037 1.059 Average 0.049 0.042 0.069 0.106 0.038 0.053 0.022 0.054 0.043 0.042 0.050 0.034 0.038 0.013 0.020 0.025 0.024 0.034 0.026 0.015 0.028 0.021 0.120 0.024 0.053 0.026 0.066 0.051 -15 Standard Error 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 n 1.084 1.326 1.228 1.215 1.399 1.254 0.964 1.487 1.507 1.352 1.113 1.189 1.265 1.369 1.612 1.620 1.223 1.358 1.183 1.312 1.634 1.558 1.470 1.498 2.051 0.946 1.272 1.412 Average 0.124 0.138 0.193 0.227 0.154 0.149 0.055 0.296 0.318 0.235 0.115 0.128 0.111 0.133 0.438 0.474 0.112 0.178 0.101 0.247 0.524 0.350 0.350 0.433 0.531 0.152 0.130 0.176 15 Standard Error 5 6 6 6 6 6 6 5 6 6 5 6 6 6 6 5 6 6 6 6 5 6 5 5 5 6 6 6 n 1.014 1.068 1.105 1.126 0.982 0.814 0.779 1.214 1.454 0.954 1.474 0.926 1.067 1.023 1.027 1.375 1.159 1.061 0.906 1.090 1.135 1.467 1.180 1.203 0.993 1.174 0.999 1.038 Average 0.106 0.084 0.207 0.171 0.136 0.075 0.059 0.226 0.174 0.056 0.471 0.063 0.040 0.060 0.105 0.272 0.217 0.103 0.061 0.070 0.129 0.353 0.140 0.267 0.164 0.383 0.075 0.089 45 Standard Error 6 6 6 6 6 6 6 5 6 6 6 6 6 5 6 6 6 6 6 6 6 6 6 6 5 6 6 5 n 312 Orcokinin Orcokinin Orcokinin Orcokinin Orcokinin Orcokinin Orcokinin Proctolin Pyrokinin RFamide RFamide RFamide RFamide RFamide RFamide RFamide RFamide RFamide RFamide RFamide RYamide RYamide SIFamide Tachykinin YRamide AST-B AST-B CCAP Family STNWSSLRSAWamide VPNDWAHFRGSWamide PFC(-H)NAFTGC(H)amide NFDEIDRSGFamide NFDEIDRSGFA NFDEIDRSGFGFA NFDEIDRSGFGFV NFDEIDRSSFGFN NFDEIDRSSFGFV NFDEIDRTGFGFH RYLPT SGGFAFSPRLamide APQRNFLRFamide APSGFLGMRamide AYNRSFLRFamide GAHKNYLRFamide GNRFLRFamide GPRNFLRFamide GYSKNYLRFamide NRNFLRFamide QDLDHVFLRFamide SENRNFLRFamide SMPSLRLRFamide FVGGSRYamide SGFYANRYamide GYRKPPNGSIFamide TPSGFLGMRamide HIGSLYRamide Time Point (min) Peptide 0.922 0.975 1.129 1.223 1.131 0.988 0.856 1.330 1.284 0.861 0.937 1.056 1.203 1.144 1.027 1.096 1.000 1.218 0.948 1.028 1.163 1.101 1.085 0.954 1.020 0.900 1.028 1.422 Average 0.110 0.055 0.190 0.226 0.107 0.133 0.055 0.273 0.197 0.133 0.109 0.061 0.088 0.068 0.055 0.036 0.078 0.092 0.062 0.075 0.107 0.055 0.200 0.059 0.120 0.110 0.064 0.353 75 Standard Error 6 6 6 6 6 6 6 4 6 6 6 5 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 n 0.928 0.949 1.024 1.008 0.955 0.794 0.762 1.797 1.058 0.801 1.285 0.959 0.894 1.008 0.902 1.066 0.896 1.095 0.838 0.882 0.985 1.085 1.093 0.937 1.215 0.825 0.962 1.146 Average 0.122 0.066 0.172 0.172 0.126 0.090 0.063 0.501 0.145 0.063 0.413 0.081 0.060 0.041 0.129 0.155 0.094 0.118 0.074 0.105 0.092 0.295 0.214 0.049 0.280 0.177 0.111 0.368 105 Standard Error 6 6 6 6 6 6 6 5 6 6 6 6 6 6 6 6 6 6 6 6 5 6 6 6 6 6 6 6 n 1.082 0.998 1.008 1.112 1.019 0.990 1.023 1.519 1.177 0.845 0.961 1.046 0.837 1.155 0.813 1.051 0.884 1.061 0.842 0.993 1.207 0.949 1.000 1.009 0.838 0.867 1.099 1.132 Average 0.174 0.100 0.130 0.177 0.091 0.155 0.175 0.317 0.181 0.063 0.135 0.133 0.112 0.179 0.062 0.161 0.069 0.102 0.065 0.146 0.217 0.128 0.139 0.071 0.163 0.115 0.105 0.276 135 Standard Error 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 n 0.971 1.039 0.991 1.018 0.947 0.817 0.863 1.688 1.337 0.997 1.082 1.028 0.918 1.263 0.931 1.080 0.874 1.029 0.903 1.026 1.005 1.133 1.051 0.931 1.069 0.943 0.986 1.174 Average 0.188 0.150 0.183 0.185 0.166 0.110 0.084 0.407 0.303 0.151 0.234 0.106 0.079 0.198 0.098 0.159 0.047 0.077 0.096 0.131 0.162 0.166 0.177 0.083 0.164 0.137 0.136 0.431 165 Standard Error 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 5 6 6 6 6 6 6 6 n 313 314 Table 7.3. Summary of statistics for GAHKYLRFamide. Significant (p<0.05) values obtained from an ANOVA followed by Tukey-Kramer post-hoc analysis are enumerated. In A), time points that are not connected by a letter are significantly different. Standard error (SE) of the difference and the confidence levels (CL) at α=0.05 are indicated. GAHKNYLRFamide A) Values that are Different Time 15 A 75 A 45 A -15 A -45 A 165 105 135 B B B B B B C C C C C C Mean 1.27 1.20 1.07 1.01 0.990 0.918 0.894 0.837 B) Statistics for Pairs of Different Values Time Compared to Time Difference SE Difference Lower CL Upper CL p-Value 15 135 0.428 0.108 0.0814 0.774 0.006 15 105 0.370 0.108 0.0243 0.717 0.0285 75 135 0.366 0.108 0.0202 0.713 0.0314 15 165 0.346 0.108 0.000242 0.693 0.0497 Table 7.4. Summary of statistics for SENRNFLRFamide. Significant (p<0.05) values obtained from an ANOVA followed by Tukey-Kramer post-hoc analysis are enumerated. In A), time points that are not connected by a letter are significantly different. Standard error (SE) of the difference and the confidence levels (CL) at α=0.05 are indicated. SENRNFLRFamide A) Values that are Different Time 15 -15 -45 75 A A A A B B B Mean 1.18 1.04 0.960 0.948 315 45 A B 0.906 165 A B 0.903 135 B 0.842 105 B 0.838 B) Statistics for Pairs of Different Values Compared to SE Time Difference Time Difference 15 105 0.345 0.0975 15 135 0.341 0.0975 Lower CL 0.0337 0.0292 Upper CL 0.657 0.652 pValue 0.0208 0.0234 Table 7.5. Summary of statistics for GPRNFLRFamide. Significant (p<0.05) values obtained from an ANOVA followed by Tukey-Kramer post-hoc analysis are enumerated. In A), time points that are not connected by a letter are significantly different. Standard error (SE) of the difference and the confidence levels (CL) at α=0.05 are indicated. GPRNFLRFamide A) Values that are Different Time Mean 15 A 1.61 45 A B 1.03 75 A B 1.03 -45 A B 1.02 -15 A B 0.983 165 A B 0.931 105 A B 0.902 135 B 0.813 B) Statistics for Pairs of Different Values Time Compared to Time Difference SE Difference Lower CL Upper CL p-Value 15 135 0.798683 0.243 0.0212 1.58 0.0404 Table 7.6. Summary of statistics for APQRNFLRFamide. Significant (p<0.05) values obtained from an ANOVA followed by Tukey-Kramer post-hoc analysis are enumerated. In A), time points that are not connected by a letter are significantly different. Standard error (SE) of the difference and the confidence levels (CL) at α=0.05 are indicated. 316 APQRNFLRFamide A) Values that are Different Time 15 A -15 A B 165 A B -45 A B 45 A B 75 A B 135 A B 105 B B) Statistics for Pairs of Different Values Time 15 Compared to Time 105 Difference 0.550 Mean 1.35 1.04 0.997 0.960 0.954 0.861 0.845 0.801 SE Difference 0.166 Lower CL 0.0197 Upper CL 1.08 p-Value 0.0373 Table 7.7. Summary of statistics for HIGSLYRamide. Significant (p<0.05) values obtained from an ANOVA followed by Tukey-Kramer post-hoc analysis are enumerated. In A), time points that are not connected by a letter are significantly different. Standard error (SE) of the difference and the confidence levels (CL) at α=0.05 are indicated. HIGSLYRamide A) Values that are Different Time 15 A 105 A 165 A -15 A 75 A 45 A -45 135 B B B B B B B Mean 2.05 1.21 1.07 1.05 1.02 0.993 0.953 0.838 B) Statistics for Pairs of Different Values Time 15 15 Compared to Time 135 -45 Difference 1.21 1.10 SE Difference 0.322 0.322 Lower CL 0.182 0.0659 Upper CL p-Value 2.244 2.13 0.0117 0.0301 317 Chapter 8: Quantification of Neuropeptides Altered in Acute and Repeated Ethanol Exposure Adapted from Schmerberg, C. M., Hayes, K. T., Putterman, L. B., and Li, L. [in preparation] 8.1. Abstract The role of neuropeptides (NPs) in motivated behaviors relevant to a variety of pathologies of the reward system is currently under a great deal of investigation [1-7]. One method to understand the pathology of these disorders is to characterize long-term changes in the neural substrates of the reward system after exposure to drugs of abuse [8]. The decapod crustacean is a novel animal model for studying NP changes after long-term drug exposure, as it expresses a variety of NPs that are homologous to some of those NPs postulated to be involved in motivated behaviors in mammals [9], and a wealth of analytical tools exist to study these compounds with high specificity [10]. Recently, evidence has also been presented describing drug-related conditioning in the crayfish Orconectes rusticus [11-15], and studies linking neuropeptide F in arthropods to reproduction-related reward [16]. In this chapter, highspecificity, information-rich tandem mass spectrometry (MS/MS) with data-independent acquisition (DIA) is conducted to quantify changes in the NP content of a major crustacean neuroendocrine organ, the pericardial organ (PO), of the rock crab, Cancer irroratus, after acute and repeated ethanol exposure. Isotopic formaldehyde labeling [17-19] was employed in duplex and triplex schemes in conjunction with DIA MS/MS for quantification. All known crustacean NPs of an appropriate size (approximately 475 NPs) were quantified after repeated ethanol exposure, and 77 changed significantly from control. Of these, large (>25%) changes were observed in 22, all of which were increased. This list of NPs was then used to quantify NP changes after short-term exposure at three different doses of ethanol, and fold-changes from 318 control were calculated. All were increased in short-term exposures for the same dose of ethanol that was used in the repeated exposures, while most were decreased or unchanged at lower and higher doses. An assay to determine the absorption efficiency of ethanol in crustaceans through immersion based on hemolymph alcohol content was also developed and applied. These results have implications for understanding NP release by the PO upon ethanol exposure, both acute and repeated, and potentially adaptation to repeated exposure to a human drug of abuse. 8.2. Introduction It is becoming clear that neuropeptides (NPs) are important chemical messengers in motivated behaviors, such as feeding and drug-induced reward [1-7]. Disorders in the brain’s motivation system can lead to a variety of debilitating pathologies, including compulsive overeating and addiction to drugs, gambling, sex, etc. The unifying hypothesis is that addiction or compulsive behavior is a disorder by which a behavior or substance alters the brain’s natural reward mechanisms, such that natural rewards no longer stimulate it sufficiently. A recent review characterized alcohol addiction as a “reward deficient disorder” with compulsive behavior caused by activation of brain stress pathways—in part by the NPs corticotropin-releasing factor (CRF), dynorphin, and neuropeptide Y (NPY)—and reductions in normal reward function [20]. A multitude of NPs have been implicated in a variety of reward-related disorders in mammals. For compulsive overeating and food reward, ghrelin, orexin/hypocretin, opioids, leptin, and neuropeptide Y are just a few of the NPs postulated to be involved [3-7, 21-23]. Orexins, melanin-concentrating hormone (MCH), brain-derived neurotrophic factor (BDNF), agouti-related protein (AgRP), urocortins, and the previously mentioned CRF, NPY, and dynorphin, are all implicated in addiction [1, 2, 6-8, 20, 23, 24]. A large and growing body of 319 evidence exists to support the role of NPs as important mediators of reward, both food- and drugrelated, in mammalian systems. Many of the NPs postulated to be critical in the reward system have a degree of conservation among mammals and arthropods, including but not limited to peptides related to oxytocin/vasopressin, NPY (neuropeptide F is the invertebrate homolog), and gastrin/cholecystokinin (CCK) [9]. A recent study has demonstrated increased ethanol drinking in Drosophila when reproduction-related reward was prohibited, along with a decrease in NPY brain immunoreactivity [16]. A crustacean model of drug-related reward in the crayfish Orconectes rusticus has also recently been described [11-15], and other arthropod species have recently been established as models of drug addiction [25, 26]. One limitation of most previous studies of NPs involved in reward is reliance on antibody-based techniques for NP detection. As discussed in previous chapters, this method suffers from antibody-cross reactivity and is only able to provide limited information. A wealth of high specificity tools for studying NP changes in the decapod crustacean based on mass spectrometry (MS) and tandem MS (MS/MS) have been developed for improved selectivity of NP identification and the potential to obtain sequence information directly from small samples [10]. A highly specific method of quantifying NPs and verifying their sequences using the newly developed MS/MS acquisition method, data-independent acquisition (DIA), has been applied previously in this dissertation to quantify NPs changed in feeding with high specificity and accuracy in the decapod crustacean (Chapter 7). Although it is currently unknown if alcohol exposure is a rewarding experience for crustaceans, other drugs that are rewarding in humans are also rewarding in crayfish [14, 15] and most drugs of abuse act on common neural pathways in mammals to produce reward. It is 320 currently unclear which of alcohol’s many effects is responsible for its reinforcing properties. In the classical model, ethanol increases dopamine in the mesolimbic dopamine system by inhibition of GABA-ergic neurons that tonically inhibit dopaminergic neurons in the ventral tegmental area (VTA) [27]. This is by no means sufficient to explain the complex mechanisms by which alcohol is rewarding, however. Alcohol also is thought to alter levels of endogenous opioids for positive reinforcement and/or cessation of negative reinforcement through currently unknown mechanisms [28]. In the crustacean, GABA is also an inhibitory neurotransmitter in the central and peripheral nervous systems, and further activation of GABA receptors by ethanol may increase inhibitory transmission. In this chapter, long-term changes in the NP complement of a major neuroendocrine organ, the pericardial organ (PO), of the Rock crab Cancer irroratus after repeated exposure to a human drug of abuse, ethanol, are determined and compared to short-term exposure changes. The advantage of this approach is that all known putative NPs can be quantified, and the data can be re-analyzed in the future as additional information becomes available. Isotopic formaldehyde labeling [17-19] was also employed in duplex and triplex schemes in conjunction with DIA MS/MS quantitation for the first time. Using an untargeted quantitation approach, approximately 475 NPs were quantified after repeated daily exposure to 0.2 M ethanol for 1 hr. Of these NPs, 77 changed significantly from controls, and 22 of these changed more than 25%, all of which were increased compared to controls. These were quantified in short-term 1hr exposure samples from crabs exposed to three different doses of ethanol (0.125 M, 0.2 M, and 0.5 M), and foldchanges from control were calculated. All were increased in short-term exposures to 0.2 M ethanol, while most were decreased or unchanged after 0.125 M and 0.5 M exposure. Twelve increases in the 0.2M dose were statistically significant compared to control animals, and no 321 statistical difference was observed at the other doses. The relevance of this dose-dependency is currently unknown. An assay to determine the extent to which ethanol is absorbed by the crab through the gills when immersed by quantifying hemolymph levels is also developed. Interestingly, after short-term exposure, the hemolymph alcohol levels were the same for crabs exposed to 0.2M or 0.5M ethanol. These results have implications for understanding NP release by the PO upon ethanol exposure, both acute and chronic, and potentially adaptation to repeated exposure to a human drug of abuse. 8.3. Methods Several of the methods have been described previously in Chapters 4, 6, and 7, and are presented in detail in Appendix A. Only where the methods differ from previous chapters will they be discussed here. 8.3.1. Materials Unless otherwise indicated, reagents were purchased from Fisher Scientific (Waltham, MA). Alcohol dehydrogenase (ADH) from Saccharomyces cerevisiae, ≥300 units/mg protein (product # A3263 Sigma Aldrich, St. Louis, MO), phenazine methosulfate (PMS, Sigma Adrich), β-nicotinamide adenine dinucleotide hydrate (NAD+, Sigma Aldrich), thiazolyl blue tetrazolium bromide (MTT, Sigma Aldrich), ethanol 100% (Sigma Aldrich), 1mL plastic syringe with ½” 26 ga needle (Becton Dickinson, Franklin Lakes, NJ), crab saline (440 mM NaCl, 26 mM MgCl 2, 13 mM CaCl2, 11.2 mM Trizma base, 11 mM KCl, 5.1 mM maleic acid, pH adjusted to 7.4), phosphate-buffered saline (PBS, 137 mM NaCl, 2.7 mM KCl, 8.1 mM NaH2PO4, 1.76 mM KH2PO4, pH 7.4), tris buffer (0.1 M Tris-HCl, 0.1% (w/v) Triton X-100, pH 8.5), sodium dodecyl sulfate (SDS, Sigma-Aldrich), dimethylformamide (DMF, Sigma-Aldrich), methanol, 322 formic acid, 200 proof ethanol (Decon Laboratories, Inc., Bryn Mawr, PA), pellet pestles (Fisher Scientific), sonicator, centrifuge 8.3.2. Animals Male rock crabs (Cancer irroratus) were obtained from Ocean Resources (Sedgwick, ME) and housed in an aquarium system with sump filtration and a protein skimmer. All tanks had crushed gravel as substrate and water quality was monitored regularly. Regular water changes were conducted and animals were fed if they were in the tanks for more than 1 week. Animals were fed raw white fish (purchased from a grocery store) and were allowed to eat for several hours. The room had temperature, humidity, and light controls. The tanks were maintained at a water temperature of 10-15°C, and salinity was maintained in the 32 to 38 ppt range. The room was on a 12-hr reverse light schedule, with lights off from 10 AM to 10 PM—in order to observe animals during their active period (dark) during normal work hours—and on from 10 PM to 10 AM. For dissection, a previously published procedure was followed [29]. Briefly, animals were put on ice for at least 15min to anesthetize them, and were weighed following anesthesia immediately prior to dissection. Sinus glands, brains, and pericardial organs were isolated and stored in tubes containing 50 µL of acidified methanol (90:9:1 MeOH: H2O: acetic acid) on ice. Tissues were kept in -80°C until extraction. 8.3.3. Ethanol Exposure All ethanol exposures were conducted in shoe box-sized solid plastic mouse cages with lids that contained 4L of liquid. Solutions of ethanol (Decon Laboratories) at 0.5, 0.2, and 0.125 M in water from the crabs’ home aquaria were made in these cages. Control animals were placed in pure aquarium water. For short-term exposure control, 8 crabs were used. For the short-term 323 exposure dosed, the following numbers of animals were used: 0.125M: 4 crabs, 0.2M: 8 crabs, and 0.5M: 3 crabs. For long-term exposure, two cohorts were used, one with 3 crabs in control exposure and 3 dosed, and the other with 4 crabs under the control condition and 4 dosed. Prior to exposure, 0.75mL of hemolymph was withdrawn, and then replaced with an equal volume of crab saline, with exceptions as noted. Crabs were placed in the cages and left undisturbed for 1hr. At this point, they were removed from the tank, 0.75mL of hemolymph was withdrawn, and they were either put on ice for dissection or rinsed by immersion in ethanol-free aquarium water and returned to their home tank. Exposed and control crabs were kept separate with a plexiglass divider. For long-term studies, 0.75 mL of hemolymph was only taken every other day to avoid undue stress on the animal; on the other days, 0.2 mL was taken and this volume was not replaced with crab saline. For long-term exposure studies, a system using colored plastic cable ties was developed to identify individuals. They were exposed to 0.2 M ethanol for 1hr daily for 5 consecutive days, with dissection occurring immediately after the last exposure. Crabs were fed on the day prior to the first exposure. 8.3.4. Sample Preparation The pericardial organs were extracted following a published procedure [18]. Briefly, tissues were ground in 100 µL acidified methanol, sonicated, and pelleted by centrifugation for 5 min at 16,100 x g. The supernatant was saved and the pellet was re-extracted two additional times. Supernatants were combined and solvent was evaporated (Savant SC10 Speedvac, Thermo Scientific). Samples were resuspended and desalted using C18 Ziptips (EMD Millipore, Billerica, MA). This was dried again and resuspended in 75 µL of 10 nM bradykinin (American Peptide, Sunnyvale, CA) internal standard solution. Formaldehyde labeling was conducted following one of two published procedures [18] or [17]. For long-term samples, a two-plex 324 dimethyl labeling strategy employing formaldehyde and deuterium formaldehyde with borane pyridine as hydride source was employed [18]. For short-term samples, a three-plex dimethyl labeling adding 13 C deuterium formaldehyde was employed, using NaCNBH3 or NaCNBD3 as the hydride source [17]. Samples were assigned to groups based on crab size. A control sample was present in each group. The assignment of light, intermediate, or heavy label was conducted randomly to blind the samples for analysis and eliminate any labeling bias. After labeling, samples were evaporated to dryness, resuspended in 10 µL 0.1% FA, and mixed. 8.3.5. Instrumental Analysis Samples were placed in LC vials and analyzed as in Chapters 6 and 7 and Protocol A. Ultrahigh performance liquid chromatography (UPLC)-MSE analysis was conducted on a Waters nanoAcquity UPLC coupled to a Waters Synapt G2 mass spectrometer (Waters, Milford, MA). Samples (2 µL) were trapped on a preconcentration column and desalted on-line. This column was then put in line with a C18 reversed-phase column (BEH130 C18, 1.7 µm, 75 µm x 100 mm, Waters, Milford, MA). The outlet of this column was connected to a fused silica capillary with a pulled tip of internal diameter ~5 µm (Sutter Instrument Company, Novato, CA). This was used as the ESI inlet. A 75 min reversed-phase run with solvent A as 0.1% FA in water and B as 0.1% FA in ACN was used. The instrument was operated in data-independent MS/MS mode with the high energy scan having a voltage ramp from 25 to 65 V and Glu-fibrinopeptide was infused for lockspray calibration. 8.3.6. Data Analysis A previously developed pseudo multiple reaction monitoring (pMRM) method for quantification of MSE data (Chapters 6, 7) using Skyline software (University of Washington [30]) was developed for use in this chapter. A list of all known crustacean neuropeptides (~475 325 NPs) smaller than 1500 Da and excluding the CHH-precursor related peptides was created for data extraction and integration of long-term exposure samples. A shorter list containing 22 peptides was created from the long-term exposure results for analysis of the short-term exposure data in a method described below. The internal standard bradykinin was added to each list, and settings for triplex dimethylation were input into the program. The iRT function with retention time predictor was used, calibrated off of results from Mascot (Matrix Science, London, UK) identifications of equine myoglobin tryptic peptides in a previous LC run of the myoglobin digest alone under the same UPLC conditions. The 2+ and 3+ precursors were monitored to the respective transitions to their most abundant 1+ and 2+ b and y ions. MS/MS filtering parameters were set to DIA with MSe isolation, and 10,000 resolving power. Peak integration was checked manually for a good match to the most abundant transitions and agreement with the predicted retention times. Variation was greater between sets of samples run on the instrument on different days than within a single day. All long-term samples were subjected to UPLC-MS/MS on the same day and analyzed in parallel. Similarly, all short-term samples were injected on UPLCMS/MS on a different day but also analyzed in parallel. Peak areas were exported to Microsoft Excel. All transitions for each isotopic form of the labeled peptide were summed. These were normalized to the corresponding isotopic-labeled signal for bradykinin and divided by the body weight of the crab. Conditions were unblinded, and the mean and variance was calculated for each isotope-labeled peptide. The average variance was also conducted, and samples whose variance was greater than 3 times the average for that peptide were considered outliers and were removed from analysis. A minimum of 2 outliers from 7 samples were observed, and most had 1 or no outliers. A student’s paired t-test was conducted, and significance was set at p<0.05. The 326 ratio of dosed to control was determined for each peptide. For those peptides with a significant difference according to the t-test and a ratio greater than 1.25 or less than 0.75, detailed results are shown. These 22 peptides were used to create the list for analysis of the short-term exposure samples. For short-term samples, all internal standard- and crab weight-normalized values were compared to control values using a student’s t-test with significance threshold at p<0.05. The values were then divided by the control value to yield fold-change. 8.3.7. Alcohol Dehydrogenase (ADH) Assay Hemolymph was collected from the crabs as described previously. Additional hemolymph from an un-dosed crab was used to make blanks and calibration curve samples. Hemolymph was put on ice immediately after collection and spun down at 16,100 x g for 5 minutes to get crab plasma. Blank plasma can be stored in the freezer for several weeks. A calibration curve consisting of 10, 5, 3, 1, and 0.1 mM EtOH (Sigma Aldrich) in crab plasma was created. To each well of the microplate (96-well Perkin Elmer Isoplate, white frame and clear well, Perkin Elmer, Waltham, MA) were added 76.7 µL of Tris buffer (0.1 M Tris-HCl, 0.1% (w/v) Triton X-100, pH 8.5), 10 µL of 600 uM MTT (in Tris buffer), and 10 µL of 37.7 mM NAD+ (in Tris buffer) solution. Samples, calibrants, and blank were then added to each well in volumes of 4 µL. Samples from crabs exposed to alcohol were also diluted 1:4 with Tris buffer and 4 µL of this was put in wells. Two µL of 5 pM phenazine methosulfate (PMS) was added followed by 4 µL of 7.33x106 U/L ADH. The plate was mixed on a rotating platform (Fisher Scientific Clinical Rotator) for 1 minute and incubated at 50 °C for 60 minutes. The reaction was stopped by adding 100 µL of inhibitor (20% SDS (w/v) in 50/50 DMF/H2O (v/v)) to each well, followed by another 1min of mixing. The absorbance at 570 nm and 655 nm was determined with a Tecan Ultra 384 microplate reader with XFluor4 software (Tecan Group Ltd., Männedorf, 327 Switzerland) and filters from Omega Optical (Omega Optical, Inv., Brattleboro, VT). Data was analyzed in Microsoft Excel, plotting absorbance at 570nm – absorbance at 655nm against EtOH concentration. A linear regression was generated and used to calculate the concentrations of unknown samples within the calibrated range. This assay was developed by modification of reference assays to crab hemolymph samples [31-33]. 8.4. Results and Discussion This study successfully quantified 475 NPs in samples labeled with H- and Dformaldehyde and analyzed via DIA MS/MS. A large number of these were found to have statistically significant changes in animals repeatedly exposed to alcohol. Large changes were observed in 22 of these, which were then quantified in samples labeled with H-formaldehyde, Dformaldehyde, and 13C D-formaldehyde and NaCNBD3. The application of isotopic labeling with DIA MS/MS quantitation has not previously been reported, and the 3-plex dimethylation scheme is less frequently used. This work demonstrates that stable isotope labeling and DIA MS/MS can be combined with pseudo-SRM data analysis for highly accurate quantitation of peptides in large-scale analysis. 8.4.1. Repeated Alcohol Exposure Two cohorts of crabs were subjected to daily, 1hr exposures to 0.2 M ethanol (or plain aquarium water) for 5 consecutive days. NPs from the POs of animals exposed to ethanol had 77 significantly altered NPs, all of which were increased relative to control animals. Large changes (>25%) were observed for 22 NPs, indicated in Fig. 8.1.A and B. One combination allatostatin (AST Combo, a prepropeptide sequence containing multiple putative active ASTs), 9 A-type ASTs (AST-A), 2 B-type ASTs (AST-B), one kinin, one YRamide, 5 RFamide-like peptides, one RYamide, and 2 tachykinin-related peptides are shown. 328 Increases in NP levels in general after alcohol exposure are not surprising. Due to its positive effects on GABA inhibitory signaling, alcohol can decrease overall neuronal activity and thus decrease any constitutive release of NPs by the PO into the hemolymph. However, over a period of several days, cells might decrease NP synthesis to compensate for a period during which release was reduced (the daily exposure period). On the other hand, inhibition of overall neuronal activity is not the only mechanism by which ethanol acts in the mammal, and other processes may be at work in the crab as well, such as effects on NP release, which ethanol stimulates in some regions of the brain. It is possible that after 5 days, changes in the synthesis rate of NPs have not occurred, or that increased levels of NPs are a result of increased synthesis of NPs starting immediately after immersion in the tank. Changes in allatostatins are of interest because most of these compounds were not observed to change significantly following feeding. Their overall activity is to reduce synthesis of juvenile hormones and decrease neuronal activity (see Ch. 2). This effect is similar to the classic effect of ethanol through activation of the GABA receptor. It is possible that their increased levels in the PO are in response to adaptation of the animal to reduced overall neuronal activity when ethanol is present, and ASTs serve the same purpose after the animal’s exposure is over, in order to maintain a certain overall level of activity. The second most affected family of NPs, the RFamide-like peptides, contains family members that stimulate and inhibit firing of muscles and neurons (See Ch. 2). Most peptides in this family increase contractions in the heart and other muscles, but not all do. In Ch. 7 it was observed that RFamides increased in the hemolymph immediately following feeding, and this is in agreement with tissue extract findings [18]. Their increase in the PO tissue after repeated alcohol exposure may be due to increased synthesis of these peptides potentially related to 329 neuropeptide Y and CCK. Longer after alcohol exposure, they may be released in large quantities into the hemolymph, potentially as a reward signal. Alternatively, they may be released after alcohol exposure to increase the basal tone of active cells to counteract negative effects of ethanol. Other peptides that were altered are also considered stimulatory peptides, and this may account for their increase. 8.4.2. Short-Term Alcohol Exposure For the peptides with large, significant changes after repeated ethanol exposure, large increases were also observed in the 0.2 M dose after short-term exposure (see Fig. 8.2.), and 12 of these were statistically significant compared to control (p<0.05). For a smaller dose, 0.125 M, most were decreased or close to unchanged compared to control, and for a larger dose, 0.5 M, little change from control was observed. Because the profiles look so similar for each peptide, it was verified statistically that they were not all simply measurements of the same background signal; values are significantly different between peptides. The relevance of these findings is currently unclear. When taken with information on the hemolymph ethanol concentrations (HECs) following short-term exposure (Table 8.1), it appears that the same HEC is causing different effects in the PO NP content between the 0.2M and 0.5M doses. The increase in PO levels of these NPs, which was statistically significant compared to control for 12 compounds, may indicate that ethanol is reducing tonic NP release by the PO in this condition by reducing overall neuronal activity, through its known augmentation of GABAergic signaling. At the lower dose, the HEC may not be high enough to alter baseline release of NPs from the PO, and thus this dose is not distinguishable from control. At the high dose, there is the possibility that NP release is stimulated, along with alterations in water elimination from the animal, or increases in the efficacy of enzymes that metabolize ethanol. Stimulated NP 330 release occurs after ethanol exposure in some regions of the mammalian brain, so it is possible that the PO is being stimulated to release NPs despite overall increased inhibitory tone in the nervous system due to the presence of ethanol. This leads to a PO NP level in the 0.5M condition that is similar to that observed in control. Since the HEC is similar at the 0.5M dose and the 0.2M dose, it is possible that the animal is eliminating water from its circulation more slowly to increase its overall hemolymph volume to reduce the effects of such a high dose of ethanol. Other possible explanations include ethanol modulation of the enzymes responsible for its metabolism. Finally, the higher numbers of animals at the 0.2M and control conditions (8 in each condition) compared to the 0.125M and 0.5M conditions (4 and 3 animals, respectively), may be the reason statistical significance is only observed in the 0.2M dose. Additional measurements of NP levels and HECs at the 0.125M and 0.5M doses may provide a clearer picture and potentially statistically significant differences from control values. A variety of additional information about the effects of different doses of ethanol in the crab is necessary to properly interpret these results. 8.4.3. Alcohol Dehydrogenase Assay The alcohol dehydrogenase assay generated a linear calibration curve for ethanol in the concentration range 0.1-10 mM (Fig. 8.3.). A representative graph showing the change in ethanol concentration for crabs exposed to 0.5 M ethanol is shown in Fig. 8.4. Average values for hemolymph alcohol concentration for short-term exposures are indicated in Table 8.1. For 0.2 and 0.5 M, the concentration was approximately equal. Daily values for before and after exposures in the repeated exposure experiment are indicated in Fig. 8.5. The experiment was conducted in two cohorts, one of 3 control and 3 dosed crabs, and one of 4 control and 4 dosed crabs. For both acute and repeated exposures, the control condition does not lead to an elevation in hemolymph ethanol levels, and levels before the test are nearly zero. No large change is 331 observed in the post-exposure alcohol levels from day to day in the repeated exposure test. This does not indicate that crabs are not developing tolerance, as increased elimination rate is not the only mechanism by which tolerance can be achieved. 8.5. Conclusions and Future Work The observation that 22 NPs increased greatly and significantly in crabs repeatedly exposed to ethanol daily over a 5-day period suggests that these NPs may be being retained in neurons due to overall reduced activity caused by ethanol’s potentiation of GABA signaling. The synthesis of these NPs could also be upregulated as a result of repeated ethanol exposure, as some (such as allatostatins) may be used to maintain overall inhibitory neuronal tone after ethanol exposure is concluded in a homeostatic mechanism. The levels of these compounds in other tissues and the hemolymph would be of great interest to determine if the increases in the PO are due to decreased release or increased synthesis. Twelve of the 22 NPs were also significantly increased after short-term exposure to the same dose of ethanol, 0.2M. Since shortterm exposure is not likely to cause significant changes in NP synthesis, it is probable that these NPs increase in PO after alcohol exposure as a result of reduced baseline release. Short-term exposure at other doses did not provide differences in the levels of these 22 NPs in the PO compared to control. This could be due to ethanol-stimulated NP release at the 0.5M dose, and not enough ethanol present at the 0.125M dose to inhibit basal NP release, or it could be an artifact of having fewer experimental animals in the low-dose conditions. Similar HECs for short-term exposure to 0.2M and 0.5M ethanol could be due to regulation of water balance in the animal, ethanol-metabolizing enzymes, or again could be an artifact of too few replicates. Additional information will be required to better understand the implications of this work, including more experimental replicates, analysis of other tissues, and following concentration 332 changes of ethanol and NPs in the hemolymph throughout ethanol exposure using microdialysis sampling. The data obtained thus far can also be mined for significant changes in the short-term samples in additional NPs that may have different expression profiles. Behavioral analysis may help determine if ethanol is a rewarding stimulus and if tolerance is occurring. More investigations into the mechanisms of ethanol uptake and elimination in the crab would also be of use. 8.6. Respective Contributions and Acknowledgments CMS designed and carried out the experiments and wrote the paper. KTH helped with sample preparation. LBP helped in developing and applying the ADH assay. LL advised and revised the paper. Additional advice on experimental design was provided by Brendan M. Walker of Washington State University, Pullman, WA. Helpful insight was provided by Chenxi Jia, Zhidan Liang, and Nicole Woodards. 8.7. Works Cited 1. Marchant, N. J.; Millan, E. Z.; McNally, G. P. The hypothalamus and the neurobiology of drug seeking. Cell Mol Life Sci. 2012, 69, 581-597. 2. Sprow, G. M.; Thiele, T. E. The neurobiology of binge-like ethanol drinking: Evidence from rodent models. Physiology & Behavior. 2012, 106, 325-331. 3. Fulton, S. Appetite and reward. Frontiers in Neuroendocrinology. 2010, 31, 85-103. 4. Parker, J. A.; Bloom, S. R. Hypothalamic neuropeptides and the regulation of appetite. Neuropharmacology. 2012, 63, 18-30. 5. Menzies, J. W.; Skibicka, K.; Egecioglu, E.; Leng, G.; Dickson, S. Peripheral Signals Modifying Food Reward. In: Joost H-G, editor. Appetite Control: Springer Berlin Heidelberg; 2012. p. 131-158. 6. Skibicka, K. P.; Dickson, S. L. Ghrelin and food reward: The story of potential underlying substrates. Peptides. 2011, 32, 2265-2273. 7. Thompson, J. L.; Borgland, S. L. A role for hypocretin/orexin in motivation. Behavioural Brain Research. 2011, 217, 446-453. 8. Briand, L. A.; Blendy, J. A. Molecular and genetic substrates linking stress and addiction. Brain Research. 2010, 1314, 219-234. 9. Grimmelikhuijzen, C. J. P.; Hauser, F. Mini-review: The evolution of neuropeptide signaling. Regulatory Peptides. 2012, 177, Supplement, S6-S9. 10. Li, L.; Sweedler, J. V. Peptides in the brain: mass spectrometry-based measurement approaches and challenges. Annu Rev Anal Chem (Palo Alto Calif). 2008, 1, 451-483. 11. Dziopa, L.; Imeh-Nathaniel, A.; Baier, D.; Kiel, M.; Sameera, S.; Brager, A.; Beatriz, V.; Nathaniel, T. I. Morphine-conditioned cue alters c-Fos protein expression in the brain of crayfish. Brain Research Bulletin. 2011, 85, 385-395. 12. Huber, R. Amines and motivated behaviors: a simpler systems approach to complex behavioral phenomena. Journal of Comparative Physiology a-Neuroethology Sensory Neural and Behavioral Physiology. 2005, 191, 231-239. 333 13. Nathaniel, T. I.; Huber, R.; Panksepp, J. Repeated cocaine treatments induce distinct locomotor effects in Crayfish. Brain Research Bulletin. 2012, 87, 328-333. 14. Nathaniel, T. I.; Panksepp, J.; Huber, R. Drug-seeking behavior in an invertebrate system: Evidence of morphine-induced reward, extinction and reinstatement in crayfish. Behavioural Brain Research. 2009, 197, 331-338. 15. Panksepp, J. B.; Huber, R. Ethological analyses of crayfish behavior: a new invertebrate system for measuring the rewarding properties of psychostimulants. Behavioural Brain Research. 2004, 153, 171-180. 16. Shohat-Ophir, G.; Kaun, K. R.; Azanchi, R.; Mohammed, H.; Heberlein, U. Sexual deprivation increases ethanol intake in Drosophila. Science. 2012, 335, 1351-1355. 17. Boersema, P. J.; Raijmakers, R.; Lemeer, S.; Mohammed, S.; Heck, A. J. R. Multiplex peptide stable isotope dimethyl labeling for quantitative proteomics. Nat Protoc. 2009, 4, 484-494. 18. Chen, R.; Hui, L.; Cape, S. S.; Wang, J.; Li, L. Comparative Neuropeptidomic Analysis of Food Intake via a Multi-faceted Mass Spectrometric Approach. ACS Chem Neurosci. 2010, 1, 204-214. 19. Kovanich, D.; Cappadona, S.; Raijmakers, R.; Mohammed, S.; Scholten, A.; Heck, A. J. Applications of stable isotope dimethyl labeling in quantitative proteomics. Anal Bioanal Chem. 2012, 404, 991-1009. 20. Koob, G. F. Theoretical frameworks and mechanistic aspects of alcohol addiction: alcohol addiction as a reward deficit disorder. Curr Top Behav Neurosci. 2013, 13, 3-30. 21. Grosshans, M.; Loeber, S.; Kiefer, F. Implications from addiction research towards the understanding and treatment of obesity. Addiction Biology. 2011, 16, 189-198. 22. Skibicka, K. P.; Dickson, S. L. Ghrelin and food reward: the story of potential underlying substrates. Peptides. 2011, 32, 2265-2273. 23. Plaza-Zabala, A.; Maldonado, R.; Berrendero, F. The Hypocretin/Orexin System: Implications for Drug Reward and Relapse. Mol Neurobiol. 2012, 45, 424-439. 24. Ryabinin, A. E.; Tsoory, M. M.; Kozicz, T.; Thiele, T. E.; Neufeld-Cohen, A.; Chen, A.; Lowery-Gionta, E. G.; Giardino, W. J.; Kaur, S. Urocortins: CRF's siblings and their potential role in anxiety, depression and alcohol drinking behavior. Alcohol. 2012, 46, 349-357. 25. Awofala, A. A. Genetic approaches to alcohol addiction: gene expression studies and recent candidates from Drosophila. Invert Neurosci. 2011, 11, 1-7. 26. Kaun, K. R.; Devineni, A. V.; Heberlein, U. Drosophila melanogaster as a model to study drug addiction. Hum Genet. 2012, 131, 959-975. 27. Kandel, E. R.; Schwartz, J. A.; Jessell, T. M. Principles of Neural Science. 4 ed. New York, NY: McGraw Hill; 2000. 28. Walker, B. M.; Valdez, G. R.; McLaughlin, J. P.; Bakalkin, G. Targeting dynorphin/kappa opioid receptor systems to treat alcohol abuse and dependence. Alcohol. 2012, 46, 359-370. 29. Ma, M.; Wang, J.; Chen, R.; Li, L. Expanding the Crustacean neuropeptidome using a multifaceted mass spectrometric approach. J Proteome Res. 2009, 8, 2426-2437. 30. MacLean, B.; Tomazela, D. M.; Shulman, N.; Chambers, M.; Finney, G. L.; Frewen, B.; Kern, R.; Tabb, D. L.; Liebler, D. C.; MacCoss, M. J. Skyline: an open source document editor for creating and analyzing targeted proteomics experiments. Bioinformatics. 2010, 26, 966-968. 31. Lim, H. H.; Buttery, J. E. Determination of ethanol in serum by an enzymatic pms-int colorimetric method. Clinica Chimica Acta. 1977, 75, 9-12. 32. Zanon, J. P.; Peres, M. F. S.; Gattás, E. A. L. Colorimetric assay of ethanol using alcohol dehydrogenase from dry baker's yeast. Enzyme and Microbial Technology. 2007, 40, 466-470. 33. Czerlinski, G. H.; Anderson, B.; Tow, J.; Reid, D. S. Coupling of redox indicator dyes into an enzymatic reaction cycle. Journal of Biochemical and Biophysical Methods. 1988, 15, 241-247. 334 8.8. Figures Figure 8.1. Fold-change from baseline in repeated exposure experiments for NPs with statistically significant changes greater than 25%. Values shown are means ± standard error of the means (SEM). P values indicated with * or **. A) AST-family peptides. B) Other peptides. A) B) 335 Figure 8.2. Fold-change from baseline in acute exposure experiments at 3 different doses for NPs identified in repeated exposure experiments as potentially of interest. Values shown are means ± SEM. Statistical significance is indicated with a * (p<0.05). A) AST-family peptides. B) Other peptides. A) 336 B) Figure 8.3. Representative graph showing linearity of ADH assay for ethanol concentration in crab hemolymph. 0.3 Abs (570-655nm) 0.25 0.2 0.15 0.1 y = 0.027x - 0.0076 R² = 0.9552 0.05 0 0 -0.05 2 4 6 8 Ethanol Concentration (mM) 10 12 337 Figure 8.4. Graph illustrating change in hemolymph ethanol levels after acute exposure to 0.5 M solution. Values indicated are means ± SEM (n=3). Significance value between before and after values for ethanol exposed crabs is indicated. 0.5M EtOH Immersion 35 EtOH Conc (mM) 30 25 20 Control 15 p=0.0006 EtOH Exposed 10 5 0 before after 338 Figure 8.5. Hemolymph ethanol levels for crabs in the repeated exposure experiments. Control and 0.2 M ethanol-exposed crabs shown. For each day and condition, n=7. Experiment was conducted in two cohorts, one of 3 control, 3 dosed, and one of 4 control, 4 dosed. Values indicated are means ± SEM. Negative values are an artifact of normalizing the response to a blank sample of crab hemolymph, which has a small amount of intrinsic activity in the assay that Control Day 1 Day 2 Day 3 Day 4 After Before After Before After Before After Before Dosed After 45 40 35 30 25 20 15 10 5 0 -5 Before Hemolymph EtOH Concentration (mM) is variable from one crab to another. Day 5 8.9. Table Table 8.1. Hemolymph alcohol concentrations of dosed crabs after short-term exposure. Means and standard deviations are indicated for each dose. A small amount of baseline activity in the assay is observed for undosed animals. Dose (M) Control 0.125 0.2 0.5 Hemolymph Alcohol Concentration (mM) After Exposure Average SEM 4.42 1.11 12.7 7.98 31.6 4.24 29.4 0.782 Number of Animals (n) 8 4 8 3 339 Chapter 9: Conclusions and Future Directions 9.1 Conclusions In this dissertation, methods were developed for analysis of neuropeptides (NPs) in the decapod crustacean based on mass spectrometry (MS) and applied to experiments in feeding and alcohol exposure. A summary of the types of NPs found in crustaceans, MS-based and traditional biochemical analysis of these compounds, and functional information was presented in Chapter 2. It was established that crustaceans are an excellent model organism for developing new analytical chemistry tools for use in neuroscience research and that much remains to be discovered, particularly in identifying the functions of these compounds. New and emerging methods for conducting neuropeptide identification and functional assignment studies were described in Chapter 3. These methods are allowing researchers to conduct untargeted peptidomics research to ensure that all possible compounds of interest are analyzed, and the decision of which compounds to analyze is based on the data itself. A technique to improve NP recovery in microdialysis (MD) sampling termed affinityenhanced MD was developed in Chapter 4. This technique is useful for increasing the concentration of low-abundance compounds in microdialysates and permits quantitation of compounds present endogenously below the limit of quantitation for the analytical instrument in use without concentrating sample and losing temporal resolution. The neuropeptidome of the crayfish Orconectes rusticus was described in Chapter 5 with two different types of MS/MS interpretation software. This led to a good understanding of the NPs present in this species and provided insight on methodology for future experiments of a similar nature. In Chapter 6, a quantification method using a new form of MS/MS acquisition, data-independent acquisition (DIA), was developed. This method used a software package commonly used for the most 340 specific form of MS/MS quantitation currently in use, MRM. Its linearity over the peptide concentration range was observed and it was successfully applied to quantitate NPs in tissue extracts of the blue crab Callinectes sapidus following feeding. These tools were applied in later chapters to behavioral neuroscience experiments. First, microdialysis sampling and DIA MS/MS quantitation were employed to study NPs that change in the hemolymph during feeding in the crab Cancer borealis. This successfully quantified 28 NPs in two replicates from each of three individual crabs. Several significant changes were observed, particularly among peptides of the RFamide-like peptide family, suggesting a possible role for these peptides in feeding. Other peptides had non-significant changes, but most appeared to follow the same trend—rapid increase in hemolymph followed by overshoot and recovery to baseline levels within 3hrs—with the exception of two groups of peptides that were unchanged and one that was released more gradually and did not decrease at the end of the time monitored. In Chapter 8, Cancer irroratus were exposed to three different doses of ethanol under an acute paradigm or one exposure conducted daily for 5 days under a repeated paradigm. The alcohol content of their hemolymph was determined using an enzymatic assay developed as part of this work. Pericardial organ (PO) tissues were analyzed for changes in NP content using a formaldehyde labeling scheme with 3-plex for acute samples and 2-plex for long-term samples. NPs were quantified using a modified version of the DIA MS/MS technique developed in Ch. 6 that allowed for relative quantitation of the isotopic peaks, and every known small-medium-sized crab NP was quantified using this method for long-term exposure samples. Significant changes were observed for 77 of the 475 NPs, and 22 of those had changes greater than 25%. These NPs were then quantified in short-term exposure PO extracts. After exposure to 0.2M ethanol, 12 were significantly increased compared to control. At 0.125M and 0.5M exposure, the values 341 were not significantly different from control. This profile, with an intermediate dose showing the highest levels of NPs, suggests two factors working in opposition. Although it is unclear whether or not alcohol is a rewarding stimulus for crabs, it is hypothesized that ethanol reduces NP release by inhibition of basal levels of secretion due to its activation of the GABA receptor. It is also causing an increase in the amount of NPs released at high doses, so another pathway may be stimulating secretion. However, these effects may also be an artifact of too few experimental animals in some conditions. 9.2 Future Directions This work provides the foundation for many possible projects. The affinity-enhanced MD could be applied in many types of experiments where NP concentration is limited, and similarly there are many potential applications for the DIA MS/MS quantitation methods developed in this dissertation. Of great significance is that all of the DIA MS/MS data could be re-analyzed to quantify any peptide of interest, should additional ones become interesting. The Orconectes rusticus neuropeptidome is expected to be useful in a study of the neuroendocrine mechanisms by which this invasive species adapts rapidly to new environmental conditions and out-competes native crayfish. A project quantifying NP changes in crayfish collected from different lakes with different environmental conditions is currently underway. Additional experiments are being conducted comparing different rearing conditions and genetic factors on behavior in O. rusticus. These animals could also be subjected to neuropeptidomics analysis. Behavioral experiments assessing response to predators and social dominance are commonly conducted in this species, and NPs could be quantified from crayfish after predator exposure or changes in social hierarchy [1-4]. An additional application could be conditioning of crayfish to express a place preference for drugs of abuse, as has been conducted by other 342 researchers [5-9], followed by analysis of NP changes in this potentially new animal model of addiction behavior. Further refinement of feeding experiment parameters, including a quantitative assessment of patterns in this behavior, would be of great benefit, along with additional replicates and a shorter time scale of analysis. The animal could be exposed to different components of the feeding experience to dissect the effects of NPs on each. For instance, it could be given a nonfood item to manipulate with its claws, exposed to the smell of food, or given an infusion of glucose into the hemolymph to determine separately the effects of motor, chemical sensation, and energy level changes that occur during feeding. Characterization of circadian changes in NPs in the crab and under other experimental conditions would be of great interest as well. A NP could be infused to determine what other NPs it causes the release of in vivo, and what behavioral effects it has. Potentially feeding-related peptides could be administered to crabs that are full but have access to food to determine the potency of their effects. Behavioral analysis of the rewarding or aversive effects of ethanol in crabs would be of great use, along with NP analysis of other neural organs. NP microdialysis could be conducted on crabs during ethanol exposure or exposure to another environmental toxin they might be more likely to encounter. A NP with large changes in one of the studies could be infused into the animal via MD concurrent with monitoring behavior and collecting samples to analyze NP release in order to obtain more information about its function. The applications of these techniques are far-reaching. Finally, these techniques could be translated into mammalian models of disease for incorporation into a traditional behavioral neuroscience workflow. The research presented in this dissertation has advanced capabilities to study NPs in a variety of species and under a myriad of physiological or behavioral manipulations. It has also 343 advanced our capabilities in MS/MS analysis by permitting simultaneous identification and quantification from a single instrument run in a data set that can be mined over and over to determine concentration changes in NPs. Opportunities also exist to implement other analytical tools in this system for analysis of different types of compounds. The work conducted in this dissertation has the potential to have great impact on the methodology used for behavioral neuroscience. 9.3. Works Cited 1. Acquistapace, P.; Hazlett, B. A.; Gherardi, F. Unsuccessful predation and learning of predator cues by crayfish. J Crustacean Biol. 2003, 23, 364-370. 2. Klocker, C. A.; Strayer, D. L. Interactions among an invasive crayfish (Orconectes rusticus), a native crayfish (Orconectes limosus), and native bivalves (Sphaeriidae and Unionidae). Northeast Nat. 2004, 11, 167-178. 3. Martin, A. L.; Moore, P. A. Field observations of agonism in the crayfish, Orconectes rusticus: shelter use in a natural environment. Ethology. 2007, 113, 1192-1201. 4. Martin, A. L.; Moore, P. A. The influence of dominance on shelter preference and eviction rates in the crayfish, Orconectes rusticus. Ethology. 2008, 114, 351-360. 5. Dziopa, L.; Imeh-Nathaniel, A.; Baier, D.; Kiel, M.; Sameera, S.; Brager, A.; Beatriz, V.; Nathaniel, T. I. Morphine-conditioned cue alters c-Fos protein expression in the brain of crayfish. Brain Research Bulletin. 2011, 85, 385-395. 6. Huber, R. Amines and motivated behaviors: a simpler systems approach to complex behavioral phenomena. Journal of Comparative Physiology a-Neuroethology Sensory Neural and Behavioral Physiology. 2005, 191, 231-239. 7. Nathaniel, T. I.; Huber, R.; Panksepp, J. Repeated cocaine treatments induce distinct locomotor effects in Crayfish. Brain Research Bulletin. 2012, 87, 328-333. 8. Nathaniel, T. I.; Panksepp, J.; Huber, R. Drug-seeking behavior in an invertebrate system: Evidence of morphineinduced reward, extinction and reinstatement in crayfish. Behavioural Brain Research. 2009, 197, 331-338. 9. Panksepp, J. B.; Huber, R. Ethological analyses of crayfish behavior: a new invertebrate system for measuring the rewarding properties of psychostimulants. Behavioural Brain Research. 2004, 153, 171-180. 344 Appendix A. Protocols A.1. Marine Crustacean Protocols A.1.1. Acquisition and Housing Jonah crabs (Cancer borealis) were obtained from the following three vendors: The Fresh Lobster Company (Gloucester, MA), James Hook and Co. (Boston, MA), and Ocean Resources (Sedgwick, ME). Male Jonah crabs were typically obtained, with the exception of a few shipments from Ocean Resources. Male and female rock crabs (Cancer irroratus) were obtained from Ocean Resources. Jonah and rock crabs were primarily caught by commercial fishermen either as a primary catch in crab pots or as bycatch in lobster pots. Some rock crabs were caught by hand by a diver. Female blue crabs (Callinectes sapidus) were obtained from local markets (Asian Midway Foods, Madison, WI; Garden Asian Market, Middleton, WI) or The Crab Place (Crisfield, MD). Animals obtained from distant vendors were shipped on ice packs with either wet newspaper or seaweed via overnight air mail. Animals from local vendors arrived at the store in boxes of wet newspaper. We took care to select the liveliest of the animals available, and put them in tanks as soon as possible. Animals were maintained in one of two aquarium systems, described in detail in the thesis of Joshua Schmidt [1]. One system was comprised of a commercial tank, complete with pump and filter. The other system employed a modified sump filtration system with protein skimmer. All tanks had crushed gravel as substrate and water quality was monitored regularly. Regular water changes were conducted and animals were fed if they were in the tanks for more than 1 week. Animals were fed white fish, shrimp, crayfish, or crab meat, and were allowed to eat for several hours. The room had temperature, humidity, and light controls. The tanks were maintained at a water temperature of 10-15°C, and salinity was maintained in the 32 to 38 ppt 345 range. The room was on a 12-hr reverse light schedule, with lights off from 10 AM to 10 PM—in order to observe animals during their active period (dark) during normal work hours—and on from 10 PM to 10 AM. A.1.2. Hemolymph Withdrawal Hemolymph was obtained from some animals via the following method, adapted from the thesis of J. Schmidt [1] and other publications [2, 3]. Several items of practical relevance based on personal experience have been noted here. A.1.2.1. Materials Crab (applicable species: Cancer borealis, Cancer irroratus, Callinectes sapidus and Carcinus maenas), needle (25 or 26 gauge, ½” long, Beckton Dickinson and Co. (BD), Franklin Lakes, NJ), plastic 1 mL or 3 mL syringe (BD), sample tubes, ice, aluminum dissecting pan (ThermoFisher Scientific, Waltham, MA), crab saline (440 mM NaCl, 26 mM MgCl2, 13 mM CaCl2, 11.2 mM Trizma base, 11 mM KCl, 5.1 mM maleic acid, pH adjusted to 7.4) A.1.2.2. Instructions 1. Prepare the needle and syringe. It was determined that the smaller gauge needles, despite a slight tendency to clog when proteins precipitate from the hemolymph, were preferable due to producing less damage to the animal. Needles of ½ inch in length were also preferred to 1 inch or 1 ½ inch needles due to the tendency of larger needles to occasionally cause internal damage to vital organs. 2. Remove the animal from the tank and either place with ventral side up in a metal dissecting pan or hold with the thumb and forefinger of the experimenter’s non-dominant hand on the tail section. This hold will make most species unable to use their chelae to harm the experimenter. C. sapidus can have a longer reach and may be able to reach this area, and it should also be noted 346 that the pereiopods of the crab are able to reach and potentially dig into the experimenter’s hand. On C. irroratus, they are particularly sharp. Crabs can be anesthetized on ice for 15 min depending on the needs of the experiment. 3. Uncap the needle and insert at one of the soft joints. The optimal location for a hemolymph withdrawal varies depending on species, amount of hemolymph needed, and experimenter preference. Typically, the best location is between the legs where they join the body. This allows access to the central compartment of the animal, which contains more hemolymph that is in direct contact with neuroendocrine and other organs. The first joint of the legs can also be sampled, and for C. sapidus, the joint where the cheliped meets the body can be accessed from the dorsal side of the animal. It is thus possible to place C. sapidus in the pan with dorsal side up, tucking the legs underneath the body (and unable to harm the experimenter), and obtain hemolymph from this joint. Regardless of placement, place the needle into the soft tissue and withdraw the plunger on the syringe to create a vacuum. Change the orientation of the needle inside the body until hemolymph is observed to enter the syringe. Once a good location is obtained, the syringe and needle are held still until either the desired amount of hemolymph is collected or the flow of hemolymph into the syringe ceases, even with additional pressure applied via the plunger. Multiple locations may need to be used to obtain enough hemolymph. 4. Replace the volume of hemolymph taken if necessary. Using a new needle (and syringe if desired), draw up a volume of crab saline equal to the volume of hemolymph taken. Inject this into a soft joint near the central compartment of the animal. Removing 0.5 mL of hemolymph or more each day will result in the animal’s death after 3-4 days, so if this volume or more is taken, it is recommended to inject an equal volume of crab saline into the animal to replace lost volume. Large volumes of hemolymph withdrawn may result in the animal’s death even if the volume is 347 replaced. The estimated total hemolymph volume of the larger crabs (Cancer spp.) is 7 mL. C. sapidus has a hemolymph volume closer to 5 mL, and C. maenas has around 3 mL. The hemolymph of C. sapidus has a thicker consistency and a tendency to clot more rapidly than the other species and should be stabilized soon after by precipitation of proteins with acidified methanol (see A.3.2.2.). A.1.3. Crab Dissection Modified from the thesis of J. Schmidt [1]. A brief description follows but the primary document should be consulted for more detailed instructions. A.1.3.1. Materials Crab (applicable species: Cancer borealis, Cancer irroratus, Callinectes sapidus, and Carcinus maenas), ice, bucket, aluminum dissecting pan (ThermoFisher Scientific, Waltham, MA), bone cutters (Fine Science Tools (FST), Foster City, CA), semimicro spatula with one round end and one tapered end (ThermoFisher), dissecting scissors: sharp, straight ended, fine or extra fine size (FST), toothed tissue forceps or curved micro mosquito hemostat (FST), curved scissors (15-20 total length, FST), crab saline (see A.1.2.), glass dish filled with black sylgard (ThermoFisher), straight pins (FST), fine dissecting forceps (Dumont #55 straight recommended, FST), spring scissors (5 mm cutting edge recommended, FST), dissecting microscope and light source, microtubes, acidified methanol solution (90/9/1 MeOH/H2O/acetic acid), pipettes. A.1.3.2. Instructions 1. Place the crab on ice for 15 min to anesthetize. 2. Remove the eyestalks and place in sylgard-filled dissection dish with crab saline on ice. 3. Remove the top portion of the carapace and dissect out the stomach. 348 4. Cut this open and rinse in crab saline prior to placing flat in the dissection dish with crab saline on ice. 5. Remove the pericardial ridges and rinse in crab saline. Keep in crab saline on ice until microdissection under a dissecting microscope. 6. Cut open the eyestalks and find the sinus gland (SG). It is immediately below the retina and is a white disc-shaped tissue. Remove with scissors and forceps. 7. Pin the stomach flat in the dish (inside of stomach down) and locate the brain. It will be underneath the epidermis near the rostral end. Remove by cutting the nerves going into this tissue. 8. To dissect the pericardial organ, first cut the lateral part of the pericardial ridges so that they can lay flat, with the side facing the heart up. Pin in place. 9. Locate the pericardial organ underneath a membrane that covers portions of the inside of the pericardial ridges. It will be a filamentous white structure with a more bluish or iridescent tint compared to muscle tissue. 10. Follow the length of the PO throughout the ridge by holding it gently with forceps in one hand and cutting any connective tissue with the spring scissors in the other hand. A diagram of the PO should be referred to in order to obtain as much of the organ as possible. 11. Put dissected tissues immediately into tubes with acidified methanol on ice. A.2. Orconectes rusticus Protocols A.2.1. Collection Orconectes rusticus were kindly provided by Alex Laztka, a graduate student in the laboratory of Jake Vander Zanden, in the Department of Limnology at the University of Wisconsin-Madison, and his co-workers. They were obtained from fresh water lakes in Northern 349 Wisconsin centered around the Limnology Department’s Trout Lake Research Station, on Trout Lake in Boulder Junction, WI. Animals were sampled as part of a National Science Foundation (NSF)-funded Long-Term Ecological Research (LTER) study, and were obtained and transported with the permission of the Wisconsin Department of Natural Resources. Permission for transport must be obtained due to the status of O. rusticus as an invasive species in Wisconsin. Regulations on their transport have been in flux over the last several years so the DNR’s website should be consulted. A recreational fishing permit is also recommended. The trapping method has been shown to preferentially collect male crayfish, and although both genders were collected in our study, around 75% of the animals were male. A.2.1.1. Materials for Catching Crayfish Gee minnow trap (Aquatic Sampling Company, Buffalo, NY), scissors, rope, buoy, bait (beef liver or chicken liver from local butcher), bucket with lid, refrigerator, Styrofoam cooler, ice packs, tape A.2.1.2. Procedure for Catching Crayfish 1. Modify the minnow trap (see Fig. A.1.A.) by increasing the entrance hole to ~5cm to accommodate mature male crayfish. 2. Bait the trap and assemble the two halves into a whole trap (See Fig. A.1.B.). Secure the two halves with the clip. 3. Attach the clip to the buoy by 1m of rope. 4. Place the trap in the lake at a depth of approximately 1 m, in an area where the lake substrate is “cobble,” or rocks approximately 6-8 cm in diameter. 5. In 20-24 hrs, remove the trap. 6. Dump crayfish from the trap into a bucket. 350 7. Separate O. rusticus from other catch by hand and place into a Styrofoam cooler with ice packs. Native crayfish, hybrid crayfish, and juvenile fish may also be caught. The fish and native crayfish should be released. Rusty crayfish are easily distinguished from native species by the presence of two large red spots on the carapace (see Fig. A.2.). It may also be useful to sort by gender/form at this point. O. rusticus should not be released, even if non-desired genders/forms are caught. There are two forms of males, the reproductive form I and the non-reproductive form II. They can be distinguished by the appearance of their reproductive gonopods (See Fig. A.3.). Form I males have feathery tips on the gonopods, whereas form II males have more solid gonopods, often with orange-colored tips. Form I males also have hooks on some leg segments. The female has gonoducts and lacks gonapods. Individuals not desired as samples should be kept, euthanized, and disposed of. Coolers should be taped shut during transport to prevent escape. Transporting to Madison, WI by personal vehicle (a trip of approximately 3.5 hrs) in this manner did not lead to significant mortality. 8. Keep animals in coolers with ice packs or buckets with lids in a refrigerator at 4ºC for approximately 12 hrs. The crayfish should not be stored in water, as the water will quickly become foul and deoxygenated. Storing animals in water in the refrigerator and/or during transport led to approximately 95% mortality in a batch of around 350 crayfish. Stored dry and cold, they enter a state similar to hibernation and can survive longer. A.2.2. Housing Crayfish A.2.2.1. Materials for Housing Crayfish Crayfish, food for crayfish (commercial bottom feeder pellets, fresh fish, etc.), freshwater aquarium setup: 20 gal tank with close-fitting lid, water, crushed gravel, filter (Aquaclear 30, 351 Rolf C. Hagen, Inc., Montreal, Canada), air pump, air line, air stone, fish net breeder isolation box (Lee’s Aquarium and Pet Products, San Marcos, CA) A.2.2.2. Housing Crayfish Suitable aquaria will contain fresh—preferably reverse osmosis—water and crushed gravel substrate. They also should be equipped with appropriate filtration and oxygenation devices, in our case aquarium hobbyist supplies including a filtration unit rated for a tank 10 gal larger than actually used, and an air bubbler with tubing and air stone. No more than 45 individuals should be housed per 20 gallon tank. Care should be taken to ensure a tight fit for the lid of the aquarium as the crayfish can escape through very small holes. Animals should not be stored more than a day at a density of 45 animals/20 gal tank, but lower densities, about 20 per 20 gal tank, can be housed indefinitely. For long-term care of the animals, O. rusticus should be fed and monitored for molting, and regular water changes should be conducted. Crayfish will cannibalize if they are not fed every 2-3 days, and fighting often occurs over food. They can be fed raw meat of any type, but white fish is recommended as it does not foul the water. Pelleted food designed for bottomfeeding fish can also be purchased from a pet store and fed to the crayfish. It must be the kind that will sink to the bottom without falling apart, and usually comes in a disc shape. Enough food should be presented so that all animals have something to eat to prevent fighting. If O. rusticus molt, they should be isolated with the old shell in an isolation net box until their new shell hardens. Other individuals will attack and kill unprotected newly molted individuals. These newly molted individuals can and should consume the old shell for nutritional purposes. Hardening takes 1-2 days. If males and females are housed together, mating may occur. If this is undesirable, separate the genders. Waste will accumulate in the substrate and water, so it is 352 advisable to clean the tank weekly with a 20% water change, or more often if housed in high density. Euthanasia of crayfish can be accomplished by freezing, putting on ice for an extended period of time, or submerging in hot tap water. Bodies of animals should be frozen prior to disposal to ensure that they are not carrying viable eggs. Boiling is an alternative method both for euthanasia and destruction of embryos. A.2.3. Orconectes rusticus Dissection Brain and eyestalk can be dissected from O. rusticus without the aid of a microscope. It is recommended to prepare whole eyestalk extracts instead of dissecting the sinus gland out of the eyestalk due to its miniscule size, which is difficult to locate even under a microscope. A.2.3.1. Materials Crayfish, ice, aluminum dissecting pan (ThermoFisher Scientific, Waltham, MA), dissecting scissors: sharp, straight ended, fine or extra fine size (Fine Science Tools (FST), Foster City, CA), fine forceps (FST), crayfish saline (195 mM NaCl, 13.5 mM CaCl2, 11.2 mM Trizma base, 5.4 mM KCl, 5.1 mM maleic acid, 2.6 mM MgCl2, pH adjusted to 7.4), sample tubes, acidified methanol (90/9/1 MeOH/H2O/acetic acid v/v/v). A.2.3.2. Dissection 1. Put crayfish on ice until movement ceases (approximately 15-20 min). 2. Place animal ventral side up. 3. Use scissors to remove appendages and cut the tail nerve to prevent tail flipping (Fig. A.4.) 4. Place the animal dorsal side up. 5. Remove antennules and antennae. (Fig. A.5.A.) 6. Snap off the rostrum (Fig. A.5.B. 353 7. Remove the eyestalks by cutting at their base, where the stalk meets the shell (Fig. A.6.). Set aside in acidified methanol. 8. Cut the carapace along a line starting below where one eyestalk was located, continuing up toward the rostrum, and then down toward the other eyestalk’s previous location (Fig. A.7.). 9. Gently pull the rostral portion away. (Fig. A.8.) 10. Locate the brain inside the shell, immediately caudal to the rostrum. (Fig. A.9.) 11. Remove with fine forceps and scissors. (Fig. A.10.) Place in a 1.5 mL tube containing 0.5 mL acidified methanol (90:9:1 methanol: water: acetic acid by volume) on ice. 25-30 brains will fit in a single tube of this size. About 20-25 pairs of eyestalks will fit in each tube. 12. Store tissues at -20°C or -80°C (if more than a month will elapse) until extraction. A.3. Sample Preparation Protocols A.3.1. Tissue Extraction A.3.1.1. Materials for Tissue Extraction Tissues, acidified methanol (90:9:1 MeOH:H2O:Acetic acid), microtubes, plastic pestles or glass vial and pestle set, sonicator, microcentrifuge, speedvac, pipettes and tips Note: Keep samples on ice whenever possible. A.3.1.2. Tissue Extraction Procedure 1. For soft tissues, use a plastic pestle to grind the tissue in the collection tube into pieces as small as possible. For whole eyestalks, place in a glass vial and grind with pestle until as homogeneous as possible. Put the eyestalk homogenate back into a microtube. Vortex the sample. 2. Sonicate the tube containing tissue and liquid for 5 min. Do not add heat. 3. Centrifuge 5 min x 16,100 g to separate liquid and solid portions. 354 4. Remove supernatant and set aside in a new tube. 5. Add a volume of acidified methanol equal to the original liquid volume to the pellet. 6. Repeat steps 1-5 twice (for a total of 3 extractions) and combine the supernatants. The pelleted solids can be saved, but it has been determined that the first and second extractions of most tissues contain the most neuropeptides. Subsequent extractions contain primarily lipids and/or no compounds of interest. 7. Evaporate the solvent using the speedvac. Medium or Low heat is recommended. A.3.2. Hemolymph Extraction A.3.2.1. Materials for Hemolymph Extraction. Materials: Hemolymph, acidified methanol (90:9:1 MeOH:H2O:Acetic acid), solution for resuspension, microtubes, plastic pestles (optional), sonicator, microcentrifuge, speedvac, pipettes and tips Note: Keep samples on ice whenever possible. A.3.2.2. Hemolymph Extraction Steps 1. For alcohol studies, collect hemolymph and spin down (5 min x 16,100g). Freeze at -80°C if necessary. Thaw and remove hemolymph for alcohol content analysis. 2. Take 0.5 mL of the remaining hemolymph and add an equal volume of acidified methanol. Vortex vigorously. This can be stored at -20°C. 3. Sonicate the tube for 5 min. Do not use heat. 4. Centrifuge 5 min x 16,100 g to separate liquid and solid portions. 5. Remove supernatant and set aside in a new tube. 6. Add a 0.5 mL acidified methanol to the pellet and resuspend. Vortexing and/or a plastic pestle can be used to aid in breaking up the pellet. 355 7. Repeat steps 3-6 twice (for a total of 3 extractions) and combine the supernatants. The pelleted solids can be saved, but it has been determined that the first and second extractions of most tissues contain the most neuropeptides. Subsequent extractions contain primarily lipids and/or no compounds of interest. 8. Evaporate the solvent using the speedvac. Medium or Low heat is recommended. 9. Resuspend the sample prior to storage at -20°C. The solvent and volume to use for resuspending will be determined by the next sample preparation step. If MWCO is to be conducted, 70/30 H2O/MeOH is used. If formaldehyde labeling is next, use H2O. If desalting is next, use H2O with 0.1% FA. It is important to resuspend prior to freezing, because it has been found that hemolymph samples frozen before resuspension will not achieve full dissolution. A.3.3. Molecular Weight Cutoff (MWCO) Hemolymph samples were usually subjected to molecular weight cutoff (MWCO) following a combination of the manufacturer’s recommended procedure and a published modification [4]. This step is conducted on the crude extracts prior to desalting or other steps. If MWCO was not conducted, the hemolymph samples were typically too thick for efficient desalting. A.3.3.1. MWCO Materials Amicon Utracel 10 kDa MWCO 0.5mL size, 70/30 H2O/MeOH, 50/50 H2O/MeOH, 0.1M NaOH, tubes, centrifuge, speedvac, pipette and tips A.3.3.2. MWCO Procedure 1. Rinse MWCO device with 0.2mL 0.1M NaOH: add solution to tube and spin 4 min x 14,000g. 2. Rinse MWCO with 0.5 mL of 50/50 H2O/MeOH: spin 8 min x 14,000g. 356 3. Load sample, dissolved in 0.5mL 30/70 MeOH/H2O (sonicate if necessary to resuspend), into device, and run through the membrane. This will take 15-20 min x 14,000g. 4. Rinse sample tube with 0.1 mL of 30/70 MeOH/H2O, and run through the membrane. This will take 5 min x 14,000g. This rinse is combined with the flow-through in step 3. 5. Both the flow-through and concentrate were saved, but only the flow-through was used for neuropeptide analysis. 6. Evaporate the solvent for the flow-through using a speedvac (Low/Med heat). A.3.4. Desalting Some tissue and hemolymph extracts were subjected to a desalting step, especially prior to analysis via MALDI ionization or formaldehyde labeling. A.3.4.1. Desalting Materials C18 Zip Tip pipette tips, 10 µL size (Millipore), acetonitrile, 0.1% FA in H2O, 50/50/0.1 ACN/H2O/FA, tubes, speedvac, pipettes and tips. A.3.4.2. Desalting procedure 1. Wet the C18 bed in the tips with 15 µL of acetonitrile (ACN). (Draw up and expel to waste) 2. Equilibrate the tips with 15 µL of 0.1% FA in water. (Draw up and expel to waste) 3. Resuspend the sample in a minimal volume of 0.1% FA in water, up to 60 µL. Sonicate 5 min. This can be done prior to wetting and equilibration of the tips. 4. Bind sample to the tip by drawing it up and expelling it back into the original tube 12 times, with 15 µL moved through the C18 bed each time. 5. Rinse the C18 bed with 15 µL of 0.1% FA in water 4 times to remove salts and highly hydrophilic compounds. For some samples, the first two rinses can be added back to the original 357 sample in case re-analysis will be required and/or the sample is very precious. Other rinses were expelled to waste. 6. Elute hydrophobic compounds, including peptides, from the C18 material by pulling up and dispensing a solution of 50/50/0.1 ACN/H2O/FA in 15 µL amounts 6 times. For MALDI analysis, a smaller volume can be used and the sample spotted directly onto the plate. Alternatively, MALDI matrix solution in 50/50/0.1 ACN/H2O/FA can be used to elute the peptides and spot immediately. 7. For UPLC analysis, dilute with up to 15 µL of 0.1% FA in water to prevent evaporation loss, and add internal standard if necessary. For formaldehyde labeling, dry down using a speedvac. A.3.5. Alternative Desalting Method Using C18 Magnetic Beads For some samples, C18 magnetic beads were used for desalting instead of Zip Tips. These had the advantage of rapid and easy separation from the liquid by holding a magnet to the side of the tube for 30 s. This was particularly useful if samples were viscous and would not go through Zip Tips easily. A.3.5.1. Magnetic C18 Desalting Materials Materials: C18 magnetic beads (Bioclone, Inc., San Diego, CA), magnet, 50/50 MeOH/H2O, acetonitrile, 0.1% FA in H2O, 50/50/0.1 ACN/H2O/FA, tubes, speedvac, pipettes and tips. A.3.5.2. Magnetic C18 Desalting Procedure 1. Suspend C18 beads in 50% MeOH/H2O at a final concentration of 2.5 g/mL. Store at 4°C. 2. Obtain a volume of beads equal to half of the sample volume. 3. Rinse beads 3 times with 2 times the bead volume of 0.1% FA in H2O. Beads are separated from solution by holding a magnet to the side of the tube for 30 s, and the liquid is removed without disturbing the particles. Beads are mixed with liquid by gentle vortexing. 358 4. Add back the original bead volume of 0.1% FA in H2O. 5. Add the beads to the resuspended samples. Allow samples to bind for 2 min at room temperature. 6. Remove the liquid portion and set aside (in case it needs to be re-analyzed) 7. Rinse the beads 3 times with a volume of 0.1% FA in H2O equal to that of the original sample. The first rinse may sometimes be added back to the original liquid portion in the event that reanalysis is required, and/or the sample is very precious. 8. Elute peptides from the beads by incubating with a volume equal to ¼ of the original sample volume of 50/50/0.1 MeOH/H2O/FA for 2 min at room temperature. 9. Save the liquid and repeat step 8 two times. 10. Prepare for LC analysis by adding 0.1% FA in H2O to prevent evaporation and internal standard, or dry in the speedvac. A.3.6. Isotopic Formaldehyde Labeling Both two-plex [5] and a three-plex [6] formaldehyde labeling schemes were used. These were modified from references. The two-plex uses formaldehyde (OCH3) and deuterium formaldehyde (OCD3) as dimethylation agents, and the three-plex adds carbon-13 deuterium formaldehyde (O13CD3) as an additional dimethylation agent. The two-plex protocol does not add sodium ions to the sample due to the use of a milder H- donor, borane pyridine as opposed to NaBH3CN. However, for the heaviest label, D- must be donated instead of H- to produce a total mass shift of four, because simply using carbon-13 deuterium formaldehyde will only produce a mass difference of two, which could have interference from naturally occurring isotopes of the compound labeled with deuterium formaldehyde. For this reason, it uses NaBH3CN for the light and intermediate label, and NaBD3CN for the heavy label. A light label incorporates 2(CH 2) per 359 primary amine; an intermediate label incorporates 2(CD2) per primary amine; and a heavy label incorporates 2(CD2-H+D) per primary amine. All peptide N-termini (except those blocked by pyroglutamation or acetylation) and lysine sidechains will be labeled. A.3.6.1. Two-Plex Formaldehyde Labeling Materials Formaldehyde, 1% solution in water (v/v) (Sigma), D-formaldehyde, 1% solution in water (v/v) (Isotech), borane pyridine, solution in isopropanol (Sigma), water, desalted sample of neuropeptides from 1-3 organs, 100 mM ammonium bicarbonate solution, water bath at 37°C, speedvac, pipette A.3.6.2. Two-Plex Formaldehyde Labeling Instructions 1. Dissolve sample in ~10 µL of water. Sonicate to ensure full dissolution. 2. Prepare formaldehyde, deuterium formaldehyde, and borane pyridine solutions in a chemical fume hood. 3. Add 10 µL of formaldehyde solution to samples for light labeling or 10µL of deuterium formaldehyde solutions to samples for intermediate labeling. 4. Add 10 µL of borane pyridine solution to each sample. 5. Incubate at 37°C for 15 min in the water bath. 6. Take samples out of the water bath and add 10 µL 100 mM ammonium bicarbonate solution. 7. Dry samples in speedvac. Samples will be fully dry when they do not smell of ammonia. A sample may look dry, but still contain ammonia. Highly basic conditions can cause peptide breakdown, so it is necessary to remove all ammonia. A.3.6.3 Materials for Three-Plex Formaldehyde Labeling Formaldehyde, 1% solution in water (v/v) (Sigma), D-formaldehyde, 1% solution in water (v/v) (Isotech), NaBH3CN solution in water (), NaBD3CN solution in water (), water, 360 desalted sample of neuropeptides from 1-3 organs, 1% NH4OH solution in water (Fisher), speedvac, pipette A.3.7. Affinity Agents: Preparation, Elution of Bound Analytes, and In Vitro Bead-binding Experiments A.3.7.1. C18 Silica Microparticles Materials: silica particles coated with octadecyl carbon (C18H37), Microsorb, 3 µM size, 300 Å pore size, (Varian, now part of Agilent), 0.5% bovine serum albumin (BSA) in water solution (w/v) with FA 0.1% (v/v), Handee Spin Cups with paper filter (Pierce, Thermo Fisher Scientific, Rockford, IL), microcentrifuge, speedvac, 0.1% FA in H2O (v/v), tubes, pipettes and tips, vials for UPLC autosampler (Waters, Millford, MA, and Micoliter Analytical Supplies, Suwanee, GA) 1. Dissolve C18 silica microparticles (C18SµP) in water with 0.5% BSA to aid solubility and 0.1% FA to improve NP binding to the beads. 2. Separate C18 silica beads from the solution by employing paper filter spin cups that fit into 1.5mL tubes. The solution containing the solid particles is loaded into the cup, which is inserted into a tube. The tube is then centrifuged, upon which the liquid flows through the filter into the tube, and the solid particles are retained. 3. Elute analyte from the C18SµP by incubating with a solution of 50/50/0.1 ACN/water/formic acid (FA) and centrifuging. 4. Save the flow-through and add to the liquid recovered from washing the beads in order to collect all analyte present in the dialysate. Dry down, resuspend in 0.1% FA, and place in UPLC vials for further analysis. A.3.7.2. Magnetic Beads A.3.7.2.1. Materials for all Magnetic Beads 361 Bio-Mag C18 magnetic beads (Bioclone Inc., San Diego, CA), protein G magnetic microbeads (2 µM diameter, New England Biolabs, Ipswich, MA), protein G magnetic nanobeads (100 nm diameter, Chemicell, Berlin, Germany), neodymium magnet, rabbit polyclonal anti-FMRFamide antibody (Abcam, Cambridge, MA), phosphate buffered saline (PBS, 140 mM NaCl, 8mM Na2HPO4, 2mM KH2PO4, 10mM KCl)., vortexer, oscillating platform, peptide standards (American Peptide, Sunnyvale, CA), 0.1% FA in H2O (v/v), 2% FA in H2O (v/v), 50/50/0.1 ACN/H2O/FA (v/v/v), tubes, pipettes and tips, 96-well plate, 1 mL round, with lid mat (Waters, Millford, MA) Magnetic particles were separated from solution as described previously by holding a neodymium magnet to the side of the tube for 30 s prior to removing the liquid by pipette. Protocols for their preparation and use were developed starting with the manufacturers’ instructions. A.3.7.2.2. C18 Magnetic Beads 1. Prepare as in protocol A.3.5. 2. For in vitro binding assessments, incubate 100 µL beads with 100 µL peptide solution at room temperature for 2 min. 3. Rinse 3 times with 300 µL of PBS or 0.1% FA. These liquid portions may be saved for determination of which analytes did not bind to the particles. 4. Elute analytes from beads by incubating for 2 min at room temperature with 100 µL of 50/50/0.1 ACN/H2O/FA. This may be analyzed directly by UPLC-MS if desired. A.3.7.2.3. Antibody-linked Magnetic Microparticles 1. Rinse 100 µL magnetic beads in solution as provided by the manufacturer three times with 300 µL of PBS. 362 2. Add the antibody at a final dilution of 1:500 (ratio of 50µL beads: 1µL of a 1:10 dilution of serum). In this instance, 2 µL of a 1:10 dilution of antibody serum would be added to the beads along with 98 µL of PBS. 3. Incubate with gentle shaking (attached to vortexer at lowest setting) at 4º C overnight. 4. Rinse 3 times with 300 µL of PBS. 5. Store in 100 µL of PBS. 6. For in vitro binding efficacy assessment, incubate beads with 100 µL of the peptide standard solution (standards in the 10-5-10-6M range) at room temperature with gentle agitation (oscillating platform) for 30 min. 7. Rinse 3 times with 300 µL of PBS to remove unbound analyte. These rinses may be saved and analyzed for peptide content to determine what does not bind to the antibody-coated beads. 8. To elute bound analyte, incubate with 100 µL of 2% FA in H2O at room temperature for 10 min. This may be analyzed directly by UPLC-MS if desired. A.3.7.2.4. Antibody-coated Magnetic Nanoparticles 1. Rinse 100 µL magnetic beads in solution as provided by the manufacturer three times with 300 µL of PBS. 2. Add the antibody at a final dilution of 1:500 (ratio of 50µL beads: 1µL of a 1:10 dilution of serum). In this instance, 2 µL of a 1:10 dilution of antibody serum would be added to the beads along with 98 µL of PBS. An alternate, higher dilution of 1:50 can be used, in this case 10 µL of a 1:10 dilution of serum and 90 µL of PBS added to the beads. 3. Incubate with gentle shaking (attached to vortexer at lowest setting) at room temperature for 15 min. 4. Rinse 3 times with 300 µL of PBS. 363 5. Store in 100 µL of PBS. 6. For in vitro binding efficacy assessment, incubate beads with 100 µL of the peptide standard solution (standards in the 10-5-10-6M range) at room temperature with gentle agitation (oscillating platform) for 15 min. 7. Rinse 3 times with 300 µL of PBS to remove unbound analyte. These rinses may be saved and analyzed for peptide content to determine what does not bind to the antibody-coated beads. 8. To elute bound analyte, incubate with 100 µL of 2% FA in H2O at room temperature for 2 min. This may be analyzed directly by UPLC-MS if desired. Note that nanoparticles will aggregate if they are sonicated, placed on ice, or allowed to dry out. A.4. Myoglobin Digest Internal Standard Equine skeletal myoglobin (ERA, Colden, CO) was digested with trypsin (bovine pancreas, Sigma-Aldrich, St. Louis, MO) following a published procedure [7]. Myoglobin was dissolved in 100 mM NH4HCO3 at 0.5 mg/mL. Trypsin was dissolved in the same solution at 1 µg/mL and added to the myoglobin solution at an enzyme: substrate ratio of 1:10. This was then diluted 1:1 with methanol and placed in a water bath at 37°C for 45 min. The reaction was stopped by adding ice-cold acetic acid to a final concentration of 5%. The sample was then spun at 15,100 x g for 5 min. A.5. Microdialysis A.5.1. Materials and Preparation CMA/20 Elite probes with 4 mm membranes of polyarylether sulfone (PAES) (CMA Microdialysis (Harvard Apparatus, Holliston, MA, USA)); syringe pump: CMA/102, KD Scientific 100 (KD Scientific Inc., Holliston, MA, USA), Harvard22 (Harvard Apparatus, Holliston, MA, USA); FEP tubing (CMA), PEEK tubing (Upchurch-Scientific, Idex Health and 364 Science, Oak Harbor, WA, USA); flanged connectors: CMA, BASi (West Lafayette, IN, USA); syringe: BD (Franklin Lakes, NJ, USA) plastic 1 mL or 3 mL; 21 gauge Luer-lock needles (included with CMA 20 series probes) with the sharp points removed by a grinding wheel on a rotary tool (Dremel, Robert Bosch LLC, Farmington Hills, MI, USA); water; crab saline; peptide standards (American Peptide, Sunnyvale, CA); tubes; pipettes and tips 1. Rinse MD probe with water or crab saline prior to use and store in liquid throughout the study to maintain the integrity of the membrane. A.5.2. In Vitro Microdialysis Additional materials: tubes, 1000µL pipette tip, scissors, orbital rotating platform, phosphate buffered saline (PBS), 96-well plate, 1 mL round, with lid mat (Waters, Millford, MA) 1. Immerse the tip of the probe or probes into a vial via a home-built apparatus to hold the probes in place. For a single probe, a 1000µL pipette tip can be cut in half and the top part placed inside a 1.5 mL tube. The probe is placed inside this holder. 2. Put microdialysis medium in the vial. This is a solution of phosphate buffered saline (PBS) solution with neuropeptide standards of interest in it at known concentrations in the micromolar range. 3. The entire vial with probe holder is placed on an orbital rotating platform to produce constant mixing of the microdialysis medium. 4. Obtain a sample of the medium prior to starting microdialysis and set aside. 5. Start flow of the perfusing liquid (0.5 µL/min) via the syringe pump. Permit the probe to equilibrate at the flow rate of the experiment for 30 min before starting dialysate collection. 6. Collect technical replicates as consecutive 30 min samples of the liquid flowing out of the tubing. A minimum of three should be collected. 365 7. Obtain a sample of the medium after all technical replicates are collected. Samples from the medium taken before and after the experiment were used to determine the relative recovery percentage. 8. Medium samples and dialysate can be placed immediately in a 96-well sample plate for UPLC-QTOF analysis. A.5.3. In Vitro Microdialysis-Affinity enhanced Additional materials: affinity agent (see protocol above), ball bearing (1/8 inch, Wheels Manufacturing, Louisville, CO, USA), rocking platform 1. Set up apparatus as in steps 1-3, with the exception that the probe is immersed in PBS alone. 2. Put a clean ball bearing into the syringe. 3. Prepare perfusate with affinity agent by diluting 50µL of bead solution (as it arrived from the manufacturer) to 3mL (a dilution of 1:60) or to 0.5mL (a 1:10 dilution) with PBS and put into syringe. Although the concentrations of beads in mg/mL vary depending on the affinity agent, it was determined that they have equal activity per mL, as the all manufacturers’ protocols recommend the same ratio of beads to sample, i.e. 50µL beads with 0.5 mL cell lysate. 4. Put syringe in pump and place on rocking platform at an angle to the axis of rotation. Start the platform to rock, causing the ball bearing to roll up and down the syringe. 5. Start perfusing the system at a higher flow rate until 3 times the volume of the system has passed through the tubing. 6. Place the probe in microdialysis medium as in step 2 above. Obtain a sample of the medium and set aside. 7. Start the pump at the flow rate desired (0.5 µL/min). Allow equilibration as in step 5 of previous protocol. Collect samples as in steps 6 and 7. Put medium samples in plate as in step 8. 366 8. For samples containing affinity agent, separate solids from perfusate liquid, which can be placed in the 96-well plate. Elute bound NPs from the solid particles according to each particle’s instructions and add to the 96-well plate. A.5.4. Crab Microdialysis A.5.4.1. Crab Microdialysis (MD) Surgery Additional materials: crab, dissection tray, ice, 30% bleach, ethanol, rotary tool (Dremel, as above), 1/32” drill bit for Dremel, hot glue and hot glue gun, food coloring, Loctite super glue gel (Loctite, Westlake, OH), Mighty Putty (epoxy materials suspended in a clay-like base, Mighty Putty, North Wales, PA), plastic electrical cord protector (from hardware store), refrigerated fraction collector (BASi Honeycomb, Bioanalytical Systems, Inc. Indianapolis, IN). Figures are provided: See Figs. A.11-19. Place a crab on ice for 15 min to anesthetize. Depth of anesthesia is monitored by watching for voluntary movement. It is maintained by keeping the crab on ice throughout the surgery. 2. Wash the crab’s shell in the area immediately above the pericardial sinus with 30% bleach followed by ethanol. Scrubbing should be vigorous and conducted until color no longer appears to come off. This can be done while the crab is being anesthetized. 3. Score the shell in this area by making cross-hatching diagonal lines using a grinding wheel on a rotary tool. 4. Apply a small amount of hot glue to the MD probe on the shaft above the membrane area to mark the deepest point to which the probe will be inserted. The tubing for the probe should already be connected and the integrity of the tubing and probe can be checked throughout surgery by gently depressing the syringe plunger. Artificial crab saline should be the perfusate. 367 5. Drill a 1/32” hole in a location estimated to be immediately above the heart. Do not apply pressure to the drill bit while drilling, and stop once the shell has been drilled through. If hemolymph does not come out of the hole, the epidermis has not been punctured. Use the needle that comes with the probe to gently puncture the epidermis. Dry off the shell 6. Carefully insert the probe. This should stop hemolymph from leaking out of the hole. 7. Apply Loctite super glue gel in a small circle around the probe. 8. Mix the Mighty Putty and apply on top of this glue circle. It is best to mix the material in the last few minutes of anesthesia induction, and form a log of approximately 2” in length with the diameter of a pen. This can then be placed on the shell and molded into a cone shape around the exposed part of the probe. 9. Fill any remaining voids with super glue gel. 10. Insert the probe tubing into a 1’ section of plastic tubing for protecting electrical cords. 11. Attach this tubing to the putty cone on the crab’s shell with hot glue. 12. Keep the crab on ice until the putty is no longer pliable, and then replace in the tank. The surgery should not take more than 45 min, and a typical surgery is around 25 min. The crab may not wake up if it is kept on ice for longer than 45 min. 13. Start the syringe pump and refrigerated fraction collector as desired. 14. When starting an experiment, determine the dead volume from the probe tip to collection vial as follows and incorporate this information to collect the proper samples. Tubing volume: 1.8 µL/10cm, probe tip to outlet: 3.0 µL, fraction collector needle: 7.1 µL 15. After the experiment is concluded, which be no more than 10 days after surgery, sacrifice the crab. The crab can be kept for other experiments if desired but the probe will probably not work anymore. 368 16. Follow the normal dissection protocol with the following changes. 17. While the crab is on ice, cut the MD tubing and protective tubing short and connect a syringe with 0.1mL of diluted food coloring (dilute 1:1 v:v) to the inlet tube. 18. Manually push the dye through the tubing and through the outlet tube. Make sure at least a few drops go all the way through. 19. When dissecting, be careful not to disturb the probe inside the shell. 20. At the point when you remove the shell over the pericardial region, take additional care. Note the location of the probe and/or dye spot. There should be a spot of dye on the crab’s heart. A.5.4.2. Crab Microdialysis-Affinity Enhanced 1. Set up syringe as indicated for in vitro affinity-enhanced MD, with the exception that crab saline is used to dilute the affinity agent instead of PBS. 2. Perfuse the system with three times its dead volume, as above, prior to equilibration and sample collection. 3. Collect samples manually into tubes suspended over ice. It is not recommended to use the fraction collector for AE-MD due to the potential for clogging and interactions between the magnetic beads and metal fraction collector needle. The tubes should not be immersed in ice, as this causes nanoparticles to aggregate. A.6. Instrumental Analysis A.6.1. Packing Chemically-Fritted C18 Columns with Integrated Tips Materials: Waters nanoAcquity UPLC system (Millford, MA), polyimide-coated glass capillary, 360 µM OD, 75 µm ID (Polymicro Technologies, Phoenix, AZ), Magic C18 particles (Michrom, Auburn, CA, USA), 3 µm diameter, 100Å pore size), potassium silicate (Kasil®, PQ Corporation, Malvern, PA) , formamide (Sigma-Aldrich), tetramethylammonium silicate (15- 369 20% in water, Sigma-Aldrich), column packing setup and gas cylinder (manufactured by UW Chemistry Shop, similar products available commercially), 2 mL vial or LC sample vial, ultramicro stir bar, sonicator, various other fittings (IDEX Health & Science LLC), capillary cutter, P-2000 Laser Puller (Sutter Instruments, Novato, CA), methanol, isopropanol, soldering iron, 0.1% FA in ACN (Optima or LC-MS grade), 0.1% FA in H2O (Optima or LC-MS grade) 1. Cut a section of capillary ~15 cm long. Burn off ~5 cm of polyimide coating from the middle, creating a clear window. Note: capillaries are much more fragile after the protective polyimide coating has been removed. 2. Mix together 100 µL potassium silicate and 100 µL tetramethylammonium silicate. 3. Add 20 µL formamide. 4. Heat up the soldering iron. 5. Dip the end of the capillary into the silicate solution. Capillary action will draw the solution up, Allow it to reach past the window. 6. Touch the tip of a hot soldering iron to the places where you want to make frits. This should be within the clear window, but closer to the part that is coated. The laser must shine on the middle to properly pull the tip and you don’t want anything else to be in that area. The area touched should briefly turn white within 5 sec. Once you see it turn white, take the soldering iron off. If it doesn’t turn white, you probably didn’t see it happen. Holding the soldering iron there too long can cause the frit to be too solid. 7. Rinse the capillary with methanol using the bomb or acetonitrile using the UPLC system. 8. Dry the capillary with the bomb or a syringe fitted with the proper adapter. Note: It is frequently useful to make several at one time, as the frit solution must be used within a few hours of being made, and these things are very fragile so it is helpful to have spares. Do not 370 allow the capillaries to sit for more than an hour with the frit solution in them, as it will start to set up. 9. Prepare column packing material. This should be ~10mg of C18 particles in 1mL of 3:1 methanol: isopropanol. Add the stir bar and sonicate for >15 sec. 10. Put the packing material in the vial in the bomb and close the bomb. 11. Put the capillary to be packed into the bomb. Make sure the end of the capillary is only a few mm above the stir bar by pushing it down until it hits the bottom of the vial, and then taking it back out a few mm. 12. Tighten the capillary in the bomb and close the vent valve. 13. Put pressure on the regulator and ~100psi on the bomb. You should start to see small clumps of C18 particles travelling up the capillary and stopping at the frit, and liquid should come out the top. 14. Once it is packed ~2cm, put full pressure on the bomb ( up to 1500 psi). Allow the column to pack for ~40 min. You can’t see part of the column because it is inside the bomb, so you will have to let it pack for longer than you expect. 15. Start to vent the bomb slowly. First, turn the pressure off at the regulator. Then open the vent valve very slowly. You should only barely be able to hear gas escaping. Sometimes you can check by putting an unstretched glove over the outlet—if it balloons up, gas is flowing out. Venting should take at least 3 hrs. 16. Attach the column to the UPLC outlet. 17. Rinse the column with 95%B at a flow rate that gives you a pressure of ~3000psi. 18. Reduce the flow rate and switch to 50/50, then increase again to obtain 3000psi. 19. Do the same thing for 95%A. Allow it to pack at 95%A and 3000 psi for at least 15 min. 371 20. Return the column to 50/50 and unhook it. 21. Dry the column on the air bomb. 22. Pull the tip. The other side will make a nice ESI emitter tip for the Synapt. A.6.2. Analytical Scale UPLC for Pre-Fractionation Materials: neutral pH solvents (7.4, solvent A: 25mM NH4HCO2, solvent B: 9:1 ACN: 25mM NH4HCO2), reversed-phase Inertsil ODS-4 column, 3µm, 2.1 x 150 mm (GL Sciences, Tokyo, Japan), Alliance HPLC with UV detection (Waters, Milford, MA), Rainin Dynamax Model FC-4 fraction collector, tubes, speedvac 1. Inject sample on a 120 min RP separation that will run 95%A to 5%A and back. 2. Collect fractions every 3 min after the first 5 min have elapsed. 3. Dry samples down with speedvac. 4. Combine early eluting fractions with late eluting fractions to reduce the total number of samples while maintaining separation. A.6.3. UPLC-MS and UPLC-MS/MS Analysis Materials: Waters nanoAcquity UPLC system, Zorbax 300SB-C18, 300 µm x 5 mm trap column (Agilent Technologies Inc., Santa Clara, CA), holder for 5 mm trap cartridges (Agilent), stainless steel nano-tee (IDEX Health & Science LLC, Oak Harbor, WA), conductive filter capsule with 2 µm filter and holding bracket (IDEX), QTOF Micro (Micromass, Cambridge, UK), Synapt G2 (Waters), home packed column, C18 reverse-phase column (BEH130 C18, 1.7 µm, 75 µm x 100 mm, Waters), trap column (Waters), home-pulled ESI tips, 0.1% FA in ACN (Optima or LC-MS grade), 0.1% FA in H2O (Optima or LC-MS grade) 1) Set up UPLC conditions. Typically, an acidic reversed phase gradient with 0.1% FA in water and 0.1% in ACN as mobile phase was employed with run time ranging from 25 min to 120 min 372 depending on the application. The change in gradient ranged from 5%B to 95%B or 10%B to 45%B (for highly hydrophilic peptides). A trapping step is employed, ranging in length from 1 min for clean samples to 5 min for very salty samples. For the Synapt, a commercial column is used with a column heater and a separate home-pulled capillary emitter tip. For the QTOF micro, a home-packed column with integrated emitter tip is used. 2) Set up MS conditions. Micromass (currently Waters) QTOF micro: can be run in MS only mode over the mass range m/z 500-700 for highly accurate quantitation of a few known peptides in that mass range. DDA can also be run on this instrument but it is not recommended due to sensitivity issues. This instrument is recommended for high concentration samples and/or standards. Waters Synapt G2 mass spectrometer: For DIA MS/MS mode, use a high energy scan having a voltage ramp from 25 to 65 V. For DDA, use a basic file that selects 3 precursors at a time. Use Glu-fibrinopeptide for lockspray calibration. A.7. Data Analysis A.7.1. QTOF Micro Data Analysis 1. Use QuanLynx software (Waters). Input the m/z’s of interest for your analytes manually. 2. Once the QuanLynx method is created, use it to analyze your data. The data must be in the QuanLynx project folder, you must be using the QuanLynx project, and you must have the correct file names in the sample list. 3. Manually check the identity of each peak. To do this, open a chromatogram in MassLynx and generate the XIC for the m/z you are looking at. Combine spectra across the peak and search for the analyte in the spectrum. Ensure it is high intensity, not an isotope of another compound, and has the correct charge state. 373 4. Manually check the integration of each peak. 5. Export results to Excel and or JMP Statistical Software (Version 9.0.2 SAS Institute, Inc., Cary, NC, USA). A.7.2. Synapt G2 Data Analysis—DDA 1) Load results into ProteinLynx Global Server (Waters). Make sure it is configured to output a .pkl file (one per sample) and note the location where these are created. 2) Create a preprocessing file to correct with the lockspray. 3) Attach the preprocessing file to the raw data and run. This will generate the .pkl file. 4) This file can be uploaded to Mascot or PEAKS for database search and/or de novo sequencing. The following PTMs are used: C-terminal amidation, pyroglutamic acid, methionine oxidation. No enzyme. Sometimes Cys dehydration is also used. 5) Export results to Microsoft Excel. A.7.3. Synapt G2 Data Analysis—DIA 1) If desired, generate a .pkl using the correct preprocessing file. This can be searched in Mascot but cannot be used in PEAKS. 2) More typically, quantitation will be conducted. Generate the appropriate quantitation file in Skyline SRM software [8] (https://skyline.gs.washington.edu/labkey/project/home/software/Skyline/begin.view). The website has downloads and helpful tutorial videos and documents. I have created the following files: 475 crab NPs labeled with H and D formaldehyde, 475 crab NPs unlabeled, 475+CPRPs and other long NPs unlabeled, NPs from Ruibing’s feeding paper [5] and myoglobin tryptic digest, sets of neuropeptide standards, NPs from long-term ethanol exposure labeled with 3-plex dimethylation. 374 3) Upload your data files into Skyline. 4) Manually check integrations for alignment with predicted retention time, retention time alignment across replicates, choice of the correct peak, uniqueness of the peak for that peptide, etc. The retention time comparison and peak area comparison windows can be helpful for this. 5) Export desired results. I have made an export file that I like to use called CMSexport. Analyze in Excel and/or JMP. Make sure to sort peptides by name and sum all transitions from the same peptide. Additional Skyline settings: Modifications were added to peptides manually. A non-specific enzyme type was used, with cleavage at every amino acid, and 9 missed cleavages permitted, the most allowed. Skyline does not have a “no enzyme” option built in and this is the closest approximation possible. Most NPs are shorter than 9 amino acids, this should be acceptable. In addition, Skyline will quantify peptides that do not meet its filter criteria if they are input manually and the exception to the filters is noted by the user. The iRT function with retention time predictor was used, calibrated off of results from Mascot identifications of (Matrix Science, London, UK) equine myoglobin tryptic peptides in a previous LC run of the myoglobin digest alone under the same conditions. The 2+ and 3+ precursors were monitored to the 4 most abundant 1+ and 2+ b and y ions, and in some cases (CCAP, somatostatin-14) the precursor ion itself was also monitored. MS/MS filtering parameters were set to DIA with MSe isolation, and 10,000 resolving power. A.8. Alcohol Dehydrogenase (ADH) Assay (with thanks to Lauren Putterman) A.8.1. Materials Alcohol dehydrogenase (ADH) from Saccharomyces cerevisiae, ≥300 units/mg protein (Sigma Aldrich, St. Louis, MO), phenazine methosulfate (PMS, Sigma Aldrich), β-nicotinamide adenine 375 dinucleotide hydrate (NAD+), thiazolyl blue tetrazolium bromide (MTT), ethanol 100% (Sigma Aldrich), crab, 1mL plastic syringe with ½” 26 ga needle (Becton Dickinson, Franklin Lakes, NJ), crab saline, centrifuge, tubes, ice, laboratory incubator/oven capable of 50°C, microplate reader capable of reading absorbance at 570 and 655nm: Tecan Ultra 384 with XFluor4 software (Tecan Group Ltd., Männedorf, Switzerland) with filters from Omega Optical (Omega Optical, Inv., Brattleboro, VT), microplates (96-well Perkin Elmer Isoplate, white frame and clear well, Perkin Elmer, Waltham, MA), phosphate-buffered saline (PBS, 137 mM NaCl, 2.7 mM KCl, 8.1 mM NaH2PO4, 1.76 mM KH2PO4, pH 7.4), tris buffer (0.1 M Tris-HCl, 0.1% (w/v) Triton X100, pH 8.5), sodium dodecyl sulfate (SDS) 20% (w/v) in 50/50 dimethylformamide (DMF)/H2O (v/v), rotating platform (Fisher Scientific Clinical Rotator), pipettes (multi-channel are useful) Note: Several kinds of ADH are available. Some have NADH in them, and are not suitable for this assay. Sigma Adrich product # A3263 is suitable. Storage notes: Plasma from unaffected crabs can be stored for weeks in the -80ºC freezer. Calibration curves are also stable for up to 2 weeks. Tris buffer should be stored at 4°C. MTT should be stored at 6 mM concentration in PBS in the dark at room temperature. This solution can be kept for up to a month. For each assay, dilute with PBS to 600 µM; do not keep this solution more than a day. ADH (7.33x106 U/L) and NAD+ (37.7 mM) solutions should be made up weekly and stored at 4°C. PMS is stored at 50 mM or 50 µM in at 4°C in the dark. These solutions are stable for up to a month. A solution of 5 pM PMS is made on the day of the experiment and not stored. SDS/DMF should not be stored in the refrigerator and is stable for several months. A.8.2. Assay Procedure 376 1. Collect hemolymph from the crab as described previously. A very small amount is required for this assay, but additional hemolymph from an un-dosed crab will be needed to make blanks and calibration curve samples. Hemolymph should be put on ice immediately after it is collected. 2. Spin down at 16.1 rcf for 5 minutes to get crab plasma, keep on ice. The plasma can be stored, especially “blank” plasma. For “blank” plasma, it appears to be stable for several weeks. 3. Turn on oven (50 °C). 4. Make calibration curve solutions in crab plasma. It is easier to make a 100 mM EtOH solution first, then dilute to 10 mM. Use this to make 5, 3, and 1 mM solutions in plasma. Use the 1 mM solution to make a 0.1 mM solution. 5. To each well of the plate to be used, add 76.7 uL of Tris-HCl Buffer 6. Add 10 uL of 600 uM MTT to each well. 7. Add 10 uL of 37.7 mM NAD+ solution to each well. 8. Add 4 uL of sample to each well. This should include a calibration curve of 0.1 mM, 1 mM, 3 mM, 5 mM, and 10 mM, sometimes done in duplicate, and a blank in addition to the actual samples. Samples from crabs exposed to alcohol should also be diluted 1:3 with Tris buffer and analyzed at this concentration to ensure they fit within the assay’s linear range. 9. Add 2 uL of 5 pM PMS to each well. 10. Add 4 uL of 7.33x106 U/L active ADH to each well 11. Mix plate on rotating platform for about 1 minute. 12. Place plate in 50 °C oven/incubator for 60 minutes. 13. Stop the reaction by adding 100 uL of inhibitor (SDS/DMF solution) to each well. 14. Mix plate on rotating platform for about 1 minute. 15. Determine the absorbance at 570 nm and 655 nm using the plate reader. 377 16. The software will output a .csv file that can be analyzed in Microsoft Excel. A.8.3. ADH Assay Analysis 1. Adjust for the blank, if not already done during data acquisition. 2. Determine the difference between absorbance at 570 nm and 655 nm. 3. Plot the concentration of ethanol versus the 570 nm-660 nm absorbance value and generate a linear fit. Sometimes, the 10 mM point will have to be excluded from the fit. 4. Determine the slope and intercept of the linear regression. Use these values to calculate the concentrations of the unknown samples (concentration = (absorbance – intercept)/slope). If the values fall outside of the range for which the calibration curve was generated, the values cannot be trusted. Dilute samples if necessary. A.9. Works Cited 1. Schmidt, J. J. From crabs to hamsters: Bioanalytical mass spectrometry for peptidomic analysis and biomarker discovery [3279009]. United States -- Wisconsin: The University of Wisconsin - Madison; 2007. 2. Behrens, H. L.; Chen, R.; Li, L. Combining microdialysis, NanoLC-MS, and MALDI-TOF/TOF to detect neuropeptides secreted in the crab, Cancer borealis. Anal Chem. 2008, 80, 6949-6958. 3. Chen, R.; Ma, M.; Hui, L.; Zhang, J.; Li, L. Measurement of neuropeptides in crustacean hemolymph via MALDI mass spectrometry. J Am Soc Mass Spectrom. 2009, 20, 708-718. 4. Cunningham, R.; Wang, J.; Wellner, D.; Li, L. Investigation and reduction of sub-microgram peptide loss using molecular weight cut-off fractionation prior to mass spectrometric analysis. J Mass Spectrom. 2012, 47, 1327-1332. 5. Chen, R.; Hui, L.; Cape, S. S.; Wang, J.; Li, L. Comparative Neuropeptidomic Analysis of Food Intake via a Multi-faceted Mass Spectrometric Approach. ACS Chem Neurosci. 2010, 1, 204-214. 6. Boersema, P. J.; Raijmakers, R.; Lemeer, S.; Mohammed, S.; Heck, A. J. R. Multiplex peptide stable isotope dimethyl labeling for quantitative proteomics. Nat Protoc. 2009, 4, 484-494. 7. Li, F.; Schmerberg, C. M.; Ji, Q. C. Accelerated tryptic digestion of proteins in plasma for absolute quantitation using a protein internal standard by liquid chromatography/tandem mass spectrometry. Rapid Commun Mass Spectrom. 2009, 23, 729-732. 8. MacLean, B.; Tomazela, D. M.; Shulman, N.; Chambers, M.; Finney, G. L.; Frewen, B.; Kern, R.; Tabb, D. L.; Liebler, D. C.; MacCoss, M. J. Skyline: an open source document editor for creating and analyzing targeted proteomics experiments. Bioinformatics. 2010, 26, 966-968. A.10. Figures 378 A.1. Modified minnow trap for collection of Orconectes rusticus A. B. A. Half of a trap, showing the enlarged hole for entry of crayfish, several crayfish in the trap, and the remains of the bait after a day in the water (pinkish blob). B. A full assembled trap. Hooks and loops interlock on opposite halves, and two loops line up for securing with a pin-like fastener. That fastener is attached to a small buoy (whitish oval) by a length of string. This trap has been pulled out of the lake after a day, and contains several crayfish along with small fish bycatch. Figure A.2. Identification of Orconectes rusticus A. B. Rusty crayfish are primarily green or brown, with red or orange accents. Dark reddish spots are a defining feature on the carapace. Arrows indicate the location of these spots in A. overhead and B. side views. 379 Figure A. 3. Genders and Stages of Orconectes rusticus b. c. b. a. a. d. Male form I, form II, and female adult O. rusticus (Left to right). Males have gonapods (a.), but those of the form I male are more feathery at the ends (b.). Male form II gonapods are more substantial and often orange at the ends. Male form I crayfish also have hooks on their legs (c.). Females lack gonapods and instead have a gonoduct (d.). Figure A.4. Dissection Step 3 A. A. Severing the tail nerve. B. Removing the legs. B. 380 Figure A.5. Dissection Step 5-6 A. B. A. Removing anntennules and antennae. C. Snapping off the rostrum. Figure A.6. Dissection Step 7 A . B . A. Cut the eyestalk at the base. B. A better view of the exposed eyestalks. Figure A.7. Dissection Step 8. 381 Make a cut all the way around the carapace as indicated. Figure A.8. Dissection Step 9 Separate the rostral portion of the shell. Figure A.9. Dissection Step 10 Locate the brain inside the rostral portion of the shell. It is the whitish structure the arrow is pointing to. Figure A.10. A. B . 382 A. Cut the nerves surrounding the brain and holding it in place. B. Remove with forceps. Figures A.11-19. Microdialysis 383 384 385 386 387 Appendix B. Crustacean Neuropeptide Database >gi|001 |LiLab|LL001 AAPYAFGL >gi|002 |LiLab|LL002 AASPYSFGL >gi|003 |LiLab|LL003 AGGAYSFGL >gi|004 |LiLab|LL004 AGLYSYGL >gi|005 |LiLab|LL005 ARPYAFGL >gi|006 |LiLab|LL006 DGPYSFGL >gi|007 |LiLab|LL007 DPYAFGLRHTSFVLYAFGL >gi|008 |LiLab|LL008 DRPYSFGL >gi|009 |LiLab|LL009 EPYEFGL >gi|010 |LiLab|LL010 ERPYSFGL >gi|011 |LiLab|LL011 FNGCNFGL >gi|012 |LiLab|LL012 FSGASPYGL >gi|013 |LiLab|LL013 FSGTYNFGL >gi|014 |LiLab|LL014 GDPYAFGL >gi|015 |LiLab|LL015 GGAYSFGL >gi|016 |LiLab|LL016 GKPYAFGL >gi|017 |LiLab|LL017 GPYSFGL >gi|018 |LiLab|LL018 GQYAFGL >gi|019 |LiLab|LL019 GRYSFGL >gi|020 |LiLab|LL020 GSGQYAFGL >gi|021 |LiLab|LL021 HGTEGPYPFGL >gi|022 |LiLab|LL022 HSPSSASYDFGL >gi|023 |LiLab|LL023 KLPYSFGL >gi|024 |LiLab|LL024 LKAYDFGL >gi|025 |LiLab|LL025 LVKYSFGL >gi|026 |LiLab|LL026 NPYSFGL >gi|027 |LiLab|LL027 Allatostatin A-type AAPYAFGLamide Cmaen Br/PO Allatostatin A-type AASPYSFGLamide Cmaen PO Allatostatin A-type AGGAYSFGLamide Cb br,PO Allatostatin A-type AGLYSYGLamide Cb PO Allatostatin A-type ARPYAFGLamide Cmaen PO Allatostatin A-type DGPYSFGLamide Csap PO,br Cb PO Allatostatin A-type DPYAFGLRHTSFVLYAFGLamide Csap Allatostatin A-type DRPYSFGLamide Cb PO Allatostatin A-type EPYEFGLamide Cmaen Br Allatostatin A-type ERPYSFGLamide Cp.PO/Cb.PO. Allatostatin A-type FNGCNFGLamide Csap Allatostatin A-type FSGASPYGLamide Cmaen Br/PO Allatostatin A-type FSGTYNFGLamide Csap PO,br Allatostatin A-type GDPYAFGLamide Cb br,PO Allatostatin A-type GGAYSFGLamide Cb.PO. Allatostatin A-type GKPYAFGLamide Cmaen Br/PO Allatostatin A-type GPYSFGLamide Cb PO Allatostatin A-type GQYAFGLamide Cb.PO. Allatostatin A-type GRYSFGLamide Csap br Allatostatin A-type GSGQYAFGLamide Cb.PO. Allatostatin A-type HGTEGPYPFGLamide Csap Allatostatin A-type HSPSSASYDFGLamide Cb PO Allatostatin A-type KLPYSFGLamide Cmaen Br Allatostatin A-type LKAYDFGLamide Cmaen Br Allatostatin A-type LVKYSFGLamide Cb PO,CoG Allatostatin A-type NPYSFGLamide Csap PO,br Cb PO Allatostatin A-type PADLYEFGLamide Cb.PO. 388 PADLYEFGL >gi|028 |LiLab|LL028 Allatostatin A-type PATDLYAFGLamide Cb.PO. PATDLYAFGL >gi|029 |LiLab|LL029 Allatostatin A-type PDMYGFGLamide Cb.PO. PDMYGFGL >gi|030 |LiLab|LL030 Allatostatin A-type pQRAYSFGLamide Csap PO Cb ,br,PO,CoG QRAYSFGL >gi|031 |LiLab|LL031 Allatostatin A-type pQRDYSFGLamide Cb PO QRDYSFGL >gi|032 |LiLab|LL032 Allatostatin A-type pQRPYSFGLamide Cp.PO QRPYSFGL >gi|033 |LiLab|LL033 Allatostatin A-type pQRTYSFGLamide Cb.PO. QRTYSFGL >gi|034 |LiLab|LL034 Allatostatin A-type PRDYAFGLamide Csap PO,br Cb br,PO,STG,CoG PRDYAFGL >gi|035 |LiLab|LL035 Allatostatin A-type PRVYSFGLamide Csap PO PRVYSFGL >gi|036 |LiLab|LL036 Allatostatin A-type PSMYAFGLamide Cb.PO. PSMYAFGL >gi|037 |LiLab|LL037 Allatostatin A-type RGPYAFGLamide Cmaen Br/PO RGPYAFGL >gi|038 |LiLab|LL038 Allatostatin A-type SGHYIFGLamide Csap PO SGHYIFGL >gi|039 |LiLab|LL039 Allatostatin A-type SKSPYSFGLamide Cb br,PO SKSPYSFGL >gi|040 |LiLab|LL040 Allatostatin A-type SNPYSFGLamide Csap PO SNPYSFGL >gi|041 |LiLab|LL041 Allatostatin A-type SPRLTYFGLamide Csap PO SPRLTYFGL >gi|042 |LiLab|LL042 Allatostatin A-type SSGQYAFGLamide Csap PO Cb PO SSGQYAFGL >gi|043 |LiLab|LL043 Allatostatin A-type SYAFGLamide Cb PO SYAFGL >gi|044 |LiLab|LL044 Allatostatin A-type TAPYAFGLamide Csap PO TAPYAFGL >gi|045 |LiLab|LL045 Allatostatin A-type TPHTYSFGLamide Csap TPHTYSFGL >gi|046 |LiLab|LL046 Allatostatin A-type TRPYSFGLamide Csap PO Cmaen Br/PO TRPYSFGL >gi|047 |LiLab|LL047 Allatostatin A-type VGPYAFGLamide VGPYAFGL >gi|048 |LiLab|LL048 Allatostatin A-type YAHSFGLamide Csap br YAHSFGL >gi|049 |LiLab|LL049 Allatostatin A-type AST-3 GGSLYSFGLamide GGSLYSFGL >gi|050 |LiLab|LL050 Allatostatin A-type Carcinustatin 1, Penaeustatin 13 YAFGLamide Cb.PO. YAFGL >gi|051 |LiLab|LL051 Allatostatin A-type Carcinustatin 10 APQPYAFGLamide Cmaen PO Cb br APQPYAFGL 389 >gi|052 |LiLab|LL052 Allatostatin A-type Carcinustatin 11 ATGQYAFGLamide Cmaen Br ATGQYAFGL >gi|053 |LiLab|LL053 Allatostatin A-type Carcinustatin 12 PDMYAFGLamide Cb.PO. PDMYAFGL >gi|054 |LiLab|LL054 Allatostatin A-type Carcinustatin 13 EYDDMYTEKRPKVYAFGLamide Cmaen PO EYDDMYTEKRPKVYAFGL >gi|055 |LiLab|LL055 Allatostatin A-type Carcinustatin 14, Penaeustatin 35 YSFGLamide Cp.PO/Cb.PO. YSFGL >gi|056 |LiLab|LL056 Allatostatin A-type Carcinustatin 15, Procastatin 27 AGPYSFGLamide Csap PO,br Cmaen Br/PO Cb PO AGPYSFGL >gi|057 |LiLab|LL057 Allatostatin A-type Carcinustatin 16 GGPYSYGLamide GGPYSYGL >gi|058 |LiLab|LL058 Allatostatin A-type Carcinustatin 17, Procastatin 10 SGQYSFGLamide SGQYSFGL >gi|059 |LiLab|LL059 Allatostatin A-type Carcinustatin 18 SDMYSFGLamide Cmaen PO Cb PO SDMYSFGL >gi|060 |LiLab|LL060 Allatostatin A-type Carcinustatin 19 APTDMYSFGLamide APTDMYSFGL >gi|061 |LiLab|LL061 Allatostatin A-type Carcinustatin 2 EAYAFGLamide Cmaen Br/PO EAYAFGL >gi|062 |LiLab|LL062 Allatostatin A-type Carcinustatin 20 GYEDEDEDRPFYALGLGKRPRTYSFGLamide Cmaen PO GYEDEDEDRPFYALGLGKRPRTYSFGL >gi|063 |LiLab|LL063 Allatostatin A-type Carcinustatin 3 EPYAFGLamide Cmaen Br/PO Cb br,PO EPYAFGL >gi|064 |LiLab|LL064 Allatostatin A-type Carcinustatin 4 DPYAFGLamide Cmaen Br Cb br,PO DPYAFGL >gi|065 |LiLab|LL065 Allatostatin A-type Carcinustatin 5 NPYAFGLamide Cmaen Br/PO NPYAFGL >gi|066 |LiLab|LL066 Allatostatin A-type Carcinustatin 6 SPYAFGLamide SPYAFGL >gi|067 |LiLab|LL067 Allatostatin A-type Carcinustatin 7 ASPYAFGLamide Cmaen Br/PO ASPYAFGL >gi|068 |LiLab|LL068 Allatostatin A-type Carcinustatin 8, Penaeustatin 6, Procastatin 17 AGPYAFGLamide Csap br Cmaen Br/PO AGPYAFGL >gi|069 |LiLab|LL069 Allatostatin A-type Carcinustatin 9 GGPYAFGLamide Cmaen Br GGPYAFGL >gi|070 |LiLab|LL070 Allatostatin A-type Lepidopteran peptide cydiastatin 1 SPHYNFGLamide SPHYNFGL 390 >gi|071 |LiLab|LL071 Allatostatin A-type Lepidopteran peptide cydiastatin 2, helicostatin 2 [11-18] LPVYNFGLamide LPVYNFGL >gi|072 |LiLab|LL072 Allatostatin A-type Lepidopteran peptide cydiastatin 2, helicostatin 2 AYSYVSEYKRLPVYNFGLamide AYSYVSEYKRLPVYNFGL >gi|073 |LiLab|LL073 Allatostatin A-type Lepidopteran peptide cydiastatin 3,helicostatin 3 SRPYSFGLamide Cb PO SRPYSFGL >gi|074 |LiLab|LL074 Allatostatin A-type Lepidopteran peptide cydiastatin 4, helicostatin 4 ARPYSFGLamide Csap PO Cmaen Br/PO Cb PO ARPYSFGL >gi|075 |LiLab|LL075 Allatostatin A-type Lepidopteran peptide cydiastatin 5 ARGYDFGLamide Csap PO ARGYDFGL >gi|076 |LiLab|LL076 Allatostatin A-type Lepidopteran peptide cydiastatin 6 LPLYNFGLamide LPLYNFGL >gi|077 |LiLab|LL077 Allatostatin A-type Lepidopteran peptide cydiastatin 7 KMYDFGLamide KMYDFGL >gi|078 |LiLab|LL078 Allatostatin A-type Lepidopteran peptide helicostatin 1 SPHYDFGLamide SPHYDFGL >gi|079 |LiLab|LL079 Allatostatin A-type Lepidopteran peptide helicostatin 5 ARAYDFGLamide Csap PO ARAYDFGL >gi|080 |LiLab|LL080 Allatostatin A-type Lepidopteran peptide helicostatin 6 LPMYNFGLamide Cb.PO. LPMYNFGL >gi|081 |LiLab|LL081 Allatostatin A-type Lepidopteran peptide helicostatin 7 ARSYNFGLamide ARSYNFGL >gi|082 |LiLab|LL082 Allatostatin A-type Lepidopteran peptide helicostatin 8 YSKFNFGLamide YSKFNFGL >gi|083 |LiLab|LL083 Allatostatin A-type Lepidopteran peptide helicostatin 9 ERDMHRFSFGLamide ERDMHRFSFGL >gi|084 |LiLab|LL084 Allatostatin A-type Lepidopteran peptide Lepidostatin I AKSYNFGLamide AKSYNFGL >gi|085 |LiLab|LL085 Allatostatin A-type Penaeustatin 1 ANEDEDAASLFAFGLamide ANEDEDAASLFAFGL >gi|086 |LiLab|LL086 Allatostatin A-type Penaeustatin 10 TPSYAFGLamide TPSYAFGL >gi|087 |LiLab|LL087 Allatostatin A-type Penaeustatin 11 pQRDYAFGLamide QRDYAFGL >gi|088 |LiLab|LL088 Allatostatin A-type Penaeustatin 12 SDYAFGLamide SDYAFGL >gi|089 |LiLab|LL089 Allatostatin A-type Penaeustatin 14 ANQYTFGLamide ANQYTFGL >gi|090 |LiLab|LL090 Allatostatin A-type Penaeustatin 15 ASQYTFGLamide 391 ASQYTFGL >gi|091 |LiLab|LL091 Allatostatin A-type Penaeustatin 16 SQYTFGLamide SQYTFGL >gi|092 |LiLab|LL092 Allatostatin A-type Penaeustatin 17 YTFGLamide YTFGL >gi|093 |LiLab|LL093 Allatostatin A-type Penaeustatin 18 SGHYNFGLamide Csap PO SGHYNFGL >gi|094 |LiLab|LL094 Allatostatin A-type Penaeustatin 19 GHYNFGLamide GHYNFGL >gi|095 |LiLab|LL095 Allatostatin A-type Penaeustatin 2 PDAEESNKRDRLYAFGLamide PDAEESNKRDRLYAFGL >gi|096 |LiLab|LL096 Allatostatin A-type Penaeustatin 20 AGPYEFGLamide AGPYEFGL >gi|097 |LiLab|LL097 Allatostatin A-type Penaeustatin 21 GGPYEFGLamide GGPYEFGL >gi|098 |LiLab|LL098 Allatostatin A-type Penaeustatin 22 AAPYEFGLamide Cmaen Br/PO AAPYEFGL >gi|099 |LiLab|LL099 Allatostatin A-type Penaeustatin 23 GPYEFGLamide GPYEFGL >gi|100 |LiLab|LL100 Allatostatin A-type Penaeustatin 24 SPYEFGLamide SPYEFGL >gi|101 |LiLab|LL101 Allatostatin A-type Penaeustatin 25 NPYEFGLamide NPYEFGL >gi|102 |LiLab|LL102 Allatostatin A-type Penaeustatin 26 NEVPDPETERNSYDFGLamide NEVPDPETERNSYDFGL >gi|103 |LiLab|LL103 Allatostatin A-type Penaeustatin 27 EVPDPETERNSYDFGLamide EVPDPETERNSYDFGL >gi|104 |LiLab|LL104 Allatostatin A-type Penaeustatin 28 PETERNSYDFGLamide PETERNSYDFGL >gi|105 |LiLab|LL105 Allatostatin A-type Penaeustatin 29 NSYDFGLamide NSYDFGL >gi|106 |LiLab|LL106 Allatostatin A-type Penaeustatin 3 DRLYAFGLamide DRLYAFGL >gi|107 |LiLab|LL107 Allatostatin A-type Penaeustatin 30 YDFGLamide YDFGL >gi|108 |LiLab|LL108 Allatostatin A-type Penaeustatin 31 AGHYSFGLamide AGHYSFGL >gi|109 |LiLab|LL109 Allatostatin A-type Penaeustatin 32 DRTYSFGLamide DRTYSFGL >gi|110 |LiLab|LL110 Allatostatin A-type Penaeustatin 33 PSAYSFGLamide PSAYSFGL >gi|111 |LiLab|LL111 Allatostatin A-type Penaeustatin 34 pQNMYSFGLamide QNMYSFGL >gi|112 |LiLab|LL112 Allatostatin A-type Penaeustatin 36 DARGALDLDQSPAYASDLGKRIGSAYSFGLamide DARGALDLDQSPAYASDLGKRIGSAYSFGL >gi|113 |LiLab|LL113 Allatostatin A-type Penaeustatin 37 TARGALDLDQSPAYASDLGKRIGSAYSFGLamide 392 TARGALDLDQSPAYASDLGKRIGSAYSFGL >gi|114 |LiLab|LL114 Allatostatin A-type SVAYGFGL >gi|115 |LiLab|LL115 Allatostatin A-type TVAYGFGL >gi|116 |LiLab|LL116 Allatostatin A-type TGGPYAFGL >gi|117 |LiLab|LL117 Allatostatin A-type GIYGFGL >gi|118 |LiLab|LL118 Allatostatin A-type SAGPYAFGL >gi|119 |LiLab|LL119 Allatostatin A-type SGHYAFGL >gi|120 |LiLab|LL120 Allatostatin A-type ANQYAFGL >gi|121 |LiLab|LL121 Allatostatin A-type AGQYAFGL >gi|122 |LiLab|LL122 Allatostatin A-type QNNYGFGL >gi|123 |LiLab|LL123 Allatostatin A-type PRNYAFGL >gi|124 |LiLab|LL124 Allatostatin TSDEEDDEDDQYYPYGLamide TSDEEDDEDDQYYPYGL >gi|125 |LiLab|LL125 Allatostatin A-type PRVYGFGL >gi|126 |LiLab|LL126 Allatostatin ADSYGLAFGNGGDALEMGLamide ADSYGLAFGNGGDALEMGL >gi|127 |LiLab|LL127 Allatostatin A-type SYDFGL >gi|128 |LiLab|LL128 Allatostatin A-type TAGPYAFGL >gi|129 |LiLab|LL129 Allatostatin A-type SGPYAFGL >gi|130 |LiLab|LL130 Allatostatin A-type TGPYAFGL >gi|131 |LiLab|LL131 Allatostatin A-type TPNYAFGL >gi|132 |LiLab|LL132 Allatostatin A-type ADPYAFGL >gi|133 |LiLab|LL133 Allatostatin A-type PNPYAFGL >gi|134 |LiLab|LL134 Allatostatin A-type DGMYSFGL >gi|135 |LiLab|LL135 Allatostatin A-type AGQYSFGL >gi|136 |LiLab|LL136 Allatostatin A-type SGPYSFGL >gi|137 |LiLab|LL137 Allatostatin A-type EDYDSSDQYSL >gi|138 |LiLab|LL138 Allatostatin A-type SGAYSFGL >gi|139 |LiLab|LL139 Allatostatin A-type Penaeustatin 38 SVAYGFGLamide Penaeustatin 39 TVAYGFGLamide Penaeustatin 4 TGGPYAFGLamide Penaeustatin 40 (X)GIYGFGLamide Penaeustatin 5 SAGPYAFGLamide Penaeustatin 7 SGHYAFGLamide Penaeustatin 8 ANQYAFGLamide Penaeustatin 9 AGQYAFGLamide Procastatin 1 QNNYGFGLamide Procastatin 11 PRNYAFGLamide A-type Procastatin 12 Procastatin 13 PRVYGFGLamide A-type Procastatin 14 Procastatin 15 SYDFGLamide Procastatin 16 TAGPYAFGLamide Procastatin 18 SGPYAFGLamide Procastatin 19 TGPYAFGLamide Procastatin 2 TPNYAFGLamide Procastatin 20 ADPYAFGLamide Procastatin 21 PNPYAFGLamide Procastatin 22 DGMYSFGLamide Procastatin 23 AGQYSFGLamide Procastatin 24 SGPYSFGLamide Procastatin 25 EDYDSSDQYSLamide Procastatin 26 SGAYSFGLamide Procastatin 3 QGMYSFGLamide 393 QGMYSFGL >gi|140 |LiLab|LL140 PDMYSFGL >gi|141 |LiLab|LL141 PDLYSFGL >gi|142 |LiLab|LL142 ADMYSFGL >gi|143 |LiLab|LL143 ADLYSFGL >gi|144 |LiLab|LL144 SGNYNFGL >gi|145 |LiLab|LL145 SRQYSFGL >gi|146 |LiLab|LL146 PO AGWNKFQGSW >gi|147 |LiLab|LL147 AGWSSMRGAW >gi|148 |LiLab|LL148 AGWSSTSRAW >gi|149 |LiLab|LL149 AWSNLGQAW >gi|150 |LiLab|LL150 GSNWSNLRGAW >gi|151 |LiLab|LL151 GVNWSNLRGAW >gi|152 |LiLab|LL152 LGNWNKFQGSW >gi|153 |LiLab|LL153 LGNWSNLRGAW >gi|154 |LiLab|LL154 LMFAPLAWPKGGARW >gi|155 |LiLab|LL155 LNNNWSKFQGSW >gi|156 |LiLab|LL156 LNWNKFQGSW >gi|157 |LiLab|LL157 MFAPLAWPKGGARW >gi|158 |LiLab|LL158 NDWSKFGQSW >gi|159 |LiLab|LL159 NNNWTKFQGSW >gi|160 |LiLab|LL160 NNWSGAFKGSW >gi|161 |LiLab|LL161 NPDWAHFRGSW >gi|162 |LiLab|LL162 SGDWSSLRGAW >gi|163 |LiLab|LL163 SKWNKFQGSW >gi|164 |LiLab|LL164 STDWSSLRSAW >gi|165 |LiLab|LL165 TGWNKFQGSW Allatostatin A-type Procastatin 4 PDMYSFGLamide Allatostatin A-type Procastatin 5 PDLYSFGLamide Allatostatin A-type Procastatin 6 ADMYSFGLamide Allatostatin A-type Procastatin 7 ADLYSFGLamide Allatostatin A-type Procastatin 8 SGNYNFGLamide Allatostatin A-type Procastatin 9 SRQYSFGLamide Allatostatin B-type AGWNKFQGSWamide Csap PO Cmaen Allatostatin B-type AGWSSMRGAWamide Csap PO Allatostatin B-type AGWSSTSRAWamide Csap br Allatostatin B-type AWSNLGQAWamide Cmaen PO Allatostatin B-type GSNWSNLRGAWamide Cmaen PO Allatostatin B-type GVNWSNLRGAWamide Cmaen VNC/PO Allatostatin B-type LGNWNKFQGSWamide Csap Allatostatin B-type LGNWSNLRGAWamide Cb PO Allatostatin B-type LMFAPLAWPKGGARWamide Csap Allatostatin B-type LNNNWSKFQGSWamide Csap Allatostatin B-type LNWNKFQGSWamide Csap Allatostatin B-type MFAPLAWPKGGARWamide Csap PO Allatostatin B-type NDWSKFGQSWamide Csap Allatostatin B-type NNNWTKFQGSWamide Csap br Allatostatin B-type NNWSGAFKGSWamide Csap Allatostatin B-type NPDWAHFRGSWamide Csap Allatostatin B-type SGDWSSLRGAWamide Csap PO,br Allatostatin B-type SKWNKFQGSWamide Hoa.brain Allatostatin B-type STDWSSLRSAWamide Csap PO,br Allatostatin B-type TGWNKFQGSWamide Csap br 394 >gi|166 |LiLab|LL166 Allatostatin B-type TNWNKFQGSWamide Hoa. Brain TNWNKFQGSW >gi|167 |LiLab|LL167 Allatostatin B-type TQNWTKFQGSWamide Csap TQNWTKFQGSW >gi|168 |LiLab|LL168 Allatostatin B-type VTWGKFQGSWamide Cmaen PO VTWGKFQGSW >gi|169 |LiLab|LL169 Allatostatin B-type CbAST-B1 VPNDWAHFRGSWamide Csap PO,br Cmaen Br/VNC/PO Cb br,PO,CoG VPNDWAHFRGSW >gi|170 |LiLab|LL170 Allatostatin B-type CbAST-B2 QWSSMRGAWamide Cmaen Br/PO QWSSMRGAW >gi|171 |LiLab|LL171 Allatostatin B-type CbAST-B3 SGKWSNLRGAWamide Csap PO Cmaen Br/PO SGKWSNLRGAW >gi|172 |LiLab|LL172 Allatostatin B-type CbAST-B4 NWNKFQGSWamide Csap PO Cb br,PO,STG NWNKFQGSW >gi|173 |LiLab|LL173 Allatostatin B-type CbAST-B5 TSWGKFQGSWamide Csap PO,br Cmaen PO Cb br,PO,STG TSWGKFQGSW >gi|174 |LiLab|LL174 Allatostatin B-type CbAST-B6 GNWNKFQGSWamide Csap PO,br Cmaen PO Cb br,PO,STG,CoG GNWNKFQGSW >gi|175 |LiLab|LL175 Allatostatin B-type CbAST-B7 NNWSKFQGSWamide Csap PO,br Cmaen PO Cb br,PO,STG,CoG NNWSKFQGSW >gi|176 |LiLab|LL176 Allatostatin B-type CbAST-B8 STNWSSLRSAWamide Csap PO,br Cmaen VNC/PO Cb PO STNWSSLRSAW >gi|177 |LiLab|LL177 Allatostatin B-type CbAST-B9 NNNWSKFQGSWamide Csap PO Cmaen Br/PO Cb br,PO,STG,CoG NNNWSKFQGSW >gi|178 |LiLab|LL178 Allatostatin B-type Lepidopteran peptide MIP IV AWSALHGAWamide AWSALHGAW >gi|179 |LiLab|LL179 Allatostatin Combos DPYAFGLGKRPADL Cb.PO. DPYAFGLGKRPADL >gi|180 |LiLab|LL180 Allatostatin Combos DPYAFGLGKRPADLYEFGLamide Cb.PO. DPYAFGLGKRPADLYEFGL >gi|181 |LiLab|LL181 Allatostatin Combos DPYAFGLGKRPDMYAFGLamide Cb.PO. DPYAFGLGKRPDMYAFGL >gi|182 |LiLab|LL182 Allatostatin Combos DPYAFGLGKRPDMYGFGLamide Cb.PO. DPYAFGLGKRPDMYGFGL >gi|183 |LiLab|LL183 Allatostatin Combos EPYAFGLGKRPATDL Cb.PO. EPYAFGLGKRPATDL >gi|184 |LiLab|LL184 Allatostatin Combos EPYAFGLGKRPATDLYAFGLamide Cb.PO. EPYAFGLGKRPATDLYAFGL >gi|185 |LiLab|LL185 Allatostatin Combos GSGQYAFGLGKKAGGAYSFGLamide Cb.PO. GSGQYAFGLGKKAGGAYSFGL >gi|186 |LiLab|LL186 Allatostatin C-type SYWKQCAFNAVSCFamide Cb cog, hoa cog, capr cog, Cmaen cog 395 SYWKQCAFNAVSCF >gi|187 |LiLab|LL187 Allatostatin C-type PISCF-AST pQIRYHQcYFNPIScF Cb PO, CoG, brain and Hoa. PO, CoG, brain QIRYHQCYFNPISCF >gi|188 |LiLab|LL188 Allatostatin C-type Lepidopteran peptide Manse-AS pEVRFRQCYFNPISCF QVRFRQCYFNPISCF >gi|189 |LiLab|LL189 Allatotropin Lepidopteran peptide Manse-AT GFKNVEMMTARGFamide Cp.SG GFKNVEMMTARGF >gi|190 |LiLab|LL190 bursicon bursicon alpha DECSLRPVIHILSYPGCTSKPIPSFACQGRCTSYVQVSGSKLWQTERSMCCQESGEREAAITLNCPKPRPGEP KEKKVLTRAPIDCMCRPCTDVEEGTVLAQKIANFIQDSMPDSVPFLK DECSLRPVIHILSYPGCTSKPIPSFACQGRCTSYVQVSGSKLWQTERSMCCQESGEREAAITLNCPKPRPGEP KEKKVLTRAPIDCMCRPCTDVEEGTVLAQKIANFIQDSMPDSVPFLK >gi|191 |LiLab|LL191 bursicon bursicon beta RSYGECETLPSTIHISKEEYDDTGRLVRVCEEDVAVNKCEGACVSKVQPSVNTPSGFLKDCRCCREVHLRARD ITLTHCYDGDGARLSGAKATQHVKLREPADCQCFKCGDSTR RSYGECETLPSTIHISKEEYDDTGRLVRVCEEDVAVNKCEGACVSKVQPSVNTPSGFLKDCRCCREVHLRARD ITLTHCYDGDGARLSGAKATQHVKLREPADCQCFKCGDSTR >gi|192 |LiLab|LL192 Calcitonin-like diuretic hormone GLDLGLGRGFSGSQAAKHLMGLAAANFANFAGGPamide GLDLGLGRGFSGSQAAKHLMGLAAANFANFAGGP >gi|193 |LiLab|LL193 CCAP CCAP PFCNAFTGCamide Csap SG,PO Cmaen Br/VNC/PO Cb br,PO PFCNAFTGC >gi|194 |LiLab|LL194 CCAP CCAP precursor related peptide DIGDLLEGKD DIGDLLEGKD >gi|195 |LiLab|LL195 CCAP Lepidopteran peptide CAP2b pELYAFPRVamide QLYAFPRV >gi|196 |LiLab|LL196 CHH (Crustacean hyperglycemic hormone) CanboCHH pQIYDTSCKGVYDRALFSDLEHVCDDCYNLYRSSYVASECRRNCYSNVVFRQCMEELLLLMEEFDKYARAVQI Vamide Cb QIYDTSCKGVYDRALFSDLEHVCDDCYNLYRSSYVASECRRNCYSNVVFRQCMEELLLLMEEFDKYARAVQIV >gi|197 |LiLab|LL197 CHH (Crustacean hyperglycemic hormone) CanpaCHH-II pEIYDTSCKGVYDRGLFSDLEHVCDDCYNLYRNSYVASACRSNCYSNVVFRQVMEELLLMEEFDKYARAVQIV amide Cancer pagarus QIYDTSCKGVYDRGLFSDLEHVCDDCYNLYRNSYVASACRSNCYSNVVFRQVMEELLLMEEFDKYARAVQIV >gi|198 |LiLab|LL198 CHH (Crustacean hyperglycemic hormone) CanpaMIH RVINDECPNLIGNRDLYKKVEWICEDCSNIFRKTGMASLCRRNCFFNEDFVWCVHATERSEELEDLEEWVGIL GAGRD RVINDECPNLIGNRDLYKKVEWICEDCSNIFRKTGMASLCRRNCFFNEDFVWCVHATERSEELEDLEEWVGIL GAGRD >gi|199 |LiLab|LL199 CHH (Crustacean hyperglycemic hormone) CanpaMOIH1 RRINNDCQNFIGNRAMYEKVDWICKDCANIFRKDGLLNNCRSNCFYNTEFLWCIDATENTRNKEQLEQWAAIL GAGWN Cancer pagarus RRINNDCQNFIGNRAMYEKVDWICKDCANIFRKDGLLNNCRSNCFYNTEFLWCIDATENTRNKEQLEQWAAIL GAGWN >gi|200 |LiLab|LL200 CHH (Crustacean hyperglycemic hormone) CanpaMOIH2 RRINNDCQNFIGNRAMYEKVDWICKDCANIFRQDGLLNNCRSNCFYNTEFLWCIDATENTRNKEQLEQWAAIL GAGWN RRINNDCQNFIGNRAMYEKVDWICKDCANIFRQDGLLNNCRSNCFYNTEFLWCIDATENTRNKEQLEQWAAIL GAGWN 396 >gi|201 |LiLab|LL201 CHH (Crustacean hyperglycemic hormone) CanproCHH-II QIYDSSCKGVYDRGLFSDLEHVCDDCYNLYRNSYVASACRSNCYSNVVFRQVMEELLLMEEFDKYARAVQIVa mide Cancer productus QIYDSSCKGVYDRGLFSDLEHVCDDCYNLYRNSYVASACRSNCYSNVVFRQVMEELLLMEEFDKYARAVQIV >gi|202 |LiLab|LL202 CHH (Crustacean hyperglycemic hormone) Capa-CHH II pEIYDTSCKGVYDRGLFSDLEHVCDDCYNLYRNSYVASACRSNCYSNVVFRQCMEELLLMDEFDKYARAVQIV Cancer pagurus QIYDTSCKGVYDRGLFSDLEHVCDDCYNLYRNSYVASACRSNCYSNVVFRQCMEELLLMDEFDKYARAVQIV >gi|203 |LiLab|LL203 CHH (Crustacean hyperglycemic hormone) CapaMIH RVINDDCPNLIGNRDLYKKVEWICEDCSNIFRNTGMATLCRKNCFFNEDFLWCVYATERTEEMSQLRQWVGIL GAGRE RVINDDCPNLIGNRDLYKKVEWICEDCSNIFRNTGMATLCRKNCFFNEDFLWCVYATERTEEMSQLRQWVGIL GAGRE >gi|204 |LiLab|LL204 CHH (Crustacean hyperglycemic hormone) Capr-CHH I QIYDTSCKGVYDRGLFSDLEHVCDDCYNLYRNSYVASACRSNCYSNVVFRQCMEELLLMEEFDKYARAVQIV QIYDTSCKGVYDRGLFSDLEHVCDDCYNLYRNSYVASACRSNCYSNVVFRQCMEELLLMEEFDKYARAVQIV >gi|205 |LiLab|LL205 CHH (Crustacean hyperglycemic hormone) Capr-CHH II QIYDSSCKGVYDRGLFSDLEHVCDDCYNLYRNSYVASACRSNCYSNVVFRQCMEELLLMEEFDKYARAVQIV QIYDSSCKGVYDRGLFSDLEHVCDDCYNLYRNSYVASACRSNCYSNVVFRQCMEELLLMEEFDKYARAVQIV >gi|206 |LiLab|LL206 CHH (Crustacean hyperglycemic hormone) CarmaCHH pQIYDTSCKGVYDRALFNDLEHVCDDCYNLYRTSYVASACRSNCYSNLVFRQCMDDLLMMDEFDQYARKVQMV amide Cmaen QIYDTSCKGVYDRALFNDLEHVCDDCYNLYRTSYVASACRSNCYSNLVFRQCMDDLLMMDEFDQYARKVQMV >gi|207 |LiLab|LL207 CHH (Crustacean hyperglycemic hormone) CarmaMIH RVINDDCPNLIGNRDLYKRVEWICEDCSNIFRNTGMATLCRKNCFFNEDFLWCVYATERTEEMSQLRQWVGIL GAGRE RVINDDCPNLIGNRDLYKRVEWICEDCSNIFRNTGMATLCRKNCFFNEDFLWCVYATERTEEMSQLRQWVGIL GAGRE >gi|208 |LiLab|LL208 CHH (Crustacean hyperglycemic hormone) CarmaPOCHH pQIYDTSCKGVYDRALFNDLEHVCDDCYNLYRTSYVASACRNNCFENEVFDVCVYQLYFPNHEEYLRSRDGLK G Cmaen PO QIYDTSCKGVYDRALFNDLEHVCDDCYNLYRTSYVASACRNNCFENEVFDVCVYQLYFPNHEEYLRSRDGLKG >gi|209 |LiLab|LL209 CHH (Crustacean hyperglycemic hormone) DappuITPL SFFDIQCKGNYDKSIFARLDRICEDCYNLFREPQLHSLCRSDFKSPYFKGCLQALLLIDEEEKFNQMVEILam ide Daphnia pulex SFFDIQCKGNYDKSIFARLDRICEDCYNLFREPQLHSLCRSDFKSPYFKGCLQALLLIDEEEKFNQMVEIL >gi|210 |LiLab|LL210 CHH (Crustacean hyperglycemic hormone) DappuITPL SFFEDINCKGLYDKSIFARLDRICQDCYSLYREPELHTLCRKNCFTTNYFKGCLDALLINDEKDIQRVMKDIS IIHQIPI Daphnia pulex SFFEDINCKGLYDKSIFARLDRICQDCYSLYREPELHTLCRKNCFTTNYFKGCLDALLINDEKDIQRVMKDIS IIHQIPI >gi|211 |LiLab|LL211 CHH (Crustacean hyperglycemic hormone) Hoa-CHH-A (pCHH-A[pQ61-V132amide]) pEVFDQACKGVYDRNLFKKLDRVCEDCYNLYRKPFVATTCRENCYSNWVFRQCLDDLLLSDVIDEYVSNVQMV amide QVFDQACKGVYDRNLFKKLDRVCEDCYNLYRKPFVATTCRENCYSNWVFRQCLDDLLLSDVIDEYVSNVQMV >gi|212 |LiLab|LL212 CHH (Crustacean hyperglycemic hormone) Hoa-CHH-B (pCHH-B[pQ61-V132amide]) pEVFDQACKGVYDRNLFKKLNRVCEDCYNLYRKPFIVTTCRENCYSNRVFRQCLDDLLMIDVIDEYVSNVQMV amide QVFDQACKGVYDRNLFKKLNRVCEDCYNLYRKPFIVTTCRENCYSNRVFRQCLDDLLMIDVIDEYVSNVQMV >gi|213 |LiLab|LL213 CHH (Crustacean hyperglycemic hormone) Hoa-MIH (pMIH[pQ61-M131]) pEVFDQACKGVYDRNLFKKLDRVCEDCYNLYRKPFVATTCRENCYSNWVFRQCLDDLLLSNVIDEYVSNVQM 397 QVFDQACKGVYDRNLFKKLDRVCEDCYNLYRKPFVATTCRENCYSNWVFRQCLDDLLLSNVIDEYVSNVQM >gi|214 |LiLab|LL214 CHH (Crustacean hyperglycemic hormone) HoaVIH ASAWFTNDECPGVMGNRDLYEKVAWVCNDCANIFRNNDVGVMCKKDCFHTMWFLWCVYATERHGEIDQFRKWV SILR ASAWFTNDECPGVMGNRDLYEKVAWVCNDCANIFRNNDVGVMCKKDCFHTMWFLWCVYATERHGEIDQFRKWV SILR >gi|215 |LiLab|LL215 CHH (Crustacean hyperglycemic hormone) HomamVIH ASAWFTNDECPGVMGNRDLYEKVAWVCNDCANIFRNNDVGVMCKKDCFHTMDFLWCVYATERHGEIDQFRKWV SILRAamide Hoa ASAWFTNDECPGVMGNRDLYEKVAWVCNDCANIFRNNDVGVMCKKDCFHTMDFLWCVYATERHGEIDQFRKWV SILRA >gi|216 |LiLab|LL216 CHH (Crustacean hyperglycemic hormone) Mee-MIH SYIENTCRGVMGNRDIYKKVVRVCEDCTNIFRLPGLDGMCRDRCFNNEWFLVCLKAANRDDELDKFKVWISIL NPGL Metepenaeus ensis SYIENTCRGVMGNRDIYKKVVRVCEDCTNIFRLPGLDGMCRDRCFNNEWFLVCLKAANRDDELDKFKVWISIL NPGL >gi|217 |LiLab|LL217 CHH (Crustacean hyperglycemic hormone) MetenCHHA SLFDPSCSGVFDRELLGRLNRVCDDCYNVFRDPKVAMECKSNCFLNPAFIQCLEYLLPEDLHEEYQSHQVVam ide Metapenaeus ensis SLFDPSCSGVFDRELLGRLNRVCDDCYNVFRDPKVAMECKSNCFLNPAFIQCLEYLLPEDLHEEYQSHQVV >gi|218 |LiLab|LL218 CHH (Crustacean hyperglycemic hormone) Orl-CHH pEVFDQACKGIYDRAIFKKLDRVCEDCYNLYRKPYVATTCRQNCYANSVFRQCLDDLLLIDVLDEYISGVQTV amide Orconectes limosus QVFDQACKGIYDRAIFKKLDRVCEDCYNLYRKPYVATTCRQNCYANSVFRQCLDDLLLIDVLDEYISGVQTV >gi|219 |LiLab|LL219 CHH (Crustacean hyperglycemic hormone) Pej-MIH SFIDNRCRGVMGNRDIYKKVVRVCEDCTNIFRLGLDGMCRNRCFYNEWFLICLKAANREDEIEKFRVWISILN AGQ M. japonicus SFIDNRCRGVMGNRDIYKKVVRVCEDCTNIFRLGLDGMCRNRCFYNEWFLICLKAANREDEIEKFRVWISILN AGQ >gi|220 |LiLab|LL220 CHH (Crustacean hyperglycemic hormone) Prc-MIH RYVFEECPGVMGNRAVHGKVTRVCEDCYNVFRDTDVLAGCRKGCFSSEMFKLCLLAMERVEEFPDFKRWIGLL NAamide Procambarus clarkii RYVFEECPGVMGNRAVHGKVTRVCEDCYNVFRDTDVLAGCRKGCFSSEMFKLCLLAMERVEEFPDFKRWIGLL NA >gi|221 |LiLab|LL221 Corazonin Corazonin pQTFQYSRGWTNamide Cmaen Br Cb br,CoG QTFQYSRGWTN >gi|222 |LiLab|LL222 CPRP (CHH precursor-related peptide) (GLN-4) RSTQGYGRMDRILAALKTSPMEPSAALAVEHGTTHPLE Cmaen SG/PO RSTQGYGRMDRILAALKTSPMEPSAALAVEHGTTHPLE >gi|223 |LiLab|LL223 CPRP (CHH precursor-related peptide) (PRO-4) RSTPGYGRMDRILAALKTSPMEPSAALAVEHGTTHPLE Cmaen SG/PO RSTPGYGRMDRILAALKTSPMEPSAALAVEHGTTHPLE >gi|224 |LiLab|LL224 CPRP (CHH precursor-related peptide) ASLKSPTVTPLR Csap SG ASLKSPTVTPLR >gi|225 |LiLab|LL225 CPRP (CHH precursor-related peptide) Bthe CPRP RSAEGFGRMERLLASIRGGADSMGHLGELTGAGEGAGHPLE RSAEGFGRMERLLASIRGGADSMGHLGELTGAGEGAGHPLE >gi|226 |LiLab|LL226 CPRP (CHH precursor-related peptide) Cama CPRP I RSTQGYGRMDRILAALKTSPMEPSAALAVENGTTHPLE RSTQGYGRMDRILAALKTSPMEPSAALAVENGTTHPLE >gi|227 |LiLab|LL227 CPRP (CHH precursor-related peptide) Cama CPRP II RSTQGYGRMDRILAALKTSPMEPSAALAVQHGTTHPLE 398 RSTQGYGRMDRILAALKTSPMEPSAALAVQHGTTHPLE >gi|228 |LiLab|LL228 CPRP (CHH precursor-related peptide) Capa CPRP RSAQGMGKMERLLASYRGALEPSTPLGDLSGSLGHPVE RSAQGMGKMERLLASYRGALEPSTPLGDLSGSLGHPVE >gi|229 |LiLab|LL229 CPRP (CHH precursor-related peptide) Capr CPRP I RSAQGMGKMEHLLASYRGALESNTPTGDLPGGLVHPVE Cp.SG RSAQGMGKMEHLLASYRGALESNTPTGDLPGGLVHPVE >gi|230 |LiLab|LL230 CPRP (CHH precursor-related peptide) Capr CPRP II RSAQGMGKMERLLASYRGAVEPNTPLGDLPGGLVHPVE Cp.SG RSAQGMGKMERLLASYRGAVEPNTPLGDLPGGLVHPVE >gi|231 |LiLab|LL231 CPRP (CHH precursor-related peptide) Capr CPRP III RSAQGMGKMEHLLASYRGALESNTPLGDLPGGLVHPVE Cp.SG RSAQGMGKMEHLLASYRGALESNTPLGDLPGGLVHPVE >gi|232 |LiLab|LL232 CPRP (CHH precursor-related peptide) Capr CPRP IV RSAQGMGKMERLLASYRAAVEPNTPLGDLPGGLVHPVE Cp.SG RSAQGMGKMERLLASYRAAVEPNTPLGDLPGGLVHPVE >gi|233 |LiLab|LL233 CPRP (CHH precursor-related peptide) Casap CPRP RSAEGLGRMGRLLASLKSDTVTPLRGFEGETGHPLE Csap SG RSAEGLGRMGRLLASLKSDTVTPLRGFEGETGHPLE >gi|234 |LiLab|LL234 CPRP (CHH precursor-related peptide) Cb CPRP I RSAQGLGKMERLLASYRGALEPNTPLGDLSGSVGHPVE Cb.SG RSAQGLGKMERLLASYRGALEPNTPLGDLSGSVGHPVE >gi|235 |LiLab|LL235 CPRP (CHH precursor-related peptide) Cb CPRP II RSAQGLGKMERLLASYRGALEPNTPLGDLSGSLGHPVE Cb.SG RSAQGLGKMERLLASYRGALEPNTPLGDLSGSLGHPVE >gi|236 |LiLab|LL236 CPRP (CHH precursor-related peptide) Cb CPRP III RSAQGLGKMEHLLASYRGALEPNTPLGDLSGSLGHPVE Cb.SG RSAQGLGKMEHLLASYRGALEPNTPLGDLSGSLGHPVE >gi|237 |LiLab|LL237 CPRP (CHH precursor-related peptide) Cb CPRP IV RSAQGLGKMERLLVSYRGAVEPNTPLGDLSGSLGHPVE Cb.SG RSAQGLGKMERLLVSYRGAVEPNTPLGDLSGSLGHPVE >gi|238 |LiLab|LL238 CPRP (CHH precursor-related peptide) DLKSDTVTPLR Csap SG DLKSDTVTPLR >gi|239 |LiLab|LL239 CPRP (CHH precursor-related peptide) GFLSQDVHS GFLSQDVHS >gi|240 |LiLab|LL240 CPRP (CHH precursor-related peptide) Hoa CPRP A (I16) RSVEGASRMEKLLSSISPSSTPLGFLSQDHSVN Hoa.SG RSVEGASRMEKLLSSISPSSTPLGFLSQDHSVN >gi|241 |LiLab|LL241 CPRP (CHH precursor-related peptide) Hoa CPRP C RSVEGVSRMEKLLSSISPSSMPLGFLSQDHSVN Hoa.SG RSVEGVSRMEKLLSSISPSSMPLGFLSQDHSVN >gi|242 |LiLab|LL242 CPRP (CHH precursor-related peptide) Hoa CPRP-A (SN16) RSVEGASRMEKLLSSSNSPSSTPLGFLSQDHSVN Hoa.SG RSVEGASRMEKLLSSSNSPSSTPLGFLSQDHSVN >gi|243 |LiLab|LL243 CPRP (CHH precursor-related peptide) Hoa CPRP-B RSVEGVSRMEKLLSSISPSSTPLGFLSQDHSVN Hoa.SG RSVEGVSRMEKLLSSISPSSTPLGFLSQDHSVN >gi|244 |LiLab|LL244 CPRP (CHH precursor-related peptide) Mlan CPRP WSLDGLARIEKLLSTSSSASAASPTRGQALNL WSLDGLARIEKLLSTSSSASAASPTRGQALNL >gi|245 |LiLab|LL245 CPRP (CHH precursor-related peptide) Mros CPRP WSVDGLARIEKLLSTSSSASAASPTRGQALNL WSVDGLARIEKLLSTSSSASAASPTRGQALNL 399 >gi|246 |LiLab|LL246 CPRP (CHH precursor-related peptide) Nnor CPRP A RSVEGASRMEKLLSSSNSPSSTPLGFLSQEHSVN RSVEGASRMEKLLSSSNSPSSTPLGFLSQEHSVN >gi|247 |LiLab|LL247 CPRP (CHH precursor-related peptide) Nnor CPRP B RSVEGASRMEKLLSSISPSSTPLGFLSQEHSVN RSVEGASRMEKLLSSISPSSTPLGFLSQEHSVN >gi|248 |LiLab|LL248 CPRP (CHH precursor-related peptide) Olim CPRP A RSVEGSSRMERLLSSGSSSSEPLSFLSQDHSVS RSVEGSSRMERLLSSGSSSSEPLSFLSQDHSVS >gi|249 |LiLab|LL249 CPRP (CHH precursor-related peptide) Olim CPRP A* RSVEGSSRMERLLSSGSSSSEPLSFLSQDQSVS RSVEGSSRMERLLSSGSSSSEPLSFLSQDQSVS >gi|250 |LiLab|LL250 CPRP (CHH precursor-related peptide) Olim CPRP B, Pcla CPRP RSVEGSSRMERLLSSGSSSSEPLSFLSQDQSVN RSVEGSSRMERLLSSGSSSSEPLSFLSQDQSVN >gi|251 |LiLab|LL251 CPRP (CHH precursor-related peptide) PSAALAVEHGTTHPLE Cmaen SG PSAALAVEHGTTHPLE >gi|252 |LiLab|LL252 CPRP (CHH precursor-related peptide) RASQGLGKMERLLASRGALEPN Cb SG RASQGLGKMERLLASRGALEPN >gi|253 |LiLab|LL253 CPRP (CHH precursor-related peptide) RGFEGETGHPN Csap SG RGFEGETGHPN >gi|254 |LiLab|LL254 CPRP (CHH precursor-related peptide) RSAEGLGRMamide Csap SG RSAEGLGRM >gi|255 |LiLab|LL255 CPRP (CHH precursor-related peptide) RSAEGLGRMG Csap SG RSAEGLGRMG >gi|256 |LiLab|LL256 CPRP (CHH precursor-related peptide) RSAEGLGRMGRL Csap SG RSAEGLGRMGRL >gi|257 |LiLab|LL257 CPRP (CHH precursor-related peptide) RSAEGLGRMGRLL Csap SG RSAEGLGRMGRLL >gi|258 |LiLab|LL258 CPRP (CHH precursor-related peptide) RSAEGLGRVGRLL Csap SG RSAEGLGRVGRLL >gi|259 |LiLab|LL259 CPRP (CHH precursor-related peptide) RSAQGLGKM(O)ERL Cb PO,CoG RSAQGLGKMERL >gi|260 |LiLab|LL260 CPRP (CHH precursor-related peptide) RSAQGLGKMEHLLASY Cb SG RSAQGLGKMEHLLASY >gi|261 |LiLab|LL261 CPRP (CHH precursor-related peptide) RSAQGLGKYLRLLASY Cb SG RSAQGLGKYLRLLASY >gi|262 |LiLab|LL262 CPRP (CHH precursor-related peptide) RSVEGASRMEKLLT RSVEGASRMEKLLT >gi|263 |LiLab|LL263 CPRP (CHH precursor-related peptide) RSVEGVSRMEKLLT RSVEGVSRMEKLLT >gi|264 |LiLab|LL264 CPRP (CHH precursor-related peptide) SLKSDTVTPLLG Csap SG 400 SLKSDTVTPLLG >gi|265 |LiLab|LL265 CPRP (CHH precursor-related peptide) Sserr CPRP RSAEGFGRMGRLLASLKADSLGPVQDFGVEGAAHPVE RSAEGFGRMGRLLASLKADSLGPVQDFGVEGAAHPVE >gi|266 |LiLab|LL266 CPRP (CHH precursor-related peptide) truncated (PRO4) 1-13 RSTPGYGRMDRIL Cmaen SG RSTPGYGRMDRIL >gi|267 |LiLab|LL267 CPRP (CHH precursor-related peptide) truncated (PRO4) 1-15 RSTPGYGRMDRILAA Cmaen SG RSTPGYGRMDRILAA >gi|268 |LiLab|LL268 CPRP (CHH precursor-related peptide) truncated (PRO4) 18-38 TSPMEPSAALAVEHGTTHPLE Cmaen SG TSPMEPSAALAVEHGTTHPLE >gi|269 |LiLab|LL269 CPRP (CHH precursor-related peptide) truncated (PRO4) 19-38 SPMEPSAALAVEHGTTHPLE Cmaen SG SPMEPSAALAVEHGTTHPLE >gi|270 |LiLab|LL270 CPRP (CHH precursor-related peptide) truncated Cama CPRP I 1-13 RSTQGYGRMDPIL Cmaen SG RSTQGYGRMDPIL >gi|271 |LiLab|LL271 CPRP (CHH precursor-related peptide) truncated Cama CPRP I 1-15 RSTQGYGRMDRILAA Cmaen SG RSTQGYGRMDRILAA >gi|272 |LiLab|LL272 CPRP (CHH precursor-related peptide) truncated Capa CPRP 1-12, Capr CPRP II 1-12 RSAQGMGKMERL Cp.SG & PO RSAQGMGKMERL >gi|273 |LiLab|LL273 CPRP (CHH precursor-related peptide) truncated Capa CPRP 24-38, CbCPRP II,III&IV 24-38 TPLGDLSGSLGHPVE Cb SG TPLGDLSGSLGHPVE >gi|274 |LiLab|LL274 CPRP (CHH precursor-related peptide) truncated Capr CPRP I 1-12 RSAQGMGKMEHL Cp.SG & PO RSAQGMGKMEHL >gi|275 |LiLab|LL275 CPRP (CHH precursor-related peptide) truncated Capr CPRP I 1-13 RSAQGMGKMEHLL Cp.SG & PO RSAQGMGKMEHLL >gi|276 |LiLab|LL276 CPRP (CHH precursor-related peptide) truncated Capr CPRP I 1-14 RSAQGMGKMEHLLA Cp.SG & PO RSAQGMGKMEHLLA >gi|277 |LiLab|LL277 CPRP (CHH precursor-related peptide) Truncated Capr CPRP I 30-38 PGGLVHPVE Cp.SG & PO PGGLVHPVE >gi|278 |LiLab|LL278 CPRP (CHH precursor-related peptide) Truncated Capr CPRP II [13-38] LASYRGAVEPNTPLGDLPGGLVHPVE Cp.SG & PO LASYRGAVEPNTPLGDLPGGLVHPVE >gi|279 |LiLab|LL279 CPRP (CHH precursor-related peptide) truncated Casap CPRP 13-25 LASLKSDTVTPLR Csap SG LASLKSDTVTPLR >gi|280 |LiLab|LL280 CPRP (CHH precursor-related peptide) truncated Casap CPRP 14-25 ASLKSDTVTPLR Csap SG ASLKSDTVTPLR >gi|281 |LiLab|LL281 CPRP (CHH precursor-related peptide) truncated Casap CPRP 15-25 SLKSDTVTPLR Csap SG SLKSDTVTPLR >gi|282 |LiLab|LL282 CPRP (CHH precursor-related peptide) truncated Casap CPRP 18-28 SDTVTPLRGFE Csap SG 401 SDTVTPLRGFE >gi|283 |LiLab|LL283 CPRP (CHH precursor-related peptide) truncated Casap CPRP 22-36 TPLRGFEGETGHPLE Csap SG TPLRGFEGETGHPLE >gi|284 |LiLab|LL284 CPRP (CHH precursor-related peptide) truncated Cb CPRP I&II 1-14 RSAQGLGKMERLLA Cb SG RSAQGLGKMERLLA >gi|285 |LiLab|LL285 CPRP (CHH precursor-related peptide) truncated Cb CPRP I&II 1-15 RSAQGLGKMERLLAS Cb SG RSAQGLGKMERLLAS >gi|286 |LiLab|LL286 CPRP (CHH precursor-related peptide) truncated Cb CPRP I&II 1-16 RSAQGLGKMERLLASY RSAQGLGKMERLLASY >gi|287 |LiLab|LL287 CPRP (CHH precursor-related peptide) truncated Cb CPRP I,II&IV 1-12 RSAQGLGKMER Cb SG RSAQGLGKMER >gi|288 |LiLab|LL288 CPRP (CHH precursor-related peptide) truncated Cb CPRP I,II&IV 1-13 RSAQGLGKMERLL Cb SG RSAQGLGKMERLL >gi|289 |LiLab|LL289 CPRP (CHH precursor-related peptide) truncated Cb CPRP II 18-38 GALEPNTPLGDLSGSLGHPVE Cb SG GALEPNTPLGDLSGSLGHPVE >gi|290 |LiLab|LL290 CPRP (CHH precursor-related peptide) truncated Cb CPRP II 2-38 SAQGLGKMERLLASYRGALEPNTPLGDLSGSLGHPVE SAQGLGKMERLLASYRGALEPNTPLGDLSGSLGHPVE >gi|291 |LiLab|LL291 CPRP (CHH precursor-related peptide) truncated Cb CPRP II 3-39 AQGLGKMERLLASYRGALEPNTPLGDLSGSLGHPVE AQGLGKMERLLASYRGALEPNTPLGDLSGSLGHPVE >gi|292 |LiLab|LL292 CPRP (CHH precursor-related peptide) truncated Cb CPRP II&III 12-38 LLASYRGALEPNTPLGDLSGSLGHPVE LLASYRGALEPNTPLGDLSGSLGHPVE >gi|293 |LiLab|LL293 CPRP (CHH precursor-related peptide) truncated Cb CPRP II&III 13-38 LASYRGALEPNTPLGDLSGSLGHPVE LASYRGALEPNTPLGDLSGSLGHPVE >gi|294 |LiLab|LL294 CPRP (CHH precursor-related peptide) truncated Cb CPRP II&III 16-38 RGALEPNTPLGDLSGSLGHPVE RGALEPNTPLGDLSGSLGHPVE >gi|295 |LiLab|LL295 CPRP (CHH precursor-related peptide) truncated Cb CPRP II&III 17-38 YRGALEPNTPLGDLSGSLGHPVE YRGALEPNTPLGDLSGSLGHPVE >gi|296 |LiLab|LL296 CPRP (CHH precursor-related peptide) truncated Cb CPRP II&III 19-38 ALEPNTPLGDLSGSLGHPVE ALEPNTPLGDLSGSLGHPVE >gi|297 |LiLab|LL297 CPRP (CHH precursor-related peptide) truncated Cb CPRP II,III&IV 23-38 NTPLGDLSGSLGHPVE NTPLGDLSGSLGHPVE >gi|298 |LiLab|LL298 CPRP (CHH precursor-related peptide) truncated Cb CPRP II,III&IV15-38 PLGDLSGSLGHPVE PLGDLSGSLGHPVE >gi|299 |LiLab|LL299 CPRP (CHH precursor-related peptide) truncated Cb CPRP IV 1-16 RSAQGLGKMERLLVSY RSAQGLGKMERLLVSY >gi|300 |LiLab|LL300 CPRP (CHH precursor-related peptide) truncated Hoa CPRP-A (SN-16) 11-23 KLLSSSNSPSSTP 402 KLLSSSNSPSSTP >gi|301 |LiLab|LL301 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 11-24 KLLSSSNSPSSTPL KLLSSSNSPSSTPL >gi|302 |LiLab|LL302 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 11-25 KLLSSSNSPSSTPLG KLLSSSNSPSSTPLG >gi|303 |LiLab|LL303 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 11-26 KLLSSSNSPSSTPLGF KLLSSSNSPSSTPLGF >gi|304 |LiLab|LL304 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 11-27 KLLSSSNSPSSTPLGFL KLLSSSNSPSSTPLGFL >gi|305 |LiLab|LL305 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 11-32 KLLSSSNSPSSTPLGFLSQDHS KLLSSSNSPSSTPLGFLSQDHS >gi|306 |LiLab|LL306 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 11-33 KLLSSSNSPSSTPLGFLSQDHSV KLLSSSNSPSSTPLGFLSQDHSV >gi|307 |LiLab|LL307 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 11-34 KLLSSSNSPSSTPLGFLSQDHSVN KLLSSSNSPSSTPLGFLSQDHSVN >gi|308 |LiLab|LL308 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 1-20 RSVEGASRMEKLLSSSNSPS RSVEGASRMEKLLSSSNSPS >gi|309 |LiLab|LL309 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 12-24 LLSSSNSPSSTPL LLSSSNSPSSTPL >gi|310 |LiLab|LL310 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 12-33 LLSSSNSPSSTPLGFLSQDHSV LLSSSNSPSSTPLGFLSQDHSV >gi|311 |LiLab|LL311 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 12-34 LLSSSNSPSSTPLGFLSQDHSVN LLSSSNSPSSTPLGFLSQDHSVN >gi|312 |LiLab|LL312 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 1-25 RSVEGASRMEKLLSSSNSPSSTPLG RSVEGASRMEKLLSSSNSPSSTPLG >gi|313 |LiLab|LL313 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 1-26 RSVEGASRMEKLLSSSNSPSSTPLGF RSVEGASRMEKLLSSSNSPSSTPLGF >gi|314 |LiLab|LL314 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 1-27 RSVEGASRMEKLLSSSNSPSSTPLGFL RSVEGASRMEKLLSSSNSPSSTPLGFL >gi|315 |LiLab|LL315 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 1-29 RSVEGASRMEKLLSSSNSPSSTPLGFLSQ RSVEGASRMEKLLSSSNSPSSTPLGFLSQ >gi|316 |LiLab|LL316 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 1-32 RSVEGASRMEKLLSSSNSPSSTPLGFLSQDHS RSVEGASRMEKLLSSSNSPSSTPLGFLSQDHS >gi|317 |LiLab|LL317 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 13-25 GFLSQDHSVN GFLSQDHSVN >gi|318 |LiLab|LL318 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 13-25 LSSSNSPSSTPLG truncated Hoa truncated Hoa truncated Hoa truncated Hoa truncated Hoa truncated Hoa truncated Hoa Truncated Hoa truncated Hoa truncated Hoa truncated Hoa Truncated Hoa Truncated Hoa Truncated Hoa Truncated Hoa Truncated Hoa truncated Hoa truncated Hoa 403 LSSSNSPSSTPLG >gi|319 |LiLab|LL319 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 1-33 RSVEGASRMEKLLSSSNSPSSTPLGFLSQDHSV RSVEGASRMEKLLSSSNSPSSTPLGFLSQDHSV >gi|320 |LiLab|LL320 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 13-32 LSSSNSPSSTPLGFLSQDHS LSSSNSPSSTPLGFLSQDHS >gi|321 |LiLab|LL321 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 13-33 LSSSNSPSSTPLGFLSQDHSV LSSSNSPSSTPLGFLSQDHSV >gi|322 |LiLab|LL322 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 13-34 LSSSNSPSSTPLGFLSQDHSVN LSSSNSPSSTPLGFLSQDHSVN >gi|323 |LiLab|LL323 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 14-33 SSSNSPSSTPLGFLSQDHSV SSSNSPSSTPLGFLSQDHSV >gi|324 |LiLab|LL324 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 14-34 SSSNSPSSTPLGFLSQDHSVN SSSNSPSSTPLGFLSQDHSVN >gi|325 |LiLab|LL325 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 15-32 SSNSPSSTPLGFLSQDHS Hoa.SG SSNSPSSTPLGFLSQDHS >gi|326 |LiLab|LL326 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 15-33 SSNSPSSTPLGFLSQDHSV SSNSPSSTPLGFLSQDHSV >gi|327 |LiLab|LL327 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 15-34 SSNSPSSTPLGFLSQDHSVN SSNSPSSTPLGFLSQDHSVN >gi|328 |LiLab|LL328 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 16-33 SNSPSSTPLGFLSQDHSV SNSPSSTPLGFLSQDHSV >gi|329 |LiLab|LL329 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 17-32 NSPSSTPLGFLSQDHS NSPSSTPLGFLSQDHS >gi|330 |LiLab|LL330 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 17-34 NSPSSTPLGFLSQDHSVN NSPSSTPLGFLSQDHSVN >gi|331 |LiLab|LL331 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 19-34, B 18-33 PSSTPLGFLSQDHSVN PSSTPLGFLSQDHSVN >gi|332 |LiLab|LL332 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 22-33, B 21-32 TPLGFLSQDHSV TPLGFLSQDHSV >gi|333 |LiLab|LL333 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 22-34, B 21-33 TPLGFLSQDHSVN TPLGFLSQDHSVN >gi|334 |LiLab|LL334 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 23-32, B&C 22-31 PLGFLSQDHS PLGFLSQDHS >gi|335 |LiLab|LL335 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 23-33, B&C 22-32 PLGFLSQDHSV PLGFLSQDHSV >gi|336 |LiLab|LL336 CPRP (CHH precursor-related peptide) CPRP-A (SN-16) 23-34, B&C 22-33 PLGFLSQDHSVN Truncated Hoa truncated Hoa truncated Hoa truncated Hoa truncated Hoa truncated Hoa truncated Hoa truncated Hoa truncated Hoa truncated Hoa truncated Hoa truncated Hoa truncated Hoa truncated Hoa truncated Hoa truncated Hoa truncated Hoa truncated Hoa 404 PLGFLSQDHSVN >gi|337 |LiLab|LL337 CPRP (CHH precursor-related CPRP-A (SN-16) 24-34, B&C 23-33 LGFLSQDHSVN LGFLSQDHSVN >gi|338 |LiLab|LL338 CPRP (CHH precursor-related CPRP-A (SN-16) 26-33 FLSQDHSV FLSQDHSV >gi|339 |LiLab|LL339 CPRP (CHH precursor-related CPRP-A (SN-16) 26-34 FLSQDHSVN FLSQDHSVN >gi|340 |LiLab|LL340 CPRP (CHH precursor-related CPRP-A (SN-16) 9-33 MEKLLSSSNSPSSTPLGFLSQDHSV MEKLLSSSNSPSSTPLGFLSQDHSV >gi|341 |LiLab|LL341 CPRP (CHH precursor-related CPRP-A (SN-16) 9-34 MEKLLSSSNSPSSTPLGFLSQDHSVN MEKLLSSSNSPSSTPLGFLSQDHSVN >gi|342 |LiLab|LL342 CPRP (CHH precursor-related CPRP-A (SN-16, I-16) 1-12 RSVEGASRMEKL RSVEGASRMEKL >gi|343 |LiLab|LL343 CPRP (CHH precursor-related CPRP-A (SN-16, I-16) 1-13 RSVEGASRMEKLL RSVEGASRMEKLL >gi|344 |LiLab|LL344 CPRP (CHH precursor-related CPRP-A (SN-16, I-16) 1-14 RSVEGASRMEKLLS RSVEGASRMEKLLS >gi|345 |LiLab|LL345 CPRP (CHH precursor-related CPRP-A (SN-16, I-16) 1-15 RSVEGASRMEKLLSS RSVEGASRMEKLLSS >gi|346 |LiLab|LL346 CPRP (CHH precursor-related CPRP-A20-32, B19-31 SSTPLGFLSQDHS SSTPLGFLSQDHS >gi|347 |LiLab|LL347 CPRP (CHH precursor-related CPRP-A20-34, B19-33 SSTPLGFLSQDHSVN SSTPLGFLSQDHSVN >gi|348 |LiLab|LL348 CPRP (CHH precursor-related CPRP-A21-32, B20-31 STPLGFLSQDHS STPLGFLSQDHS >gi|349 |LiLab|LL349 CPRP (CHH precursor-related CPRP-A21-34, B20-33 STPLGFLSQDHSVN STPLGFLSQDHSVN >gi|350 |LiLab|LL350 CPRP (CHH precursor-related CPRP-B 11-23 KLLSSISPSSTPL KLLSSISPSSTPL >gi|351 |LiLab|LL351 CPRP (CHH precursor-related CPRP-B 11-25 KLLSSISPSSTPLGF KLLSSISPSSTPLGF >gi|352 |LiLab|LL352 CPRP (CHH precursor-related CPRP-B 11-26 KLLSSISPSSTPLGFL KLLSSISPSSTPLGFL >gi|353 |LiLab|LL353 CPRP (CHH precursor-related CPRP-B 11-28 KLLSSISPSSTPLGFLSQ KLLSSISPSSTPLGFLSQ >gi|354 |LiLab|LL354 CPRP (CHH precursor-related CPRP-B 11-32 KLLSSISPSSTPLGFLSQDHSV peptide) truncated Hoa peptide) truncated Hoa peptide) truncated Hoa peptide) truncated Hoa peptide) truncated Hoa peptide) Truncated Hoa peptide) Truncated Hoa peptide) Truncated Hoa peptide) Truncated Hoa peptide) truncated Hoa peptide) truncated Hoa peptide) truncated Hoa peptide) truncated Hoa peptide) truncated Hoa peptide) truncated Hoa peptide) truncated Hoa peptide) truncated Hoa peptide) truncated Hoa 405 KLLSSISPSSTPLGFLSQDHSV >gi|355 |LiLab|LL355 CPRP (CHH precursor-related CPRP-B 11-33 KLLSSISPSSTPLGFLSQDHSVN KLLSSISPSSTPLGFLSQDHSVN >gi|356 |LiLab|LL356 CPRP (CHH precursor-related CPRP-B 12-32 LLSSISPSSTPLGFLSQDHSV LLSSISPSSTPLGFLSQDHSV >gi|357 |LiLab|LL357 CPRP (CHH precursor-related CPRP-B 12-33 LLSSISPSSTPLGFLSQDHSVN LLSSISPSSTPLGFLSQDHSVN >gi|358 |LiLab|LL358 CPRP (CHH precursor-related CPRP-B 1-24 RSVEGVSRMEKLLSSISPSSTPLG RSVEGVSRMEKLLSSISPSSTPLG >gi|359 |LiLab|LL359 CPRP (CHH precursor-related CPRP-B 1-26 RSVEGVSRMEKLLSSISPSSTPLGFL RSVEGVSRMEKLLSSISPSSTPLGFL >gi|360 |LiLab|LL360 CPRP (CHH precursor-related CPRP-B 1-28 RSVEGVSRMEKLLSSISPSSTPLGFLSQ RSVEGVSRMEKLLSSISPSSTPLGFLSQ >gi|361 |LiLab|LL361 CPRP (CHH precursor-related CPRP-B 1-32 RSVEGVSRMEKLLSSISPSSTPLGFLSQDHSV RSVEGVSRMEKLLSSISPSSTPLGFLSQDHSV >gi|362 |LiLab|LL362 CPRP (CHH precursor-related CPRP-B 13-31 SISPSSTPLGFLSQDHS SISPSSTPLGFLSQDHS >gi|363 |LiLab|LL363 CPRP (CHH precursor-related CPRP-B 13-32 LSSISPSSTPLGFLSQDHSV LSSISPSSTPLGFLSQDHSV >gi|364 |LiLab|LL364 CPRP (CHH precursor-related CPRP-B 13-32 SISPSSTPLGFLSQDHSV SISPSSTPLGFLSQDHSV >gi|365 |LiLab|LL365 CPRP (CHH precursor-related CPRP-B 13-33 LSSISPSSTPLGFLSQDHSVN LSSISPSSTPLGFLSQDHSVN >gi|366 |LiLab|LL366 CPRP (CHH precursor-related CPRP-B 13-33 SISPSSTPLGFLSQDHSVN SISPSSTPLGFLSQDHSVN >gi|367 |LiLab|LL367 CPRP (CHH precursor-related CPRP-B 14-31 SSISPSSTPLGFLSQDHS SSISPSSTPLGFLSQDHS >gi|368 |LiLab|LL368 CPRP (CHH precursor-related CPRP-B 14-32 SSISPSSTPLGFLSQDHSV SSISPSSTPLGFLSQDHSV >gi|369 |LiLab|LL369 CPRP (CHH precursor-related CPRP-B 14-33 SSISPSSTPLGFLSQDHSVN SSISPSSTPLGFLSQDHSVN >gi|370 |LiLab|LL370 CPRP (CHH precursor-related CPRP-B 2-12 SVEGVSRMEKL SVEGVSRMEKL >gi|371 |LiLab|LL371 CPRP (CHH precursor-related CPRP-B 2-33 SVEGVSRMEKLLSSISPSSTPLGFLSQDHSVN SVEGVSRMEKLLSSISPSSTPLGFLSQDHSVN >gi|372 |LiLab|LL372 CPRP (CHH precursor-related CPRP-B 9-33 EKLLSSISPSSTPLGFLSQDHSVN peptide) truncated Hoa peptide) truncated Hoa peptide) truncated Hoa peptide) truncated Hoa peptide) truncated Hoa peptide) truncated Hoa peptide) truncated Hoa peptide) truncated Hoa peptide) truncated Hoa peptide) truncated Hoa peptide) truncated Hoa peptide) truncated Hoa peptide) truncated Hoa peptide) truncated Hoa peptide) truncated Hoa peptide) truncated Hoa peptide) truncated Hoa peptide) truncated Hoa 406 EKLLSSISPSSTPLGFLSQDHSVN >gi|373 |LiLab|LL373 CPRP (CHH precursor-related peptide) truncated Hoa CPRP-B&C 1-10 RSVEGVSRME RSVEGVSRME >gi|374 |LiLab|LL374 CPRP (CHH precursor-related peptide) truncated Hoa CPRP-B&C 1-12 RSVEGVSRMEKL RSVEGVSRMEKL >gi|375 |LiLab|LL375 CPRP (CHH precursor-related peptide) truncated Hoa CPRP-B&C 1-14 RSVEGVSRMEKLLS RSVEGVSRMEKLLS >gi|376 |LiLab|LL376 CPRP (CHH precursor-related peptide) truncated Hoa CPRP-B&C 1-19 RSVEGVSRMEKLLSSISPS RSVEGVSRMEKLLSSISPS >gi|377 |LiLab|LL377 CPRP (CHH precursor-related peptide) truncated Hoa CPRP-C 11-23 KLLSSISPSSMPLG KLLSSISPSSMPLG >gi|378 |LiLab|LL378 CPRP (CHH precursor-related peptide) truncated Hoa CPRP-C 11-33 KLLSSISPSSMPLGFLSQDHSVN KLLSSISPSSMPLGFLSQDHSVN >gi|379 |LiLab|LL379 CPRP (CHH precursor-related peptide) truncated Hoa CPRP-C 1-23 RSVEGVSRMEKLLSSISPSSMPL RSVEGVSRMEKLLSSISPSSMPL >gi|380 |LiLab|LL380 CPRP (CHH precursor-related peptide) truncated Hoa CPRP-C 1-24 RSVEGVSRMEKLLSSISPSSMPLG RSVEGVSRMEKLLSSISPSSMPLG >gi|381 |LiLab|LL381 CPRP (CHH precursor-related peptide) truncated Hoa CPRP-C 1-32 RSVEGVSRMEKLLSSISPSSMPLGFLSQDHSV RSVEGVSRMEKLLSSISPSSMPLGFLSQDHSV >gi|382 |LiLab|LL382 CPRP (CHH precursor-related peptide) truncated Hoa CPRP-C 16-32 ISPSSMPLGFLSQDHSV ISPSSMPLGFLSQDHSV >gi|383 |LiLab|LL383 CPRP (CHH precursor-related peptide) truncated Hoa CPRP-C 16-33 ISPSSMPLGFLSQDHSVN ISPSSMPLGFLSQDHSVN >gi|384 |LiLab|LL384 CPRP (CHH precursor-related peptide) truncated Hoa CPRP-C 20-33 SMPLGFLSQDHSVN SMPLGFLSQDHSVN >gi|385 |LiLab|LL385 Ecdysis triggering hormone ETH DPSPEPFNPNYNRFRQKIPRIamide DPSPEPFNPNYNRFRQKIPRI >gi|386 |LiLab|LL386 Eclosion hormone AVAANRKVSICIKNCGQCKKMYTDYFNGGLCGDFCLQTEGRFIPDCNRPDILIPFFLQRLE AVAANRKVSICIKNCGQCKKMYTDYFNGGLCGDFCLQTEGRFIPDCNRPDILIPFFLQRLE >gi|387 |LiLab|LL387 Kinins cholecyctokinin cck8 DYMGWMDFamide DYMGWMDF >gi|388 |LiLab|LL388 Kinins Lepidopteran peptide helicokinin I YFSPWGamide YFSPWG >gi|389 |LiLab|LL389 Kinins Lepidopteran peptide helicokinin II VRFSPWGamide VRFSPWG >gi|390 |LiLab|LL390 Kinins Lepidopteran peptide helicokinin III KVKFSAWGamide KVKFSAWG 407 >gi|391 |LiLab|LL391 Kinins pev-Kinin 1 ASFSPWGamide ASFSPWG >gi|392 |LiLab|LL392 Kinins pev-Kinin 2 DFSAWAamide Cp.PO. DFSAWA >gi|393 |LiLab|LL393 Kinins pev-Kinin 3 PAFSPWGamide PAFSPWG >gi|394 |LiLab|LL394 Kinins pev-Kinin 4 VAFSPWGamide VAFSPWG >gi|395 |LiLab|LL395 Kinins pev-Kinin 5 pEAFSPWAamide QAFSPWA >gi|396 |LiLab|LL396 Kinins pev-Kinin 6 AFSPWAamide AFSPWA >gi|397 |LiLab|LL397 myosuppressin pQDLDHVFLR Csap PO QDLDHVFLR >gi|398 |LiLab|LL398 Neuroparsin APRCDRHDEEAPKNCKYGTTQDWCKNGVCAKGPGETCGGYRWSEGKCGEGTPCSCGICGGCSPFDGKCGPTSI C APRCDRHDEEAPKNCKYGTTQDWCKNGVCAKGPGETCGGYRWSEGKCGEGTPCSCGICGGCSPFDGKCGPTSI C >gi|399 |LiLab|LL399 Orcokinin DEIDRSGFGFA Cp.SG Csap PO DEIDRSGFGFA >gi|400 |LiLab|LL400 Orcokinin DFDEIDRSGFA Csap SG Cb br,PO,SG DFDEIDRSGFA >gi|401 |LiLab|LL401 Orcokinin DFDEIDRSGFG Csap SG DFDEIDRSGFG >gi|402 |LiLab|LL402 Orcokinin DFDEIDRSGFGFA Csap SG DFDEIDRSGFGFA >gi|403 |LiLab|LL403 Orcokinin DFDEIDRSGFGFV Csap SG DFDEIDRSGFGFV >gi|404 |LiLab|LL404 Orcokinin DFDEIDRSSFA Csap SG,PO DFDEIDRSSFA >gi|405 |LiLab|LL405 Orcokinin DFDEIDRSSFG Csap SG DFDEIDRSSFG >gi|406 |LiLab|LL406 Orcokinin DFDEIDRSSFGFA Csap SG DFDEIDRSSFGFA >gi|407 |LiLab|LL407 Orcokinin DFDEIDRSSFGFN Csap SG DFDEIDRSSFGFN >gi|408 |LiLab|LL408 Orcokinin DFDEIDRSSFGFV DFDEIDRSSFGFV >gi|409 |LiLab|LL409 Orcokinin DFEDIERSGFGFV Cmaen VNC/PO DFEDIERSGFGFV >gi|410 |LiLab|LL410 Orcokinin EIDRSGFGF Csap SG EIDRSGFGF >gi|411 |LiLab|LL411 Orcokinin EIDRSGFGFA Csap SG,PO Cmaen PO Cb br,STG,CoG EIDRSGFGFA >gi|412 |LiLab|LL412 Orcokinin EIDRSSFGFN Cp.SG EIDRSSFGFN >gi|413 |LiLab|LL413 Orcokinin FDEIDRSGFA Csap SG FDEIDRSGFA >gi|414 |LiLab|LL414 Orcokinin FDEIDRSGFG Csap SG FDEIDRSGFG >gi|415 |LiLab|LL415 Orcokinin FDEIDRSGFGFA Csap PO Cmaen PO FDEIDRSGFGFA 408 >gi|416 |LiLab|LL416 Orcokinin FDEIDRSSFA Csap SG FDEIDRSSFA >gi|417 |LiLab|LL417 Orcokinin NFDEIDRSA Csap SG NFDEIDRSA >gi|418 |LiLab|LL418 Orcokinin NFDEIDRSGFA Csap SG,PO,br Cmaen VNC/PO Cb br,Sg,STG,CoG NFDEIDRSGFA >gi|419 |LiLab|LL419 Orcokinin NFDEIDRSGFamide Hoa. SG/CNS Csap PO,br Cmaen Br Cb br,SG,CoG NFDEIDRSGF >gi|420 |LiLab|LL420 Orcokinin NFDEIDRSGFGFH Hoa. SG/CNS NFDEIDRSGFGFH >gi|421 |LiLab|LL421 Orcokinin NFDEIDRSSFamide Csap SG,PO,br Cmaen Br NFDEIDRSSF >gi|422 |LiLab|LL422 Orcokinin NFDEIDRSSFG Csap SG,PO Cmaen VNC NFDEIDRSSFG >gi|423 |LiLab|LL423 Orcokinin NFDEIDRSSFGF Csap SG,PO,br NFDEIDRSSFGF >gi|424 |LiLab|LL424 Orcokinin NFDEIDRSSFGFA Csap br NFDEIDRSSFGFA >gi|425 |LiLab|LL425 Orcokinin SSEDMDRLGFA Hoa. SG/CNS SSEDMDRLGFA >gi|426 |LiLab|LL426 Orcokinin VYGPRDIANLY Hoa. SG/CNS VYGPRDIANLY >gi|427 |LiLab|LL427 Orcokinin [Ala13]-orcokinin NFDEIDRSGFGFA Csap SG,PO,br Cmaen Br/VNC/SG/PO Cb br,PO,SG,STG,CoG NFDEIDRSGFGFA >gi|428 |LiLab|LL428 Orcokinin [Ala8-Ala13]-Orcokinin NFDEIDRAGFGFA NFDEIDRAGFGFA >gi|429 |LiLab|LL429 Orcokinin [Asn13]-OrcoKinin NFDEIDRSGFGFN Hoa.SG/CNS Cmaen PO NFDEIDRSGFGFN >gi|430 |LiLab|LL430 Orcokinin [Ser9]-Orcokinin NFDEIDRSSFGFN Csap SG,PO,br Cmaen Br/VNC/SG/PO Cb br,SG,STG,CoG NFDEIDRSSFGFN >gi|431 |LiLab|LL431 Orcokinin [Ser9-val13]-Orcokinin NFDEIDRSSFGFV Csap SG,PO,br Cmaen VNC/SG/PO Cb br,PO,SG,STG,CoG NFDEIDRSSFGFV >gi|432 |LiLab|LL432 Orcokinin [Thr8-His13]-Orcokinin NFDEIDRTGFGFH Csap br Cb br,PO,SG,STG,CoG NFDEIDRTGFGFH >gi|433 |LiLab|LL433 Orcokinin [val13]-OrcoKinin NFDEIDRSGFGFV Csap SG,PO, br Cmaen VNC/SG/PO Cb br,PO,SG,STG,CoG NFDEIDRSGFGFV >gi|434 |LiLab|LL434 Orcokinin Hoa-Orcokinin SSEDMDRLGFG Hoa. SG/CNS SSEDMDRLGFG >gi|435 |LiLab|LL435 Orcokinin Hoa-Orcokinin SSEDMPSSLGFGFN Hoa. SG/CNS SSEDMPSSLGFGFN >gi|436 |LiLab|LL436 Orcokinin Orcokinin[1-11] NFDEIDRSGFG Csap SG,PO,br Cmaen Br/VNC Cb br,PO,SGSTG,CoG NFDEIDRSGFG >gi|437 |LiLab|LL437 Orcokinin Orcokinin[1-12] NFDEIDRSGFGF Csap SG,PO,br Cmaen VNC/PO Cb br,PO,SG,STG,CoG NFDEIDRSGFGF 409 >gi|438 |LiLab|LL438 Orcokinin Orcokinin-like peptides NFDEIDRSSFA Csap SG,PO,br Cmaen Br NFDEIDRSSFA >gi|439 |LiLab|LL439 Orcokinin/Orcomyotropin-related TPRDIANLYamide Csap SG TPRDIANLY >gi|440 |LiLab|LL440 Orcomyotropin FDAFTTGFamide Hoa.CNS FDAFTTGF >gi|441 |LiLab|LL441 Orcomyotropin FDAFTTGFGHN Hoa. SG/CNS FDAFTTGFGHN >gi|442 |LiLab|LL442 Orcomyotropin FDAFTTGFGHS Csap PO,SG,br Cmaen Br/VNC/SG/PO Cb br,PO,SG,STG,CoG FDAFTTGFGHS >gi|443 |LiLab|LL443 Orcomyotropin FPAFTTGFGSH Csap SG FPAFTTGFGSH >gi|444 |LiLab|LL444 Others AVLLPKKTEKK AVLLPKKTEKK >gi|445 |LiLab|LL445 Others DLPKVDTALK DLPKVDTALK >gi|446 |LiLab|LL446 Others EVEEPEAPAPPAK EVEEPEAPAPPAK >gi|447 |LiLab|LL447 Others GPSGGFNGALAR GPSGGFNGALAR >gi|448 |LiLab|LL448 Others HL/IGSL/IYRamide Csap PO,br Cmaen Br/SG/PO Cb br,PO,SG,STG,CoG HLGSLYR >gi|449 |LiLab|LL449 Others KPKTEKK KPKTEKK >gi|450 |LiLab|LL450 Others [Leu]-enkephalin YGGFL YGGFL >gi|451 |LiLab|LL451 Others [Met]-enkephalin YGGFM YGGFM >gi|452 |LiLab|LL452 PDH(Pigment-dispersing hormone) NSELINSLLGISRLMNEAamide Cp.SG NSELINSLLGISRLMNEA >gi|453 |LiLab|LL453 PDH(Pigment-dispersing hormone) pQELHVPEREA Csap SG QELHVPEREA >gi|454 |LiLab|LL454 PDH(Pigment-dispersing hormone) Pandalus borealis PDH NSGMINSILGIPRVMTEAamide NSGMINSILGIPRVMTEA >gi|455 |LiLab|LL455 PDH(Pigment-dispersing hormone) PDH-armadillidium QDLNPTEKEVLSNMLDFLQRHSRTTYMFPLLSESKRNSELINSLLGAPRVLNNAamide armadillidium, predicted QDLNPTEKEVLSNMLDFLQRHSRTTYMFPLLSESKRNSELINSLLGAPRVLNNA >gi|456 |LiLab|LL456 PDH(Pigment-dispersing hormone) PDH-Callinectes-I QELKYQEREMVAELAQQIYRVAQAPWAAAVGPHKRNSELINSILGLPKVMNDAamide Callinectes-I, predicted QELKYQEREMVAELAQQIYRVAQAPWAAAVGPHKRNSELINSILGLPKVMNDA >gi|457 |LiLab|LL457 PDH(Pigment-dispersing hormone) PDH-Callinectes-II QELHVPEREAVANLAARILKIVHAPHDAAGVPHKRNSELINSLLGISALMNEAamide CallinectesII, predicted QELHVPEREAVANLAARILKIVHAPHDAAGVPHKRNSELINSLLGISALMNEA 410 >gi|458 |LiLab|LL458 PDH(Pigment-dispersing hormone) PDH-Cancer-I QDLKYQAREMVAELAQQIYRVAQAPQAGAVGPHKRNSELINSILGLPKVMNDAamide Cancer-I, predicted QDLKYQAREMVAELAQQIYRVAQAPQAGAVGPHKRNSELINSILGLPKVMNDA >gi|459 |LiLab|LL459 PDH(Pigment-dispersing hormone) PDH-Cancer-Iia QEVNVSEREAVATLAAHILKVVCAPLEGAGGLPHKRNSELINSILGLPKVMNEAamide Cancer-IIa, predicted QEVNVSEREAVATLAAHILKVVCAPLEGAGGLPHKRNSELINSILGLPKVMNEA >gi|460 |LiLab|LL460 PDH(Pigment-dispersing hormone) PDH-Carcinus QDLKYQEREMVAELAQQIYRVAQAPWAGAVGPHKRNSELINSILGLPKVMNDAamide carcinus, predicted QDLKYQEREMVAELAQQIYRVAQAPWAGAVGPHKRNSELINSILGLPKVMNDA >gi|461 |LiLab|LL461 PDH(Pigment-dispersing hormone) PDH-eurydice QSRDFSISEREIVASLAKQLLRVARMGYVPEGDLPRKRNAELINSLLGVPRVMSDAamide eurydice, predicted QSRDFSISEREIVASLAKQLLRVARMGYVPEGDLPRKRNAELINSLLGVPRVMSDA >gi|462 |LiLab|LL462 PDH(Pigment-dispersing hormone) PDH-Marsupenaeus-I DSSLKYFEREVVSELAAQILRVAQGPSAFVAGPHKRNSELINSLLGIPKVMTDAamide MarsupenaeusI, predicted DSSLKYFEREVVSELAAQILRVAQGPSAFVAGPHKRNSELINSLLGIPKVMTDA >gi|463 |LiLab|LL463 PDH(Pigment-dispersing hormone) PDH-Marsupenaeus-II QREPTASKCQAATELAIQILQAVKGAHTGVAAGPHKRNSELINSLLGLPKFMIDAamide Marsupenaeus-II, predicted QREPTASKCQAATELAIQILQAVKGAHTGVAAGPHKRNSELINSLLGLPKFMIDA >gi|464 |LiLab|LL464 PDH(Pigment-dispersing hormone) PDH-Marsupenaeus-III QEDLKYFEREVVSELAAQILRVAQGPSAFVAGPHKRNSELINSLLGIPKVMNDAamide MarsupenaeusIII, predicted QEDLKYFEREVVSELAAQILRVAQGPSAFVAGPHKRNSELINSLLGIPKVMNDA >gi|465 |LiLab|LL465 PDH(Pigment-dispersing hormone) PDH-orconectes QELKYPEREVVAELAAQIYRVAQAPWAGAVGPHKRNSELINSILGLPKVMNEAamide orconectes, predicted QELKYPEREVVAELAAQIYRVAQAPWAGAVGPHKRNSELINSILGLPKVMNEA >gi|466 |LiLab|LL466 PDH(Pigment-dispersing hormone) Penaeus aztecus PDH NSELINSLLGIPKVMNDAamide NSELINSLLGIPKVMNDA >gi|467 |LiLab|LL467 PDH(Pigment-dispersing hormone) Procambarus clarkii PDH NSELINSILGLPKVMNEAamide NSELINSILGLPKVMNEA >gi|468 |LiLab|LL468 PDH(Pigment-dispersing hormone) Uca pugilator, Cancer magister PDH NSELINSILGLPKVMNDAamide Cmaen SG Cb br, SG NSELINSILGLPKVMNDA >gi|469 |LiLab|LL469 Proctolin Proctolin RYLPT Csap PO Cmaen PO Cb br,PO,SG,STG,CoG RYLPT >gi|470 |LiLab|LL470 Pyrokinin FSPRLamide Hoa.brain Cb brain FSPRL >gi|471 |LiLab|LL471 Pyrokinin LYFAPRLamide Cmaen Br Cb br LYFAPRL >gi|472 |LiLab|LL472 Pyrokinin SGGFAFSPRLamide Cb br,STG,CoG SGGFAFSPRL >gi|473 |LiLab|LL473 Pyrokinin TDGFAFSPRLamide Cmaen Br TDGFAFSPRL >gi|474 |LiLab|LL474 Pyrokinin TSFAFSPRLamide Cmaen Br TSFAFSPRL 411 >gi|475 |LiLab|LL475 br,CoG TNFAFSPRL >gi|476 |LiLab|LL476 AHKNFLRF >gi|477 |LiLab|LL477 AHRNFLRF >gi|478 |LiLab|LL478 APNKNFLRF >gi|479 |LiLab|LL479 APQGNFLRF >gi|480 |LiLab|LL480 APRNFLRF >gi|481 |LiLab|LL481 ARPRNFLRF >gi|482 |LiLab|LL482 ASNNLRF >gi|483 |LiLab|LL483 AYPSLRLRF >gi|484 |LiLab|LL484 DARTAPLRLRF >gi|485 |LiLab|LL485 DENRNFLRF >gi|486 |LiLab|LL486 DGNRNFLRF >gi|487 |LiLab|LL487 DGPLAPFLRF >gi|488 |LiLab|LL488 DHVPFLRF >gi|489 |LiLab|LL489 DQNRNFLRF >gi|490 |LiLab|LL490 DRNFVLRF >gi|491 |LiLab|LL491 DTSTPALRLRF >gi|492 |LiLab|LL492 EFLRF >gi|493 |LiLab|LL493 EFVDPNLRF >gi|494 |LiLab|LL494 ELNFLRF >gi|495 |LiLab|LL495 EMPSLRLRF >gi|496 |LiLab|LL496 ENRNFLRF >gi|497 |LiLab|LL497 EPLDHVFLRF >gi|498 |LiLab|LL498 ETNFLRF >gi|499 |LiLab|LL499 FDDFLRF >gi|500 |LiLab|LL500 FEPSLRLRF >gi|501 |LiLab|LL501 Pyrokinin Cb Pyrokinin I TNFAFSPRLamide Csap PO Cb RFamide AHKNFLRFamide Csap PO Cb PO RFamide AHRNFLRFamide Cb PO RFamide APNKNFLRFamide Cb PO RFamide APQGNFLRFamide Csap PO Cmaen Br/PO RFamide APRNFLRFamide Cmaen VNC/PO Cb br CoG RFamide ARPRNFLRFamide Cb PO RFamide ASNNLRFamide Csap PO RFamide AYPSLRLRFamide Hoa.CNS RFamide DARTAPLRLRFamide Csap br Cmaen Br/VNC RFamide DENRNFLRFamide Csap PO, Cb PO RFamide DGNRNFLRFamide Cmaen Br/VNC/PO RFamide DGPLAPFLRFamide Csap PO RFamide DHVPFLRFamide Cb PO RFamide DQNRNFLRFamide Hoa.CNS RFamide DRNFVLRFamide Cb PO RFamide DTSTPALRLRFamide Hoa.CNS RFamide EFLRFamide Csap PO RFamide EFVDPNLRFamide Csap PO RFamide ELNFLRFamide Csap PO RFamide EMPSLRLRFamide Cmaen VNC RFamide ENRNFLRFamide Cb PO RFamide EPLDHVFLRFamide Csap RFamide ETNFLRFamide Csap PO RFamide FDDFLRFamide Csap PO RFamide FEPSLRLRFamide Hoa.CNS RFamide FTSKNYLRFamide Cb,PO,STG 412 FTSKNYLRF >gi|502 |LiLab|LL502 RFamide GAHKNYLRFamide Csap SG,PO,br Cmaen PO Cb br,PO,STG,CoG GAHKNYLRF >gi|503 |LiLab|LL503 RFamide GGGEYDDYGHLRFamide Hoa.CNS GGGEYDDYGHLRF >gi|504 |LiLab|LL504 RFamide GHRNFLRFamide Cb PO GHRNFLRF >gi|505 |LiLab|LL505 RFamide GLSRNYLRFamide Csap PO,br Cmaen Br/VNC/PO GLSRNYLRF >gi|506 |LiLab|LL506 RFamide GNRNFLRFamide Csap PO Cmaen Br/PO Cb br,PO,STG,CoG GNRNFLRF >gi|507 |LiLab|LL507 RFamide GPFLRFamide Csap PO Cb PO GPFLRF >gi|508 |LiLab|LL508 RFamide GPKNFLRFamide Csap GPKNFLRF >gi|509 |LiLab|LL509 RFamide GPPSLRLRFamide Hoa.CNS GPPSLRLRF >gi|510 |LiLab|LL510 RFamide GPRNFLRFamide Csap PO GPRNFLRF >gi|511 |LiLab|LL511 RFamide GRNFLRFamide Cb br,PO GRNFLRF >gi|512 |LiLab|LL512 RFamide GYNRSFLRFamide GYNRSFLRF >gi|513 |LiLab|LL513 RFamide GYSDRNYLRFamide Hoa.CNS GYSDRNYLRF >gi|514 |LiLab|LL514 RFamide GYSKNYLRFamide Csap PO Cb br,PO,STG,CoG GYSKNYLRF >gi|515 |LiLab|LL515 RFamide GYSVGLNYLRFamide Csap PO GYSVGLNYLRF >gi|516 |LiLab|LL516 RFamide HDLVQVFLRFamide Csap HDLVQVFLRF >gi|517 |LiLab|LL517 RFamide HDSPHVFLRFamide Csap HDSPHVFLRF >gi|518 |LiLab|LL518 RFamide HPLSFVSALRFamide Csap HPLSFVSALRF >gi|519 |LiLab|LL519 RFamide HVFLRFamide Csap PO HVFLRF >gi|520 |LiLab|LL520 RFamide LAPQRNFLRFamide Csap LAPQRNFLRF >gi|521 |LiLab|LL521 RFamide LAYNRSFLRFamide Csap LAYNRSFLRF >gi|522 |LiLab|LL522 RFamide LDGPLAPFLRFamide Csap LDGPLAPFLRF >gi|523 |LiLab|LL523 RFamide LDRNFLRFamide Csap LDRNFLRF >gi|524 |LiLab|LL524 RFamide LELNFLRFamide Csap LELNFLRF >gi|525 |LiLab|LL525 RFamide LETNFLRFamide Csap LETNFLRF >gi|526 |LiLab|LL526 RFamide LFDDFLRFamide Csap LFDDFLRF >gi|527 |LiLab|LL527 RFamide LGDRNFLRFamide Csap 413 LGDRNFLRF >gi|528 |LiLab|LL528 LGRPNFLRF >gi|529 |LiLab|LL529 LNPFLRF >gi|530 |LiLab|LL530 LNPSNFLRF >gi|531 |LiLab|LL531 LNQPNFLRF >gi|532 |LiLab|LL532 LNRNFLRF >gi|533 |LiLab|LL533 LNRSFLRF >gi|534 |LiLab|LL534 LPGVNFLRF >gi|535 |LiLab|LL535 LRNLRF >gi|536 |LiLab|LL536 LRQFLRF >gi|537 |LiLab|LL537 LSPRNFLRF >gi|538 |LiLab|LL538 LTGNRNFLRF >gi|539 |LiLab|LL539 LTHPFLRF >gi|540 |LiLab|LL540 LTNRNFLRF >gi|541 |LiLab|LL541 LTNYGGFLRF >gi|542 |LiLab|LL542 LTRPLRF >gi|543 |LiLab|LL543 LYEQDFLRF >gi|544 |LiLab|LL544 MPYLRF >gi|545 |LiLab|LL545 NFLRF >gi|546 |LiLab|LL546 NPSDFLRF >gi|547 |LiLab|LL547 NPSNFLRF >gi|548 |LiLab|LL548 NQPNFLRF >gi|549 |LiLab|LL549 br,PO NRSFLRF >gi|550 |LiLab|LL550 PGVNFLRF >gi|551 |LiLab|LL551 PKSNFLRF >gi|552 |LiLab|LL552 QDNDHVFLRF >gi|553 |LiLab|LL553 QGNFLRF RFamide LGRPNFLRFamide Csap RFamide LNPFLRFamide RFamide LNPSNFLRFamide Csap RFamide LNQPNFLRFamide Csap RFamide LNRNFLRFamide Csap RFamide LNRSFLRFamide Csap RFamide LPGVNFLRFamide Csap RFamide LRNLRFamide Csap PO RFamide LRQFLRFamide Csap RFamide LSPRNFLRFamide Csap RFamide LTGNRNFLRFamide Csap RFamide LTHPFLRFamide Csap RFamide LTNRNFLRFamide Csap RFamide LTNYGGFLRFamide Csap RFamide LTRPLRFamide Csap br RFamide LYEQDFLRFamide Csap RFamide MPYLRFamide Csap PO RFamide NFLRFamide Hoa.CNS Csap PO Cb br PO RFamide NPSDFLRFamide Csap PO Cb PO RFamide NPSNFLRFamide Csap PO Cb PO RFamide NQPNFLRFamide Csap PO RFamide NRSFLRFamide Csap PO,br Cmaen Br/VNC/PO Cb RFamide PGVNFLRFamide Csap PO RFamide PKSNFLRFamide Cb PO RFamide pQDNDHVFLRFamide Csap br RFamide pQGNFLRFamide Csap PO Cmaen PO 414 >gi|554 |LiLab|LL554 RFamide pQRNFLRFamide Cb br,PO QRNFLRF >gi|555 |LiLab|LL555 RFamide PSLRLRFamide Hoa.CNS Cmaen Br/VNC Cb br PSLRLRF >gi|556 |LiLab|LL556 RFamide PSMRLRFamide Csap br Cmaen Br/VNC Cb br CoG PSMRLRF >gi|557 |LiLab|LL557 RFamide QYFMRLFRamide Csap PO QYFMRLFR >gi|558 |LiLab|LL558 RFamide RDRNFLRFamide Cb br,PO RDRNFLRF >gi|559 |LiLab|LL559 RFamide RNFLRFamide Csap PO Cmaen Br/VNC/PO Cb br PO RNFLRF >gi|560 |LiLab|LL560 RFamide RQFLRFamide Csap RQFLRF >gi|561 |LiLab|LL561 RFamide RSFLRFamide Cp.PO RSFLRF >gi|562 |LiLab|LL562 RFamide SDRNYLRFamide SDRNYLRF >gi|563 |LiLab|LL563 RFamide SENRNFLRFamide Csap PO Cmaen VNC Cb br,PO,STG,CoG SENRNFLRF >gi|564 |LiLab|LL564 RFamide SGRNFLRFamide Hoa.CNS SGRNFLRF >gi|565 |LiLab|LL565 RFamide SKNYLRFamide Csap PO Cb PO CoG SKNYLRF >gi|566 |LiLab|LL566 RFamide SMPTLRLRFamide Csap br SMPTLRLRF >gi|567 |LiLab|LL567 RFamide SPRNFLRFamide Csap PO SPRNFLRF >gi|568 |LiLab|LL568 RFamide SQGLNSDLRFamide Csap PO SQGLNSDLRF >gi|569 |LiLab|LL569 RFamide SQPSKNYLRFamide Csap PO Cb PO SQPSKNYLRF >gi|570 |LiLab|LL570 RFamide SRNYLRFamide Cmaen Br/VNC/PO SRNYLRF >gi|571 |LiLab|LL571 RFamide TGNRNFLRFamide Csap PO TGNRNFLRF >gi|572 |LiLab|LL572 RFamide THPFLRFamide Csap PO THPFLRF >gi|573 |LiLab|LL573 RFamide TNYGGFLRFamide Csap PO TNYGGFLRF >gi|574 |LiLab|LL574 RFamide TRNFLRFamide TRNFLRF >gi|575 |LiLab|LL575 RFamide YEQDFLRFamide Csap PO YEQDFLRF >gi|576 |LiLab|LL576 RFamide YGNRPHLRFamide Csap YGNRPHLRF >gi|577 |LiLab|LL577 RFamide YGSDRNFLRFamide Cb PO YGSDRNFLRF >gi|578 |LiLab|LL578 RFamide [ala1]-FaRP AYNRSFLRFamide Csap PO Cb br,PO,STG,CoG AYNRSFLRF 415 >gi|579 |LiLab|LL579 RFamide [glu2-leu3]-SchistoFLRFa pQDLDHVFLRFamide Csap PO,br Cmaen Br/VNC/PO Cb br,PO,STG,CoG QDLDHVFLRF >gi|580 |LiLab|LL580 RFamide [val2] pem-FLP 6 DVRTPALRLRFamide Cb br,CoG DVRTPALRLRF >gi|581 |LiLab|LL581 RFamide DF 2, Mar-FLP 1, PrcFaRP 5 DRNFLRFamide Csap PO,br Cmaen Br/SG/PO Cb br,PO,STG,CoG DRNFLRF >gi|582 |LiLab|LL582 RFamide F1 SDRNFLRFamide SDRNFLRF >gi|583 |LiLab|LL583 RFamide F2 TNRNFLRFamide Csap PO TNRNFLRF >gi|584 |LiLab|LL584 RFamide Lepidopteran peptide FLRFamide I pEDVVHSFLRFamide QDVVHSFLRF >gi|585 |LiLab|LL585 RFamide Lepidopteran peptide FLRFamide II GNSFLRFamide GNSFLRF >gi|586 |LiLab|LL586 RFamide Lepidopteran peptide FLRFamide III DPSFLEFamide DPSFLEF >gi|587 |LiLab|LL587 RFamide Mar-FLP 2 ADKNFLRFamide ADKNFLRF >gi|588 |LiLab|LL588 RFamide Mar-FLP 3 NYDKNFLRFamide NYDKNFLRF >gi|589 |LiLab|LL589 RFamide Mar-FLP 4, sNPF (short neuropeptide F) APALRLRFamide APALRLRF >gi|590 |LiLab|LL590 RFamide Mar-FLP 5 DRTPALRLRFamide DRTPALRLRF >gi|591 |LiLab|LL591 RFamide Mar-FLP 6 DGGRNFLRFamide DGGRNFLRF >gi|592 |LiLab|LL592 RFamide Mar-FLP 7 GYGDRNFLRFamide GYGDRNFLRF >gi|593 |LiLab|LL593 RFamide Mar-FLP 8 VSHNNFLRFamide VSHNNFLRF >gi|594 |LiLab|LL594 RFamide NF 1, PrcFaRP 2 NRNFLRFamide Csap PO,br Cmaen Br/VNC/SG/PO Cb br,PO,STG,CoG NRNFLRF >gi|595 |LiLab|LL595 RFamide NPF (neuropeptide F) KPDPSQLANMAEALKYLEQELDKYYSQVSRPRFamide KPDPSQLANMAEALKYLEQELDKYYSQVSRPRF >gi|596 |LiLab|LL596 RFamide NPY/PP peptide pem-PYF 3 YAIAGRPRFamide YAIAGRPRF >gi|597 |LiLab|LL597 RFamide NPY/PP peptide pem-PYF2 YSQVSRPRFamide YSQVSRPRF >gi|598 |LiLab|LL598 RFamide NPY/PP peptide pem-PYF4 YSLRARPRFamide YSLRARPRF >gi|599 |LiLab|LL599 RFamide pem-FLP 1 GDRNFLRFamide GDRNFLRF >gi|600 |LiLab|LL600 RFamide pem-FLP 2 AYSNLNYLRFamide AYSNLNYLRF >gi|601 |LiLab|LL601 RFamide pem-FLP 3 AQPSMRLRFamide AQPSMRLRF 416 >gi|602 |LiLab|LL602 RFamide pem-FLP 4 SQPSMRLRFamide SQPSMRLRF >gi|603 |LiLab|LL603 RFamide pem-FLP 5 SMPSLRLRFamide Cmaen Br/VNC/SG Cb br,CoG SMPSLRLRF >gi|604 |LiLab|LL604 RFamide pem-FLP 6 DGRTPALRLRFamide DGRTPALRLRF >gi|605 |LiLab|LL605 RFamide pem-PYF1 RARPRFamide RARPRF >gi|606 |LiLab|LL606 RFamide PrcFaRP 1 FLRFamide AYSDRNFLRFamide AYSDRNFLRF >gi|607 |LiLab|LL607 RFamide PrcFaRP 1 YLRFamide AYSDRNYLRFamide AYSDRNYLRF >gi|608 |LiLab|LL608 RFamide PrcFaRP 3 SRNFLRFamide SRNFLRF >gi|609 |LiLab|LL609 RFamide PrcFaRP 4 ALDRNFLRFamide ALDRNFLRF >gi|610 |LiLab|LL610 RFamide PrcFaRP 6 APQRNFLRFamide Csap PO,br Cmaen VNC Cb br,PO,STG,CoG APQRNFLRF >gi|611 |LiLab|LL611 RFamide S I/LNFTHKFamide I/LNFTHKF >gi|612 |LiLab|LL612 RFamide sulfakinin pQFDEY(sulf)GHMRFamide QFDEYGHMRF >gi|613 |LiLab|LL613 RPCH Anaim-AKH pEVNFSPSWamide Anax imperator EVNFSPSW >gi|614 |LiLab|LL614 RPCH Corpu-AKH pELNFSPSWamide Corixa punctata ELNFSPSW >gi|615 |LiLab|LL615 RPCH Dappu-RPCH pEVNFSTSWamide Daphnia pulex, predicted EVNFSTSW >gi|616 |LiLab|LL616 RPCH Grybi-Akh pEVNFSTGWamide Gryllus bimaculatus EVNFSTGW >gi|617 |LiLab|LL617 RPCH Ile2-Panbo-RPCH pEINFSPGWamide unnatural EINFSPGW >gi|618 |LiLab|LL618 RPCH Manto-CC pEVNFSPGWamide species of order Mantophasmatodea EVNFSPGW >gi|619 |LiLab|LL619 RPCH Nepci-AKH pELNFSSGWamide Nepa cinerea ELNFSSGW >gi|620 |LiLab|LL620 RPCH Panbo-RPCH pELNFSPGWamide Pandalus borealis ELNFSPGW >gi|621 |LiLab|LL621 RPCH RPCH pQLNFSPGWamide Csap SG,br Cmaen SG Cb SG QLNFSPGW >gi|622 |LiLab|LL622 RPCH Schgr-AKH-III pELNFSTGWamide Schistocera gregaria ELNFSTGW >gi|623 |LiLab|LL623 RYamide (X)YANRYamide Cmaen PO YANRY >gi|624 |LiLab|LL624 RYamide EWYSQRYamide Csap PO EWYSQRY >gi|625 |LiLab|LL625 RYamide FVGGSRYamide Csap PO Cmaen PO Cb br,PO,SG FVGGSRY >gi|626 |LiLab|LL626 RYamide FVNSRYamide 417 FVNSRY >gi|627 |LiLab|LL627 RYamide FYANRYamide Csap PO Cmaen PO Cb PO FYANRY >gi|628 |LiLab|LL628 RYamide FYSQRYamide Csap PO Cmaen PO Cb PO FYSQRY >gi|629 |LiLab|LL629 RYamide GFVSNRYamide Csap PO GFVSNRY >gi|630 |LiLab|LL630 RYamide L/IFVGGSRYamide Cp.PO LFVGGSRY >gi|631 |LiLab|LL631 RYamide LEWYSQRYamide Csap LEWYSQRY >gi|632 |LiLab|LL632 RYamide LFYSQRYamide Csap LFYSQRY >gi|633 |LiLab|LL633 RYamide LGFVSNRYamide Csap LGFVSNRY >gi|634 |LiLab|LL634 RYamide LSGFYANRYamide Csap LSGFYANRY >gi|635 |LiLab|LL635 RYamide LSSRFVGGSRYamide Csap LSSRFVGGSRY >gi|636 |LiLab|LL636 RYamide PAFYSQRYamide PAFYSQRY >gi|637 |LiLab|LL637 RYamide pQGFYSQRYamide Csap SG,PO Cmaen PO Cb br,PO,SG,STG,CoG QGFYSQRY >gi|638 |LiLab|LL638 RYamide RFVGGSRYamide Cp.PO RFVGGSRY >gi|639 |LiLab|LL639 RYamide RFYANRYamide Csap PO RFYANRY >gi|640 |LiLab|LL640 RYamide SGFYADRYamide Cmaen PO SGFYADRY >gi|641 |LiLab|LL641 RYamide SGFYANRYamide Csap PO Cmaen PO Cb PO SGFYANRY >gi|642 |LiLab|LL642 RYamide SGFYAPRYamide Csap PO Cmaen PO SGFYAPRY >gi|643 |LiLab|LL643 RYamide SRFVGGSRYamide Csap PO SRFVGGSRY >gi|644 |LiLab|LL644 RYamide SSRFVGGSRYamide Csap PO Cmaen PO Cb PO SSRFVGGSRY >gi|645 |LiLab|LL645 RYamide VGFYANRYamide Csap PO VGFYANRY >gi|646 |LiLab|LL646 SIFamide AYRKPPFNGSIFamide Insect AYRKPPFNGSIF >gi|647 |LiLab|LL647 SIFamide RKPPFNGSIFamide Hoa.CNS Cmaen Br Cb br,STG,CoG RKPPFNGSIF >gi|648 |LiLab|LL648 SIFamide VYRKPPFNGSIFamide Hoa.CNS Cmaen VNC VYRKPPFNGSIF >gi|649 |LiLab|LL649 SIFamide pem-FLP 7 GYRKPPFNGSIFamide Csap br Cmaen Cmaen br/VCN/SG Cb br,SG,STG,CoG GYRKPPFNGSIF >gi|650 |LiLab|LL650 Tachykinin APSGFLGM Csap br APSGFLGM >gi|651 |LiLab|LL651 Tachykinin APSGFLGMRamide-NH3-loss Csap br APSGFLGMR 418 >gi|652 |LiLab|LL652 APSGFLGMRG >gi|653 |LiLab|LL653 PSGFLGMR >gi|654 |LiLab|LL654 Br/VNC/SG Cb br,CoG TPSGFLGMR >gi|655 |LiLab|LL655 br,STG,CoG SGFLGMR >gi|656 |LiLab|LL656 YPSGFLGMR >gi|657 |LiLab|LL657 APSFGQ >gi|658 |LiLab|LL658 GFLGMR Tachykinin APSGFLGMRG Csap PO Cmaen Br Cb SG,CoG Tachykinin PSGFLGMRamide Cmaen Br/VNC/SG Tachykinin CabTRP II TPSGFLGMRamide Csap PO Cmaen Tachykinin CabTRP lb SGFLGMRamide Cmaen Br/VNC Cb Tachykinin CalsTRP YPSGFLGMRamide Csap br Tachykinin TRP APSFGQamide Tachykinin TRP GFLGMRamide Neuropeptides Found in Orconectes rusticus Neuropeptide Family NP Matching Sequence AST-B TNWNKFQGSWamide FMRFamide EPLDHVFLRFamide FMRFamide EPLDHVFLRFamide FMRFamide EPLDHVFLRFamide FMRFamide YAIAGRPRFamide Myosuppressin pQDLDHVFLRFamide Myosuppressin pQDLDHVFLRFamide Myosuppressin pQDLDHVFLRFamide Orcokinin Orcokinin Orcokinin Orcokinin Orcokinin Orcokinin Orcokinin Orcokinin Orcokinin Orcokinin Orcokinin Orcokinin Orcokinin Orcokinin Orcokinin Orcokinin Orcomyotropin FDAFTTGFGHS CPRP CPRP CPRP CPRP CPRP CPRP CPRP CPRP CPRP CPRP CPRP CPRP CPRP CPRP CPRP CPRP CPRP CPRP CPRP Sequence of PSM TNWNKFQGSWa EPLDHVFLR EPLDHVFLRF EPLDHVFLRFa YAIAGRPR pQDLDHVFLR pQDLDHVFLRF pQDLDHVFLRFa DFDEIDRSGFA EIDRSGFGFN FDEIDRSGFGFN NFDEIDR NFDEIDRS NFDEIDRSGFA NFDEIDRSGFG NFDEIDRSGFGF NFDEIDRSGFGFA NFDEIDRSGFGFN NFDEIDRSGFGFV NFDEIDRSSFG NFDEIDRTGFa NFDEIDRTGFG NFDEIDRTGFGF NFDEIDRTGFGFH FDAFTTGFGHS GSSRMERLL LLSSGSSSSEPLS LLSSGSSSSEPLSF LLSSGSSSSEPLSFL LLSSGSSSSEPLSFLSQ LLSSGSSSSEPLSFLSQDQSVN LLSSGSSSSEPLSFLSQDQSVS LSSGSSSSEPLSFL LSSGSSSSEPLSFLSQDQSVS MERLLSSGSSSSEPLSFLSQDQSVS PLSFLSQDQSVS RLLSSGSSSSEPL RLLSSGSSSSEPLS RLLSSGSSSSEPLSF RLLSSGSSSSEPLSFL RLLSSGSSSSEPLSFLSQDQSVS RSVEGSSRM(O)ERLLSSGSSSSEPLSFLSQDQSVS RSVEGSSRMERL RSVEGSSRMERLL 25.83 43.97 64.89 51.49 83.14 109.36 21.4 45.31 116.9 64.49 40.65 63.06 70.99 54.37 92.46 64.5 37.43 21.59 53.36 46.01 75.71 49.52 19.91 18.8 33.24 29.98 53.35 Eyestalk PEAKS Score (-10logP) 32.36 Appendix C. Neuropeptides and Proteins Identified in Orconectes rusticus 33.81 151.69 82.22 46.53 94.94 92.77 59.16 100.88 47.02 28.97 15.38 63.15 16.43 33.61 24.56 34.29 58.65 51.41 110.22 28.28 34.47 64.87 41.7 22.13 35.44 16.55 48.09 35.44 16.55 51.99 Mascot Score 27.52 38.98 51.82 58.31 75.5 48.84 76.44 69.98 16.85 23.52 29.92 45.87 30.71 32.87 Brain PEAKS Score (-10logP) 21.27 43.02 36.02 46.55 45.69 101.5 58.89 46.25 33.66 47.52 20.73 Mascot Score 419 CPRP CPRP CPRP CPRP CPRP CPRP CPRP CPRP CPRP CPRP CPRP CPRP PDH PDH PDH PDH PDH PDH PDH PDH PDH PDH PDH PDH PDH PDH PDH PDH PDH PDH PDH PDH PDH PDH PDH PDH PDH PDH PDH PDH PDH PDH SIFamide SIFamide SIFamide SIFamide SIFamide SIFamide GYRKPPFNGSIFa GYRKPPFNGSIFa GYRKPPFNGSIFa GYRKPPFNGSIFa GYRKPPFNGSIFa GYRKPPFNGSIFa RSVEGSSRMERLLSSGSSSSEPLSFLSQDQSVN RSVEGSSRMERLLSSGSSSSEPLSFLSQDQSVNa RSVEGSSRMERLLSSGSSSSEPLSFLSQDQSVS SEPLSFLSQDQSVS SGSSSSEPLSFL SGSSSSEPLSFLSQDQSVN SGSSSSEPLSFLSQDQSVS SSEPLSFLSQDQSVS SSGSSSSEPLSFLSQDHSV SSGSSSSEPLSFLSQDQSVS SSSEPLSFLSQDQSVS SSSSEPLSFLSQDQSVS AAQllRVAQGPSAFVAGPHK AGGlPHKRNa AQGPSAFVAGPHK DlNPTEKa EAVATlAAHllKVVCAPlEGAGGlPHKa EYllKFa lGTM(O)AGGPHKa llGlPKVM(O)T llGlPKVMNEAa MVAM(O)SlQlT NSEllNAllGSPTl NSEllNAllGSPTlM NSEllNSllGl NSEllNSllGlP NSEllNSllGlPK NSEllNSllGlPKVMNEAa pQElKYPEREVVAEl pQElKYPEREVVAElA pQElKYPEREVVAElAA pQElKYPEREVVAElAAQ SEllNSllGlPa SllGlPKVM(O) SllGlPKVMNEAa SlM(O)SEEVa SlTM(O)DVES SSlKYFEREVVSElAAQll SSlKYFEREVVSElAAQllR SSlKYFEREVVSElAAQllRV VAQGPSAFVAGPHK VVlAAVlTQG GYRKPPFN GYRKPPFNG GYRKPPFNGSIF GYRKPPFNGSIFa KPPFNGSIF KPPFNGSIFa 19.79 24.71 18.82 30.95 15.39 24.73 33.91 19.25 30.71 56.9 64.97 21.04 20.16 28.04 34.24 49.1 53.8 36.15 17.94 19.05 19.96 25.29 31.32 25.71 38.13 41.45 65.89 76.48 102.29 19.06 45.91 21.47 32.27 50.76 82.63 67 16.23 16.66 26.61 29.27 48.58 73.48 20.08 36.65 69.16 38.04 21.71 24.69 87.6 28.58 34.91 86.67 35.58 38.6 81.38 71.45 129.83 16.35 15.74 19.11 47.45 21.08 55.2 51.55 17 21.09 16.58 29.83 19.03 36.46 37.37 16.36 25.71 420 SIFamide SIFamide SIFamide SIFamide SIFamide SIFamide GYRKPPFNGSIFa GYRKPPFNGSIFa GYRKPPFNGSIFa GYRKPPFNGSIFa GYRKPPFNGSIFa GYRKPPFNGSIFa RKPPF RKPPFN RKPPFNG RKPPFNGSI RKPPFNGSIF RKPPFNGSIFa 30.91 39.46 38.42 27.45 21.18 23.42 23.7 33.66 21.41 17.23 421 CHH CHH CPRP CPRP CPRP CPRP Crustacean female hormone Crustacean female hormone Crustacean female hormone EH Eyestalk peptide Eyestalk peptide FMRFamide FMRFamide Insulin-like O15981.1 O97384.1 AAM21927.1 AAS46643.1 ABS01332.1 LL343 ADO00266.1 AFK81936.1 AF112986_1 AF112986_1 BAE06263.1 EFX80896.1 ADA67878.1 ADO00266.1 ADO00266.1 CHH LL206 NPs Found in O. rusticus, novel Accession Number Peptide Family ABX55995.1 Bursicon ACG50067.1 Bursicon ACG50067.1 Bursicon ACG50067.1 Bursicon ADI86242.1 Bursicon ACX46386.1 Calcitonin ACX46386.1 Calcitonin ABB46292.1 CCAP BAF34909.1 CCAP BAF34909.1 CCAP AAD45236.1 CHH ACN65120.1 CHH ACS35346.1 CHH AER27833.1 CHH AFD28272.1 CHH AFD28272.1 CHH AFG16934.1 CHH AFM29133.1 CHH crustacean female hormone [Callinectes sapidus] eclosion hormone [Amphibalanus amphitrite] eyestalk peptide [Jasus edwardsii] eyestalk peptide [Jasus edwardsii] FLRFamide precursor protein B [Procambarus clarkii] putative sulfakinin-like peptide [Daphnia pulex] insulin-like androgenic gland hormone precursor [Penaeus monodon] crustacean female hormone [Callinectes sapidus] crustacean female hormone [Callinectes sapidus] Description bursicon alpha [Carcinus maenas] bursicon hormone alpha subunit [Callinectes sapidus] bursicon hormone alpha subunit [Callinectes sapidus] bursicon hormone alpha subunit [Callinectes sapidus] bursicon alpha subunit [Homarus gammarus] prepro-calcitonin-like diuretic hormone [Homarus americanus] prepro-calcitonin-like diuretic hormone [Homarus americanus] crustacean cardioactive peptide [Homarus gammarus] crustacean cardioactive peptide [Procambarus clarkii] crustacean cardioactive peptide [Procambarus clarkii] hyperglycemic hormone-like neuropeptide 43-4 [Metapenaeus ensis] crustacean hyperglycemic hormone [Panulirus homarus] Crustacean hyperglycemic hormone isoform 1 [Rimicaris kairei] Crustacean hyperglycaemic hormone [Ptychognathus pusillus] CHH protein [Scylla paramamosain] CHH protein [Scylla paramamosain] Hyperglycemic hormone [Pandalopsis japonica] Crustacean hyperglycmic hormone [Portunus pelagicus] Crustacean hyperglycemic hormone Hoa-CHH-A (pCHH-A[pQ61V132amide]) [Homarus amerianus] Crustacean hyperglycemic hormones 5; Pej-SGP-V [Marsupenaeus japonicus] Crustacean hyperglycemic hormone 2; Pm-SGP-II [Penaeus monodon] Crustacean hyperglycemic hormone precursor [Pachygrapsus marmoratus] cCustacean hyperglycemic hormone precursor [Macrobrachium rosenbergii] Prepro-crustacean hyperglycemic hormone [Galathea strigosa] CHH precursor-related peptide Capr CPRP II [13-38] [Cancer productus] QGPLPIQPVa PGLASLGGSTST DVRQDGQTNa TPARQR PAEYSRNVR PVELGSSNPa VGMLMVLSLT ERQIQGP DGGYIDLSENa GDLPGGLVHamide RIEKLLST LSFMPEHP DRSLFGKL SSPVASLIRG LVAGASSAGT LDDLLLSDV PSM Sequence VTVLVVIG AM(O)VGAAVT M(O)VGAAVTV VLTRAPIDCMCRPCT TVVWSDECSLT LTRLGHSIIRANELEKFVRSSGSA TRLGHSIIRANELEKF TPHTQPR AGPLAKRDIG KLWEQLQ pEAASPGASSPWVEHR SPPPAGRTATVIGCSVNVSTTYa HPSGLAALTASH PAGHPLEKRQamide LVACIAMATLPQTQamide pEGAAHPLEKRamide EEHVDIR APM(O)QGYGTEamide 20.84 20.02 17.36 15.07 21.73 15.63 15.04 21.37 19.2 22.2 15.02 17.91 PEAKS Eyestalk 30.15 28.32 19.46 Mascot 37.72 19.38 19.84 15.44 17.93 18.55 16.77 18.19 19.29 18.05 16.15 22.81 21.51 15.38 18.99 PEAKS Brain 18.3 18.17 28.86 58.81 Mascot 422 Orcokinin Orcokinin Orcokinin Oxytocinneurophysin RPCH RPCH SIFamide-related peptide Tachykinin Tachykinin Tachykinin Q9NL83.1 Q9NL83.1 Q9NL83.2 Q867W1.1 BAC82426.1 BAC82426.1 BAC82426.1 ADD24046.1 ADN95181.1 ADQ73633.1 Insulin-like Insulin-like Insulin-like EFX70023.1 EFX76240.1 EFX76240.1 GYRKPPFNGSIF-amide; FRP1_PROCL [Procambarus clarkii] preprotachykinin [Procambarus clarkii] preprotachykinin [Procambarus clarkii] preprotachykinin [Procambarus clarkii] Oxytocin-neurophysin 1 [Lepeophtheirus salmonis] red pigment concentrating hormone precursor [Scylla paramamosain] red pigment concentrating hormone precursor [Scylla olivacea] Insulin/IGF/relaxin-like peptide 2 [Daphnia pulex] insulin/IGF/relaxin-like peptide 1 [Daphnia pulex] insulin/IGF/relaxin-like peptide 1 [Daphnia pulex] Orcomyotropin/orcokinin precursor-like peptide [Procambarus clarkii] Orcomyotropin/orcokinin precursor-like peptide [Procambarus clarkii] Orcomyotropin/orcokinin precursor-like peptide [Procambarus clarkii] AGGDSLYEPGK AGMDSELETLL HFDDESEIDAY HFDDESEIDAYIQAL QYKQCSSCGP SVGGAPGGVVPSSPGSSSGDSa PPGSSSGDSCGP VYVPRYIANLY VYVPRYIAN MTAQM(O)FTIALLLSLS SLGLTTTAATPNa M(O)IIPSTVGRCWM(O) VTPAEP 51.8 17.89 38.47 77.24 38.23 18.39 21.23 45.72 22.85 18.11 18.54 18.49 45.89 16.8 37.25 87.4 29.9 22.28 22.03 423 EGFAPHTTFGT E(-18.01)LGDAPDVAAEKAR EGFAPHTTCST DPSGSTLAQR DSYGYHLDR DSYGYHLDR E(-18.01)EEVAAVG DPSGSTLAQR DGFAPHTTFGT DLPVSAAPVVESAPVVES DPSGLELAGA DPSGLPLAAA DPSGLPLAAA DPSGLPLAAA DPSGLPLAQ DPSGLPLAQ DPSGLPLAQ DPSGPALAKAA DPSGPALANR DGAPAAEDVKAPES DGAPAAEDVKAPES APVLGYWNLR ATLGVGAE AVGFLPALNLT APVFGYWNLR ALVGPSGLLLDDGTPVQFK ADVPDLGA AGFAGDDANGP(-.98) ADVPDLAA AALPLGNKEKTGA AAPPPVPTPEPT Peptide Best BLAST E-value XP_001951442 predicted serine/threonine protein kindase MAKlike Acyrthosiphon pisum 27 EFX85740 hypothetical protein dappudraft_309057 0.046 ACO14744.1 general transcription factor IIF subunit 3 Caligus clemensi 6.1 ACO15766.1 U3 small nucleolar ribonucleoprotein protein IMP4 Caligus clemensi 6.1 ACT82962.1 beta-actin Penaeus monodon 0.28 ABM54460.1 cuticle protein CUT 9 Portunus pelagicus 2.00E-08 ADV59553.1 glutathione S-transferase mu Parcyclopina nana 0.069 ADV59553.1 glutathione S-transferase mu Parcyclopina nana 0.069 ACO15187.1 tetraspanin-13 Caligus clemensi 4.4 EFX61408.1 hypothetical protein dappudrapt_339726 2.3 ACO12558.1 tissue factor pathway nhibitor precursor Lepeophtheirus salmonis 0.86 ACO12558.1 tissue factor pathway nhibitor precursor Lepeophtheirus salmonis 0.86 ABV58636.1 hemocyanin-like protein Metapenaeus ensis 0.03 EFX64340.1 hypothetical protein dappudraft_305068 0.006 EFX60846.1 hypothetical protein dappudraft_341180 9.5 EFX78346.1hypothetical protein dappudraft_246411 3.7 EFX78346.1hypothetical protein dappudraft_246411 3.7 EFX78346.1hypothetical protein dappudraft_246411 3.7 EFX64858.1 hypothetical protein dappudraft_65872 6.8 EFX64858.1 hypothetical protein dappudraft_65872 6.8 EFX64858.1 hypothetical protein dappudraft_65872 6.8 EFD86972.1 hypothetical protein dappudraft_187411 15 EFX75855.1 hypothetical protein dappudraft_322949 18 P83180.1 hemocyanin B chain Pontastacus leptodactylus 10 P83180.1 hemocyanin B chain Pontastacus leptodactylus 10 hemocyanin (multiple) 2.00E-05 hemocyanin (multiple) 2.00E-05 EFX81277.1 hypothetical protein dappudraft_102637 3.1 ABB9165.1 calcified cuticle protein CP19.0 isoform B Callinectes sapidus, others 2.00E-04 ACS44711.1 hemocyanin Porcellio scaber 0.058 ACS44713.1 hemocyanin subunit 2 or ACS44712.1 hemocyanin subunit 1 Eurydice pulchra 0.017 De Novo Sequenced Peptides from O. rusticus, from PEAKS Software 70 7 9.6 6.7 68 61 94 76 6.9 7.5 74 61 53 79 60 75 7.4 6.1 4.3 14.9 7.8 9.1 8 73 73 68 73 8 6.5 6.1 78 67 59 60 80 56 77 56 72 7.1 7.4 10.6 6 8 6.1 8.5 4.5 9.3 7.7 8.6 6.3 7.2 70 78 63 79 67 55 90 85 72 80 66 58 76 5.5 9 8.5 7.2 7.2 5.9 5.3 8.4 6.7 63 68 75 53 61 59 61 78 67 7 9.5 10.5 5.9 6.1 5.9 4.8 8.5 5.4 9.7 7.4 7 6.3 11.7 5.9 4.5 6 14.9 4.1 7.1 69 92 77 63 65 59 56 60 78 52 59 ALC ALC TLC_brain (%)_brain ALC TLC_brain (%)_brain ALC 1 1 TLC_eye1 (%)_eye1 2 2 TLC_eye2 (%)_eye2 424 AABZ79135.1 arginine kinase Procambarus clarkii EFX82438.1 hypothetical protein dappudraft_30262 EFX86228.1 hypothetical protein dappudraft_308434 EFX66745.1 hypothetical protein dappudraft_116065 ACV73257.1 cytochrome C oxidase subunit I (Cyzicus spp WRH-2009) or ACV73163.1 same Limnadia lenticularus ACO15009.1 torsin-1B precursor Caligus clemensi EFX79110.1 hypothetical protein dappudraft_245307 ABI34072.1 ATP/ADP translocase Pacifastacus leniusculus or AEZ68611.1 adenine nucleotide translocase Litopenaeus vannamei ADD38275.1 probable pyruvate dehydrogenase E1 component subunit alpha, others P05547.1 troponin I Pontastacus leptodactylus P05547.1 troponin I Pontastacus leptodactylus EFX59854.1 hypothetical protein dappudraft_126524, others BAE94239.1 aryl hydrocarbone receptor nuclear translocator, daphnia magna, others EFX76176.1 hypothetical protein dappudraft_107260 EFX60320.1 hypothetical protein dappudraft_278059 EFX85893.1 hypothetical protein dappudraft_98546 AEB54632.1 arg kinase Procambarus clarkii AEB54632.1 arg kinase Procambarus clarkii AEB54632.1 arg kinase Procambarus clarkii AEB54632.1 arg kinase Procambarus clarkii AEB54632.1 arg kinase Procambarus clarkii AEB54632.1 arg kinase Procambarus clarkii ABX44762.3 cytosolic manganese superoxide dismutase Procambarus clarkii EFX74602.1 hypothetical protein dappudraft_251748 EFX66222.1 hypothetical protein dappudraft_263488 EFX82754.1 hypothetical protein dappudraft_101360 EFX82754.1 hypothetical protein dappudraft_101361 ABQ41245.1 male reproductive-related LIM protein Macrobrachium rosenbergii EFX90150.1 hypothetical protein dappudraft_299958 EFX78266.1 hypothetical protein dappudraft_305250 EFX70769.1 hypothetical protein dappudraft_112349 ACP19741.1 thymosin-repeated protein 2 Eriocheir sinensis and others CAR64036.1 NADH dehydrogenase subunit 1 Catroptrus nitidus EFX77232.1 hypothetical protein dappudraft_305825 EFX76032.1 hypothetical protein dappudraft_322770 EMQDGLLELLKLEQEM EVTPVVAA EVVTTKLTKK FAGVGLATRHGT QLLVKVVNH QTDTPNVAAVT(-.98) RLPDNGV QGLEGFSPDKGG(-.98) LSMDTGSHLEEKK PHYGMST Q(-17.03)LGYGGFQQPVGSYPR QGAPDTPNADTAA LDLNAYLAALEKKLAELSG LEGDLPDLPKVE LETPNKL LGLGGGFGR LGLGGGFGR HLYDHLH LAEFEHFDKF LDAAASPAPAK LDAAASPAPAQKATK LDAATLAAGLEEGF LDAATLAGALEEGFK LDAATLAGALEEGFK LDAATLAGALEEGFK LDAATLAGALEEGFK LDAATLAKLE GSADLPGFGSV(-.98) FNHYGMST GDAPAAEDVKAPES GDAPAAEDVKAPES FLAGGLAAANLT FAGVGLHVGAT FAGVGLVGAT FDDDFLTSPT ABM54471.1 cryptocyanin Portunus pelagicus EGFSPQTTCST 1 8.7 19 0.002 1.00E-04 10 0.059 7.3 7.00E-10 0.22 4.2 1.9 2.9 0.9 1.5 0.87 0.5 0.62 0.075 1.075 2.075 3.075 0.33 9 1.3 1.2 1.2 0.99 1.2 14 0.47 6.00E-07 1.8 10 6.5 0.5 5 8.8 55 80 55 64 10.2 6.6 81 65 73 71 70 57 9.1 11 10.7 10.5 8.6 10.5 81 73 8.9 10.2 80 88 59 74 5.9 8.9 8.8 8.8 64 10.9 4 10.2 5.3 58 79 75 70 57 53 62 6.2 13.3 6.8 3.7 72 68 59 68 77 78 74 67 61 60 76 10.8 10.2 4.1 6.8 8.5 10.9 10.4 8.1 6.1 4.8 8.3 5.2 8.1 5.7 14.1 7.9 4 4.8 4.5 9.2 11.1 4 6.6 5 10 8.8 7 4.3 7.4 58 62 81 74 66 57 53 51 66 74 58 66 63 72 73 70 54 68 7.5 6.2 10.8 11 6.3 9 10.9 6.6 6.1 8.5 9.7 8.9 5.2 8.1 11.2 69 52 67 84 63 82 73 60 76 53 88 89 52 68 66 425 YGGYFPRSPDNK WSGPGHAPAAGGW(-.98) VSGGSGFGHGGGFPNYGGAW(.98) NYGGGGYGRY(-.98) WPSGLVLSNGQNVQYRT VVLPVTLQGA WPNTPEDNAAYLERN(-.98) VKAPGLLPR VKVVNH VSSTLSGMSLPLE(-.98) TVSALPDPPVAA TEEASLKDLPKVDT TLGDTPEVAAEQAR TLSPTVVGP TDVPDLAA SSGGFAAASGP(-.98) STAGFVTRP TDLPDLAA TDPVVGPSGLLDPTGK TDPVVGPSGLLDPTGK TDPVVGPSGLLTPTGK TDPVVGPSGLLTPTGK TDPVVGPSGLLTPTGK SPVGALGLLLDDGTPVQF SEGPSGLVLSNGQNVQYRT RPTDQNLLPSSTGAA RLPDNGV 0.007 1.2 0.025 0.11 EFX73534.1 hypothetical protein dappudraft_325069 EFX83421.1 hypothetical protein dappudraft_315888 2.00E-04 0.84 0.55 2 31 0.62 1.9 1.00E-04 0.32 4.6 6.1 6.5 12 4.5 0.39 1.39 2.39 3.39 4.39 0.003 1.00E-06 0.099 20 EFX76032.1 hypothetical protein dappudraft_322771 AAY89318.1 glyceraldehyde-3-phosphate dehydrogenase Tigriopus japonicus ABM54457.1 cuticle protein CUT2 Portunus pelagicus, other spp ABM54460.1 cuticle protein CUT 9 Portunus pelagicus or ABM54459.1 cuticle protein CUT8 Portunus pelagicus ABQ59095.1 or ABQ59.96.1 chitinase Homarus americanus EFX62683.1 hypothetical protein dappudraft_336600 EFX87331.1 hypothetical protein dappudraft_97181 EFX70090.1 hypothetical protein dappudraft_300586 EFX70090.1 hypothetical protein dappudraft_300587 EFX70090.1 hypothetical protein dappudraft_300588 EFX70090.1 hypothetical protein dappudraft_300589 EFX70090.1 hypothetical protein dappudraft_300590 ACO14744.1 general transcription factor IIF subunit 3 Caligus clemensi AEL23126.1 thymosin-repeated protein 1 Cherax quadricarinatus P81583.1 cuticle protein CP1499 Cancer pagurus EFX86349.1 hypothetical protein dappudraft_299671 EFX62924.1 or EFX67775.1 hypothetical protein dappudraft_336202 or 330715 EFX70812.1 hypotherical proteind appudraft_309281 or ADC55251.1 mitochondrial ATP synthase alpha precursor Litopenaeus vannamei 3 diff hypothetical daphia prots ACB46937.1 arg kinase Eurtium limosum and others YP_003084316.1 cytochrome c oxidase subunit II Metacrangonyx longipes EFX71970.1 hypothetical protein dappudraft_308613 ABM54457.1 cuticle protein CUT2 Portunus pelagicus ABM74407.1 hemocyanin Portunus pelagicus and other spp EFX90425.1 hypothetical protein dappudraft_30013 10.2 5.4 7.9 60 54 61 61 68 56 73 69 80 79 78 11.8 11 12.9 12.7 12.4 8.6 9.5 5.1 64 65 60 70 53 7.1 5.8 10.7 13.4 7.9 10.7 5.9 4.3 6.7 4.7 5 9.1 4 71 65 72 56 58 62 61 57 7.7 10.3 5.3 3.4 7 5.4 4.4 64 69 59 56 58 68 55 10.6 5.4 8.2 6.6 9.3 5.2 7.9 9.6 9 5.6 6.7 5.2 9.5 12.1 53 54 68 51 55 52 61 68 64 63 60 58 53 63 426 Brain 1 beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| Protein Description 14-3-3 [Portunus trituberculatus] 14-3-3 protein [Scylla paramamosain] 14-3-3 zeta [Artemia franciscana] 14-3-3 zeta [Fenneropenaeus merguiensis] 14-3-3 zeta [Fenneropenaeus merguiensis] 14-3-3 zeta [Scylla paramamosain] 14-3-3 zeta protein [Artemia franciscana] 14-3-3-like protein [Penaeus monodon] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] Protein Accession gi|401816688|gb|AFQ20813.1| gi|380003174|gb|AFD28274.1| gi|161898814|gb|ABX80390.1| gi|298570899|gb|ADI87601.1| gi|298570899|gb|ADI87601.1| gi|380042038|gb|AFD33362.1| gi|282895616|gb|ADB03180.1| gi|66774602|gb|AAY56092.1| Proteins found in O. rusticus, Mascot 285 285 285 285 285 285 285 285 285 285 285 285 285 285 285 285 285 140 140 140 140 140 140 140 140 140 Protein Mascot Score 56 56 56 56 56 56 56 56 68.37 63.32 57.19 55.38 48.21 47.45 40.83 39.74 36.31 35.9 34.01 30.46 30.11 29.9 29.58 29.46 27.85 17.18 17.78 20.62 28.53 30.08 30.79 42.88 55.38 66.14 Peptide Mascot Score 55.65 55.65 55.65 55.65 15.58 55.65 55.65 55.65 QVFRRLTSAVNE AGFAGDDAPRAVFPSI VG AGFAGDDAPR TEAPLNPKANR TEAPLNPKANR LTEAPLNPK AGFAGDDAPRAVF TEAPLNPK HQGVMVGMGQ GDDAPRAVFPSIVG FAGDDAPR EAPLNPKANR TEAPLNPK FAGDDAPRAVFPSIVG DDAPRAVFPSIVG APLNPK AGFAGDDAPRAVFPS DEAQSKRGILT FAPLFDPIIEDY FAPLFDPIIED LFAPLFDPIIED APLFDPIIED DVIQSGVENLDSGVG ATLLDVIQSGVENL ATLLDVIQSGVENLDS GVG EVQQKLIDDHFLFK Peptide Sequence ASNPESKVFYLK ASNPESKVFYLK ASNPESKVFYLK ASNPESKVFYLK CTLIMQLLR ASNPESKVFYLK ASNPESKVFYLK ASNPESKVFYLK 2 Oxidation (M) Oxidation (M) Peptide Variable Modifications 0.0000400400.0 0.000040000.0 Position of Variable Modifications 427 beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Procambarus clarkii] beta-actin [Procambarus clarkii] beta-actin [Procambarus clarkii] beta-actin [Procambarus clarkii] beta-actin [Procambarus clarkii] beta-actin [Procambarus clarkii] beta-actin [Procambarus clarkii] beta-actin [Procambarus clarkii] beta-actin [Procambarus clarkii] beta-actin [Procambarus clarkii] beta-actin [Procambarus clarkii] Chain A, 1.2a Resolution Structure Of A Crayfish Trypsin Complexed With A Peptide Inhibitor, Sgti Chain A, 1.2a Resolution Structure Of A Crayfish Trypsin Complexed With A Peptide Inhibitor, Sgti Chain A, X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A, X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A, X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A, X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A, X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A, X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A, X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A, X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A, X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A, X-Ray Structure And Catalytic gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|211947526|gb|ACJ13518.1| gi|211947526|gb|ACJ13518.1| gi|211947526|gb|ACJ13518.1| gi|211947526|gb|ACJ13518.1| gi|211947526|gb|ACJ13518.1| gi|211947526|gb|ACJ13518.1| gi|211947526|gb|ACJ13518.1| gi|211947526|gb|ACJ13518.1| gi|211947526|gb|ACJ13518.1| gi|211947526|gb|ACJ13518.1| gi|211947526|gb|ACJ13518.1| gi|402715413|pdb|1PDZ|A gi|402715413|pdb|1PDZ|A gi|402715413|pdb|1PDZ|A gi|402715413|pdb|1PDZ|A gi|402715413|pdb|1PDZ|A gi|402715413|pdb|1PDZ|A gi|402715413|pdb|1PDZ|A gi|402715413|pdb|1PDZ|A gi|402715413|pdb|1PDZ|A gi|402715413|pdb|1PDZ|A gi|99032198|pdb|2F91|A gi|99032198|pdb|2F91|A beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| 152 152 152 152 152 152 152 152 152 152 42 42 167 167 285 285 285 285 285 167 167 167 167 167 167 167 167 167 285 285 285 285 20.17 18.13 24.13 27.58 30.55 32.79 33.99 34.3 67.64 79.96 23.7 44.11 17.65 18.63 17.65 16.81 16.68 15.87 15.69 57.19 55.38 48.21 45.31 40.83 34.01 30.46 29.58 24.19 18.63 26.31 24.44 21.18 EVILPVPAF NVINGGSHAGNK MGTEVYHHLK NVINGGSHAGNKLA EVILPVP EVILPVPAFNVINGGS HAGNKLA EVILPVPAF DEGGFAPNILN AGAAELGIPLYR DEGGFAPNILNNK AGELDMSVNEGS IVGGTDATLGEFPYQL EAPLNPK EAPLN EAPLNPK LKYPIE DEAQSKRGILT AGFAGDDAPRA LPHAILR TEAPLNPKANR TEAPLNPKANR LTEAPLNPK DSGDGVSHTVPIYEG TEAPLNPK EAPLNPKANR TEAPLNPK APLNPK VSHTVPIYEG EAPLN FAGDDAPRAVFPS RGILTLK LDLAGR Oxidation (M) Amidated (Cterm) Amidated (Cterm) Glu->pyro-Glu (N-term E) Amidated (Cterm) Glu->pyro-Glu (N-term E) 0.4000000000.0 0.000000000000.1 3.0000000.0 0.00000.1 3.0000000.0 0.00000.1 428 gi|310743896|dbj|BAJ23879.1| gi|310743896|dbj|BAJ23879.1| gi|310743896|dbj|BAJ23879.1| gi|310743896|dbj|BAJ23879.1| gi|310743896|dbj|BAJ23879.1| gi|310743896|dbj|BAJ23879.1| gi|310743896|dbj|BAJ23879.1| gi|310743896|dbj|BAJ23879.1| gi|310743896|dbj|BAJ23879.1| gi|62701385|dbj|BAD95643.1| gi|62701385|dbj|BAD95643.1| gi|62701385|dbj|BAD95643.1| gi|62701385|dbj|BAD95643.1| gi|62701385|dbj|BAD95643.1| gi|62701385|dbj|BAD95643.1| gi|62701385|dbj|BAD95643.1| gi|62701385|dbj|BAD95643.1| gi|62701385|dbj|BAD95643.1| gi|62701385|dbj|BAD95643.1| gi|62701385|dbj|BAD95643.1| gi|225709118|gb|ACO10405.1| gi|225709118|gb|ACO10405.1| gi|290784917|dbj|BAI79321.2| gi|402715413|pdb|1PDZ|A duplex-specific nuclease [Chionoecetes opilio] GDP-fucose protein O-fucosyltransferase 1 precursor [Caligus rogercresseyi] GDP-fucose protein O-fucosyltransferase 1 precursor [Caligus rogercresseyi] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] Mechanism Of Lobster Enolase Chain A, X-Ray Structure And Catalytic Mechanism Of Lobster Enolase 28 28 28 28 28 28 28 28 28 106 106 106 106 106 106 106 106 106 106 106 17 17 20 152 15.32 15.42 15.83 17.88 18.07 18.65 20.12 22.08 23.07 17.61 18.97 22.05 26.08 26.58 28.62 32.31 32.92 48.72 49.96 60.57 15.25 17.24 19.84 17.87 KSPNNFNLKFFN FVPRTVMIGGK LPAPHEP VLYPNDNFFEGK GANIE VVHLDQL VDIEGLPWAKAW QEYFMVA AMPYDNPIPGYK TVDGPSAKDWR DGPSAKDWR TVDGPSAKDWR TVDGPSAKDWR GGRGAAQNIIPSS AQNIIP GGRGAAQNIIPSSTG GGRGAAQNIIPS GPSAKDWR TVDGPSAKDWR GPSAKDWR HMDLII LPNAPF MIKAYSQSSQLV DEGGFAPNIL Oxidation (M) Amidated (Cterm) Oxidation (M) Oxidation (M) Oxidation (M) Amidated (Cterm) Amidated (Cterm) 0.000000000000.1 0.00000040000.0 0.0000400.0 0.040000000000.0 0.040000.0 0.000000.1 0.000000000000.1 429 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] 51.9 49.76 52.47 53.05 54.12 54.65 55.05 56.84 56.91 57.6 59.56 60.73 61.17 67.99 68.81 69.31 71.47 72.37 77.53 78.13 79.09 83.5 95.24 102.96 108.07 VFKVQNQHGQVVK VSFPQLIHDADEAVA DGFAPHTTYK FAPHTTYK FNMPPGVMEHFET EVTPHLFTNSEVIDQA YAAK YGGEFPARPDNK FNMPPGVMEHFET DTDVAHQQQAINR DTDVAHQQQAINRL EVTPHLFTNSEVIDQ NEHGIDILGDIIE VQNQHGQVVKIFHH DEAVANGAELPHK YDAERLSNFLPAVDE L DTDVAHQQQAINR DAERLSNFLPAVDEL NEHGIDILGDIIESS DTDVAHQQQAINRLL DTDVAHQQQAINRLL EVTPHLFTNSEVIDQA YAAK DTDVAHQQQAINRLL YKVT DTDVAHQQQAINRLL YK DTDVAHQQQAINRLL YK VSFPQLIHDADEAVA NGAELPHKESR 2 Oxidation (M) Amidated (Cterm) 0.0040000400000.0 0.000000000000000.1 430 AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] 30.83 30.88 30.98 31.41 31.54 32.5 34.02 35.01 35.24 35.59 38.54 38.59 38.68 39.24 39.8 40.24 40.42 41.36 42.35 44.23 44.6 45.01 45.39 47.05 KFNMPPGVMEHFET VFKVQNQHGQVVK DEAVANGAELPH LPPLYEVTPHLF SSESSVAIPDRVSFPQL VQNQHGQVVKIFHH SSESSVAIPDR VSFPQLIHDADEAVA NGAELPH SSESSVAIPDRVSFPQ YGGEFPARPDNKEF EAAETWNPRDHTDK GAELPHKESR LFTNSEVIDQ LIHDADEAVANGAEL PH AETWNPRDHTDK EAAETWNPRDHTDK VSFPQLIHDADEAVA N SSESSVAIPDR EAAETWNPR DTDVAHQQQAINRLL ALHNQAHRVLG NEHGIDILGDIIESS MTQTPGNFK VMEHFETATR NGAELPHK 2 Oxidation (M) Amidated (Cterm) Oxidation (M) Oxidation (M) 0.00040000400000.0 0.0000000000.1 0.400000000.0 0.0400000000.0 431 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 1345 hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] 17.9 17.85 18.55 19.28 19.82 19.94 20.13 21.22 21.39 21.76 21.82 22.29 22.95 24.32 24.35 26.46 26.53 26.6 27.23 27.46 27.88 28.55 28.79 29.83 30.62 YGGEFPARPD NEHGIDILGDIIES MPPGVMEHFET DADEAVANGAELPH ATRDPAFFRLHK TWNPRDHTDK NEHGIDILGDIIES APHTTYK ATRDPAFFRLH KEAAETWNPRDHTD K EVTPHLF KEAAETWNPRDHTD K ATRDPAFFR EAAETWNPRDHTDK DTDVAHQQQAINRLL Y LLLPKGK DTDVAHQQQAINRLL Y LSNFLPAVDEL GFPFDRPIPDLRV VTPHLFTNSEVIDQ MPPGVMEHFETATR EVTPHLFTNSE ATRDPAF ETWNPRDHTDK DPAFFR 2 Oxidation (M) 2 Oxidation (M) Amidated (Cterm) 0.40000400000.0 0.00000000000000.1 0.40000400000000.0 432 gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 1345 1345 1345 1345 1345 1345 1345 1345 1345 hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] 116 116 116 116 116 116 116 116 116 116 116 116 116 116 1345 hemocyanin [Pacifastacus leniusculus] 15.85 16.51 17.31 20.45 21.4 25.04 27.82 28.41 32.01 33.31 35.51 36.75 46.57 47.93 15.15 15.41 15.69 16.24 17.06 17.18 17.56 17.59 17.72 17.84 LPNRFLLP EVIHEAYKA KAIDE LPNRFLLPK TAHIMLGR QYPDKRPHGYPLDR EVTPHMFTNSE TRDPSFFR LPPGVLEHF DPSFFR NTAHIMLGR EALPN NTAHIMLGR DPSFFRLH RPDNKEF EVTPHLFT IDILGD DGFAPHTTYKYG EVTPHLF KPFSLNIH GFAPHTTYK VSFPQLIHDA WAHHQLTAR PHTTYK Amidated (Cterm) Oxidation (M) Glu->pyro-Glu (N-term E) Oxidation (M) Oxidation (M) Amidated (Cterm) Amidated (Cterm) Glu->pyro-Glu (N-term E) Glu->pyro-Glu (N-term E) 0.000000000.1 0.00004000.0 3.00000000000.0 0.000004000.0 0.00000.1 0.000004000.0 3.00000000.0 0.000000.1 3.0000000.0 433 I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 116 116 116 17.93 17.89 17.59 18.15 18.13 18.06 18.05 17.94 18.22 18.69 18.68 18.83 18.8 20.8 20.75 19.98 19.47 19.13 19.1 21.04 20.96 23.37 22.07 23.59 23.58 26.02 24.07 23.65 36.04 28.26 26.84 15.22 15.55 15.83 RKPKTEK LTTRLAGEVE KEEQATSIISEHQSLTV PDTRETIQ KPNSLQVIDVT QENGLAH KVPEQV DFIIE AEGERCHLE EKIEEVSEE KPFKTDKAKEPESVS DKPDIQKAE HLDIP KDDKPKE APVDIKE EDGTEALTLPYAVL IPQKSVNVS QFKLKPKTKKKVK VTVKEIPTDI KMKPKKKP AVVPRGE KPKEKVVEEI KILDKPSQP KAKKVMPQQESI KPGKKPSKKI QIKFKPGK KKPETQQ FEARLVPT KPFEKPKP PKPER DEEKPKPEE ALELDRF KYGGYFPSRPDN DPSFFRL 0.0000000.1 0.000000000.1 0.000000000.1 Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) 0.0000000.1 0.04000000.1 Amidated (Cterm); Oxidation (M) Amidated (Cterm) 0.0000000000.1 Amidated (Cterm) 0.00000000.1 0.000000000000.1 Amidated (Cterm) 0.000000000.1 0.00000000.1 Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) 0.00000000.1 Amidated (Cterm) 434 16 16 16 I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] inhibitor of apoptosis protein [Penaeus monodon] inhibitor of apoptosis protein [Penaeus monodon] inhibitor of apoptosis protein [Penaeus gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|133754273|gb|ABO38431.1| gi|133754273|gb|ABO38431.1| gi|133754273|gb|ABO38431.1| 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 25 I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| 25 25 I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| 17.58 16.13 17.87 15.1 15.16 15.12 15.12 15.41 15.29 15.42 15.42 15.44 15.65 15.5 15.48 15.69 15.67 15.65 15.7 15.89 15.89 16.6 16.36 15.92 16.68 17.25 17.23 17 16.79 17.56 17.31 IRMCKVCMD ETRQTLQADPIESEA KDLDPL EFKET EHAEVI KRPPM IPKTRQ EKLDESDKK EPEVEKRV AKAIIADDVKDS QTSLL VIKPEE EKEEVIEEV ITQAPRFVVK VRLIPTSDP IDIVEG EVVVETVA SDPFMR QTRTVREVQQGV GIILEP KAPTEEKFE KKRILIVKS PEEEGP KPSEVQP EYEARAVNK QQQQSMSQQ PEEIPEKE HNETTGVVT VQYDQNGASCLQIL EAKPV QSQVP 3.000000.0 0.00004.0 Glu->pyro-Glu (N-term E) Oxidation (M) Glu->pyro-Glu (N-term E) Amidated (Cterm) Amidated (Cterm); 2 Oxidation (M) Glu->pyro-Glu 0.000000000.1 0.004000040.1 3.000000000000000.0 0.000000.1 3.00000.0 0.000000000000.1 2.00000.0 0.000000.1 0.000000000.1 0.000040.0 Amidated (Cterm) Gln->pyro-Glu (N-term Q) Amidated (Cterm) Amidated (Cterm) Oxidation (M) Amidated (Cterm) 0.000000.1 0.000000000000.1 0.000000.1 Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) 0.000000000.1 3.000000000.0 0.000004000.1 Amidated (Cterm); Oxidation (M) Glu->pyro-Glu (N-term E) Amidated (Cterm) 0.00000.1 Amidated (Cterm) 435 Brain 2 ABC protein, subfamily ABCH [Daphnia pulex] ABC protein, subfamily ABCH [Daphnia pulex] ABC protein, subfamily ABCH [Daphnia pulex] ABC protein, subfamily ABCH [Daphnia pulex] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] gi|321467478|gb|EFX78468.1| gi|321467477|gb|EFX78467.1| gi|321467477|gb|EFX78467.1| gi|321467477|gb|EFX78467.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| Protein Description 14-3-3 protein [Scylla paramamosain] 14-3-3 protein [Scylla paramamosain] 14-3-3 zeta [Artemia franciscana] 14-3-3 zeta [Artemia franciscana] 14-3-3 zeta [Fenneropenaeus merguiensis] 14-3-3 zeta [Fenneropenaeus merguiensis] 14-3-3 zeta [Scylla paramamosain] 14-3-3 zeta [Scylla paramamosain] 14-3-3-like protein [Penaeus monodon] 14-3-3-like protein [Penaeus monodon] ABC protein, subfamily ABCH [Daphnia pulex] putative DH31 receptor [Daphnia pulex] putative DH31 receptor [Daphnia pulex] RecName: Full=NAD-dependent protein deacylase; AltName: Full=Regulatory protein SIR2 homolog 5; Flags: Precursor ubiquitin carboxyl-terminal esterase L3 [Scylla paramamosain] ubiquitin carboxyl-terminal esterase L3 [Scylla paramamosain] ubiquitin carboxyl-terminal esterase L3 [Scylla paramamosain] ubiquitin carboxyl-terminal esterase L3 [Scylla paramamosain] ubiquitin carboxyl-terminal esterase L3 [Scylla paramamosain] ubiquitin carboxyl-terminal esterase L3 [Scylla paramamosain] Protein Accession # gi|380003174|gb|AFD28274.1| gi|380003174|gb|AFD28274.1| gi|161898814|gb|ABX80390.1| gi|161898814|gb|ABX80390.1| gi|298570899|gb|ADI87601.1| gi|298570899|gb|ADI87601.1| gi|380042038|gb|AFD33362.1| gi|380042038|gb|AFD33362.1| gi|66774602|gb|AAY56092.1| gi|66774602|gb|AAY56092.1| gi|321467478|gb|EFX78468.1| gi|225908473|gb|ACO36738.1| gi|225908473|gb|ACO36738.1| gi|225908473|gb|ACO36738.1| gi|225908473|gb|ACO36738.1| gi|225908473|gb|ACO36738.1| gi|225908473|gb|ACO36738.1| gi|387935383|sp|E9GD30.1|SIR 5_DAPPU gi|321458499|gb|EFX69566.1| gi|321458499|gb|EFX69566.1| gi|83638451|gb|ABC33915.1| gi|133754273|gb|ABO38431.1| monodon] inhibitor of apoptosis protein [Penaeus monodon] kazal-type proteinase inhibitor [Fenneropenaeus chinensis] 97 97 97 31 17 31 31 39 Protein Mascot Score 106 106 106 106 106 106 106 106 106 106 17 39 39 39 39 39 43 32 32 24 16 27.67 52.96 70.36 15.04 17.46 31.05 22.81 15.34 Peptide Mascot Score 66.45 64.1 66.45 64.1 66.45 64.1 66.45 64.1 66.45 64.1 22.81 15.89 17.92 22.6 23.86 30.44 43.47 31.97 16.08 23.85 15.24 ASVHIKLP ATLLDVIQSGVENL NTTIVQVPF ATLLDVIQSGVENLDS GVG DLPKLAAPSSQ FLLPAA IPGPRVGYMP Peptide Sequence ASNPESKVFYLK ASNPESKVFYLK ASNPESKVFYLK ASNPESKVFYLK ASNPESKVFYLK ASNPESKVFYLK ASNPESKVFYLK ASNPESKVFYLK ASNPESKVFYLK ASNPESKVFYLK IPGPRVGYMP VMNKFMAGLGVPES VRWLPLESNPAVMNK EEQEAAGGDVPE VRWLPLE VRWLPLESNPAVMNK VRWLPLESNPAVMNK F IAEFEHRMT QSLKCTR LPAVVT KHDGPCAAKVI PSTSGAQA Oxidation (M) Amidated (Cterm) Oxidation (M) Amidated (Cterm) Oxidation (M) Peptide Variable Modifications Oxidation (M) Oxidation (M) Oxidation (M) Oxidation (M) Amidated (Cterm) (N-term E) 0.000000000.1 0.0000000040.0 0.00000000000.1 0.0000000040.0 Position of Variable Modifications 0.00000400000000.0 0.000000000000400.0 0.0000000000004000.0 0.000000000000400.0 0.000000040.0 0.0000000.1 436 caspase 1, partial [Daphnia parvula] caspase 5, partial [Daphnia pulex] caspase 5, partial [Daphnia pulex] gi|321478601|gb|EFX89558.1| gi|385048432|gb|AFI39977.1| gi|385048432|gb|AFI39977.1| gi|385048432|gb|AFI39977.1| gi|385048432|gb|AFI39977.1| gi|385048472|gb|AFI39997.1| gi|385048472|gb|AFI39997.1| 21 21 15 23 15 15 15 Btk family kinase at 29A-like protein [Daphnia pulex] caspase 1, partial [Daphnia parvula] caspase 1, partial [Daphnia parvula] caspase 1, partial [Daphnia parvula] gi|321478601|gb|EFX89558.1| 281 281 127 127 127 127 127 127 127 127 97 281 281 281 281 281 281 281 281 281 281 281 281 281 281 281 281 281 281 281 281 281 281 23 arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Procambarus clarkii] beta-actin [Procambarus clarkii] beta-actin [Procambarus clarkii] beta-actin [Procambarus clarkii] beta-actin [Procambarus clarkii] beta-actin [Procambarus clarkii] beta-actin [Procambarus clarkii] beta-actin [Procambarus clarkii] Btk family kinase at 29A-like protein [Daphnia pulex] gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|211947526|gb|ACJ13518.1| gi|211947526|gb|ACJ13518.1| gi|211947526|gb|ACJ13518.1| gi|211947526|gb|ACJ13518.1| gi|211947526|gb|ACJ13518.1| gi|211947526|gb|ACJ13518.1| gi|211947526|gb|ACJ13518.1| gi|211947526|gb|ACJ13518.1| gi|551380|gb|AAB31477.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| 21.58 20.87 15.39 18.89 22.5 19.53 15.74 23.18 17.14 16.48 72.03 48.24 44.53 37.2 34.87 24.06 19.89 17.78 27.4 72.03 66.87 57.89 48.24 47.87 43.42 42.27 41.74 37.2 35.54 34.87 34.43 32.21 27.21 26.61 25.84 24.06 22.77 22.62 19.89 17.78 17.33 KMMKEGTM LDAGTTMLPP LDAGTTMLPP LPNVTET QGDRLDAGTTMLPKV TETDS DLLNKLINVLNDLQF N DLLNKLINVLNDLQF SIPTEPAVVP RGILTLK FAGDDAPR TEAPLNPKANR EAPLNPKANR VSHTVPIYEG APLNPKANR TEAPLNPK EAPLNPK TEAPLNPK TEAPLNPK LFAPLFDPIIED TEAPLNPKANR AGFAGDDAPR HQGVMVGMGQK EAPLNPKANR AGFAGDDAPR FAGDDAPR AGFAGDDAPR AGFAGDDAPR APLNPKANR DAPRAVFPSIVG TEAPLNPK AGFAGDDAPRAVFPS GFALPHAILR AGFAGDDAPRA DEAQSKRGILT FAGDDAPRAVFP EAPLNPK LVVDNGSGMVK VVDNGSGMVK TEAPLNPK TEAPLNPK LKYPIE Amidated (Cterm) Amidated (Cterm) Amidated (C- Amidated (Cterm); Oxidation (M) Oxidation (M) Oxidation (M) Amidated (Cterm) 0.0000000000000000.1 0.00000000000000000. 0.000000000000000000 00.1 0.00000004.1 0.0000004000.0 0.0000004000.0 0.0000000.1 437 caspase 5, partial [Daphnia pulex] caspase 5, partial [Daphnia pulex] caspase 5, partial [Daphnia pulex] caspase 5, partial [Daphnia pulex] caspase 5, partial [Daphnia pulex] Chain A, X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A, X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A, X-Ray Structure And Catalytic Mechanism Of Lobster Enolase gi|385048478|gb|AFI40000.1| gi|385048478|gb|AFI40000.1| gi|385048464|gb|AFI39993.1| gi|385048464|gb|AFI39993.1| gi|385048464|gb|AFI39993.1| gi|310743896|dbj|BAJ23879.1| gi|310743896|dbj|BAJ23879.1| gi|310743896|dbj|BAJ23879.1| gi|310743896|dbj|BAJ23879.1| gi|310743896|dbj|BAJ23879.1| gi|310743896|dbj|BAJ23879.1| gi|310743900|dbj|BAJ23881.1| gi|310743900|dbj|BAJ23881.1| gi|402715413|pdb|1PDZ|A gi|402715413|pdb|1PDZ|A gi|402715413|pdb|1PDZ|A gi|402715413|pdb|1PDZ|A gi|402715413|pdb|1PDZ|A gi|402715413|pdb|1PDZ|A gi|402715413|pdb|1PDZ|A fructose 1,6-bisphosphatase [Marsupenaeus japonicus] fructose 1,6-bisphosphatase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] Chain A, X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A, X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A, X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A, X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A, X-Ray Structure And Catalytic Mechanism Of Lobster Enolase caspase 5, partial [Daphnia pulex] gi|385048478|gb|AFI40000.1| gi|402715413|pdb|1PDZ|A caspase 5, partial [Daphnia pulex] gi|385048472|gb|AFI39997.1| 29 29 29 29 29 29 28 28 97 97 97 97 97 97 97 97 21 21 21 21 21 21 21 15.02 16.03 18.5 22.31 23.39 29.54 16.77 28.02 16.68 18.54 19.1 19.18 27.67 28.58 41.71 82.06 20.48 20.87 21.58 20.48 20.87 21.58 20.48 SLAIPEL LPAPHEP ILPRHLQ TNGITPR RDIFKDF NNVVNTMRL KNNEAVPSVSDALQ MAGDVNVQ SITKVFART LAKYNQILRIEEELGS GA KDFPI PIVSIEDPFDQ MILPTGA DEGGFAPNILN DEGGFAPNILNNK DEGGFAPNILNNK KV DLLNKLINVLNDLQF KV DLLNKLINVLNDLQF N DLLNKLINVLNDLQF KV DLLNKLINVLNDLQF KV DLLNKLINVLNDLQF N DLLNKLINVLNDLQF KV DLLNKLINVLNDLQF KV Amidated (Cterm) Amidated (Cterm); Oxidation (M) Amidated (Cterm); Oxidation (M) term) Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) 0.0000000.1 0.40000000.1 0.4000000.1 0.0000000000000000.1 0.00000000000000000. 1 0.00000000000000000. 1 0.0000000000000000.1 0.00000000000000000. 1 0.00000000000000000. 1 1 0.00000000000000000. 1 438 25 994 994 994 994 994 994 994 994 994 994 994 994 994 994 994 994 994 994 994 994 994 994 glycogen synthase [Marsupenaeus japonicus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] gi|310743898|dbj|BAJ23880.1| gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| 25 25 glycogen synthase [Marsupenaeus japonicus] glycogen synthase [Marsupenaeus japonicus] gi|310743898|dbj|BAJ23880.1| gi|310743898|dbj|BAJ23880.1| 42.2 41.86 42.99 44.14 44.31 44.47 45.69 47.65 54.3 56.62 58.85 60.11 61 62.14 62.87 68.02 70.13 72.32 73.5 74.67 77.72 83.48 15.9 25.11 18.49 VMEHFETATR EAAETWNPRDHTDK ATRDPAFFRLH DTDVAHQQQAINR VFKVQNQHGQVVK SSESSVAIPDR RPMGFPFDRPIPDLR AETWNPRDHTDK DTDVAHQQQAINRLL DTDVAHQQQAINRLL DTDVAHQQQAINR VFKVQNQHGQVVKIF HH DTDVAHQQQAINRLL Y SSESSVAIPDRVSFPQ VQNQHGQVVKIFHH EAAETWNPRDHTDK FNMPPGVMEHFETAT RDPAF DTDVAHQQQAINR LIHDADEAVANGAEL PHK KEAAETWNPRDHTD K KEAAETWNPRDHTD K DTDVAHQQQAINRLL Y EKINDFIRG EASVRMEVE LTTAINNIR 0.000004000.1 0.000000000.1 Amidated (Cterm); Oxidation (M) Amidated (Cterm) 439 AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 994 994 994 994 994 994 994 994 994 994 994 994 994 994 994 994 994 994 994 994 994 994 994 994 hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] 24.52 24.61 24.73 24.99 25.03 25.13 25.86 29.74 29.74 30.42 30.72 31.14 31.94 32.88 33.17 33.89 33.95 35.32 38.56 39.81 39.94 40.06 40.86 41.66 LLLPKGK EVTPHLFTNS LIHDADEAVANGAEL PHK MEFTGSKKNR GEFPARPDNK ATRDPAFFRLHK DAERLSNFLPAVDEL NEHGIDILGDIIES PFDRPIPDLR FPQLIHDADEAVAN EAAETWNPR AERLSNFLPAVDEL EVTPHLFTNSEVID DPAFFRLH LSNFLPAVDEL AETWNPRDHTDK ETWNPRDHTDK KFNMPPGVMEHFETA TRDPAF KEAAETWNPRDHTD K EVTPHLFTNSE EAAETWNPRDHTDK MPPGVMEHFET TDLKEAAETWNPRDH TDK EAAETWNPRDHTDK Glu->pyro-Glu (N-term E) Glu->pyro-Glu (N-term E) 3.0000000000.0 3.00000000000000.0 440 gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 994 994 994 994 994 994 994 994 994 994 994 994 994 994 994 994 994 994 994 994 hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus 110 110 110 110 994 hemocyanin [Pacifastacus leniusculus] 34.79 34.2 37.6 42.73 15.08 15.44 15.73 15.86 16.04 16.63 16.98 18.3 18.32 19.07 19.08 19.26 19.49 20.21 20.71 21.31 21.53 21.53 21.82 23.01 23.92 YMDNIFR LPPGVLEHFET EVTPHMFTNSE LHNTAHIMLGR GFPFDRP VTPHLFT PFDRPIPDLRV EVTPHLF AAETWNPRDHTDK QVVKIFHH VSFPQLIHDADEAVA NGAELPH VFKVQNQHGQVVK EVTPHLFTNSEVID VFKVQNQHGQVVKIF HH VSFPQLIHDADEAVA NGAELPHK LFTNSEVIDQ RPMGFPFDRPIPDLRV DVAHQQQAINR NQHGQVVKIFHH QNQHGQVVKIFHH LLLPKGK YDAERLSNFLPAVDE L AERLSNFLPAVDEL FNMPPGVMEHFET VSFPQLIHDADEAVA N Glu->pyro-Glu (N-term E) Glu->pyro-Glu (N-term E) Amidated (Cterm) 3.00000000000.0 3.0000000.0 0.00000000000000.1 441 I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] 33 33 33 33 33 33 33 33 33 33 33 33 33 33 33 33 33 33 33 110 110 110 110 110 110 110 110 17.76 17.47 18.2 18.8 18.95 18.81 19.09 19.07 19.18 21.61 21.35 20.63 21.89 21.73 23.23 23.37 24.5 27.11 33.95 16.58 17.89 23.75 25.63 26.61 28.07 29.51 32.17 EYEARAVNK PLQDPNPLPE AVSIHE EKLDESDKK DKPDIQKAE KDEVE KPFEKPKP HNETTGVVT KEIPEVKE PEVPKPVE LMVSSKES HNETTGVVT KEEVKEEIILK IKAPLWRVKKLP EKLDESDKK QIKFKPGK APSAPFRPH LLLPQDSIAV KPFEKPKP KYGGYFPSRPDN QYPDKRPHGYPLDR YMDNIFREHK GYPLDR QYPDKRPHGYPLDR NTAHIMLGR LPNRFLLPK LPPGVLEHF 0.00000000.1 0.04000000.0 Amidated (Cterm) Oxidation (M) 3.000000000.0 0.0000000000.1 0.000000.1 0.000000000.1 0.000000000.1 Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) Glu->pyro-Glu (N-term E) Amidated (C- 0.00000000.1 0.00000000.1 Amidated (Cterm) Amidated (Cterm) 0.000000000.1 0.00000000.1 0.000000000.1 0.0000000000.1 0.00000000000.1 0.00000000.1 0.000004000.0 Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) Oxidation (M) 442 I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] JHE-like carboxylesterase 1 [Pandalopsis gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|326579691|gb|ADZ96217.1| 33 21 33 33 33 33 33 33 33 33 33 33 33 33 33 33 33 33 33 33 33 33 33 33 33 33 33 33 33 15.25 21.26 15.31 15.48 15.36 15.61 15.6 15.61 15.67 15.73 15.91 15.88 15.99 16.01 16.38 16.19 16.12 16.09 16.45 16.54 16.75 16.58 16.56 16.96 17 17.09 17.16 17.36 17.3 KHEDIEE MKLLFLCVAIATWLG KIEEVTEEVTIKKP KPEDEE TKIAQLE QEVTVKGVPAD EKIKELEDT ADVAPSIAPLE GIILEP KPGKKP TLLIIEAVPEDSA RVEPRN QNKLQEKI QRVSVIVS MQQMVSPKF QCRAVGTPTP TVQEVAVKE AGQYTVVAR KPTKIIEPE EGLEKIRQLE QGRAGLEKVE QENGLAH KRPPM QLEEPVKP QATSVISPLE QPDILPGI KDEVEPH NKTGEA KPSEVQP 0.000000.1 Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) Amidated (C- 0.00000000000.1 0.0000000.1 0.0000000000000000.1 0.00000000000000.1 0.00000000000.1 0.000000.1 0.000000.1 0.0000000000000.1 0.00000000.1 0.00000000.1 0.000400000.1 0.000000000.1 0.0000000000.1 0.00004.0 0.0000000000.1 0.00000000.1 2.0000000000.0 0.00000000.1 0.0000000.1 Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) Oxidation (M) Amidated (Cterm) Amidated (Cterm) Amidated (Cterm); Oxidation (M) Amidated (Cterm) Amidated (Cterm) Gln->pyro-Glu (N-term Q) Amidated (Cterm) Amidated (Cterm) term) 443 Eyestal k1 phantom [Marsupenaeus japonicus] putative G-protein coupled receptor, orphan [Daphnia pulex] putative G-protein coupled receptor, orphan [Daphnia pulex] RecName: Full=NAD-dependent protein deacylase; AltName: Full=Regulatory protein SIR2 homolog 5; Flags: Precursor RecName: Full=NAD-dependent protein deacylase; AltName: Full=Regulatory protein SIR2 homolog 5; Flags: Precursor ubiquitin carboxyl-terminal esterase L3 [Scylla paramamosain] vitellogenin fused with superoxide dismutase [Daphnia magna] vitellogenin fused with superoxide dismutase [Daphnia magna] vitellogenin fused with superoxide dismutase [Daphnia magna] vitellogenin fused with superoxide dismutase [Daphnia magna] vitellogenin fused with superoxide dismutase [Daphnia pulex] vitellogenin fused with superoxide dismutase [Daphnia pulex] vitellogenin fused with superoxide dismutase [Daphnia pulex] vitellogenin fused with superoxide dismutase [Daphnia pulex] vitellogenin fused with superoxide dismutase [Daphnia pulex] vitellogenin fused with superoxide dismutase [Daphnia pulex] gi|224471277|dbj|BAH24005.1| gi|224471277|dbj|BAH24005.1| gi|46398235|gb|AAS91795.1| gi|225712206|gb|ACO11949.1| Protein Accession # gi|321465377|gb|EFX76379.1| gi|321465377|gb|EFX76379.1| gi|321465377|gb|EFX76379.1| gi|321465377|gb|EFX76379.1| gi|321465377|gb|EFX76379.1| gi|321465377|gb|EFX76379.1| gi|95113632|dbj|BAE94322.1| gi|95113632|dbj|BAE94322.1| gi|95113632|dbj|BAE94322.1| gi|95113632|dbj|BAE94322.1| gi|225908473|gb|ACO36738.1| gi|387935383|sp|E9GD30.1|SIR 5_DAPPU gi|387935383|sp|E9GD30.1|SIR 5_DAPPU gi|321477804|gb|EFX88762.1| gi|321477804|gb|EFX88762.1| Protein Description Serine/threonine-protein phosphatase 2A regulatory subunit B [Lepeophtheirus salmonis] intestinal trypsin 4 precursor [Lepeophtheirus salmonis] phantom [Marsupenaeus japonicus] gi|225713512|gb|ACO12602.1| gi|326579691|gb|ADZ96217.1| japonica] JHE-like carboxylesterase 1 [Pandalopsis japonica] Peptidyl-tRNA hydrolase 2, mitochondrial precursor [Lepeophtheirus salmonis] 24 26 28 Protein Mascot Score 28 28 28 28 28 25 25 25 25 55 43 43 37 37 19 19 20 21 23.58 25.59 15.54 Peptide Mascot Score 16.15 17.32 20.25 21.2 27.98 17.03 20.25 21.2 25.21 54.57 22.28 43.58 28.78 40.82 17.79 18.88 20.5 17.03 EFTPTGE VDEIPLEEQSQ Peptide Sequence EVKNTANQIT VGEQAVQAISALVP VRIIGCTSAEG GARAQETVEKKLG QRIKSI MEGSVTGECET KIVTVLL GARAQETVEKKLG QRIKSI ASPSTIRQ VRWLPLESNPAVM IAEFEHRMT IAEFEHRMT HSIQITSTGGD AALLDRL LLLALLR VVGIGPGPLE APHLTLTNLVEKYGR VYSLKMGGVSVVVIA DPDLI KDVILVV V Amidated (Cterm); Glu- Oxidation (M) Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) Glu->pyro-Glu (N-term E) Peptide Variable Modifications Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) Oxidation (M) Oxidation (M) Amidated (Cterm) Amidated (Cterm) term) Amidated (Cterm) 3.0000000.1 Position of Variable Modifications 3.0000000000.0 0.00000000000000.1 0.00000000000.1 0.0000000000000.1 0.000000.1 0.40000000000.0 0.0000000.1 0.0000000000000.1 0.000000.1 0.00000000.1 0.000000040.0 0.000000040.0 0.00000000000.1 0.0000000.1 0.0000000.1 444 I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| 110 110 110 110 110 110 110 110 110 110 110 110 110 110 110 110 110 110 110 110 110 110 110 110 110 110 110 110 110 110 110 110 15.88 15.83 16.4 16.35 16.16 16.02 15.88 17.15 16.94 16.91 17.38 17.29 17.22 17.83 17.73 18.82 18.71 18.05 19.53 18.94 19.55 21.35 20 19.97 23.54 22.55 22.22 21.65 21.42 30.07 64.48 78.22 VVLQSQVPK EGMPSI IEDKAKTVLSQRVS KSVKRVNQK VTSASVKVT LEPLKPLK DKYLMTQ QHIRSVEEM SQIPK KTEQAKSILSPRE LPAPIEE QENGLAH SDPFMR ERTKPEKEE ISQDTVQVQEI KEAPYSMEED KDSITVTEKIP ISQDTVQVQEI LEAQVQPA QLEMDVRLVAIP KSPLE KPFEKPKP LEPLKPLK AAPIEEKF KAKKVMPQQESI KPRFLTKLK LSQNMH QMQIQVAA TVQEVSVKEAP KRILIVKSV IIEPEPEPEPELIILRPV E IIEPEPEPEPELIILRPV ER 0.0000004000.1 0.000000000.1 0.0000000.1 Amidated (Cterm) Amidated (Cterm) 0.000000000.1 3.000000.0 0.0000400.0 0.00000000000000.1 Amidated (Cterm) Oxidation (M) Amidated (Cterm) Glu->pyro-Glu 2.000000004.0 0.000040.0 Oxidation (M) Gln->pyro-Glu (N-term Q); Oxidation (M) 0.00000000.1 0.000400000000.0 0.00000.1 0.00000000.1 Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) Oxidation (M) Amidated (Cterm); Oxidation (M) 0.000000000000.1 0.000000000.1 Amidated (Cterm) Amidated (Cterm) >pyro-Glu (Nterm E) 445 110 110 510 510 510 510 510 510 510 510 510 510 510 510 510 510 510 510 510 510 510 510 510 I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 110 110 110 I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| 18.68 19.66 23.45 25.81 26.17 28.21 30.52 37.63 38.39 42.19 42.62 45.16 46.67 48.06 61.82 62.06 67.74 68.3 83.43 99.5 107.57 15.12 15.04 15.8 15.63 15.31 RPMGFPFDRPIPDLR DLKELESR EVTPHL LLLPKGK PMGFPFDRPIPDL VFKVQNQHGQVV DEAVANGAELPHK VFKVQNQHGQVVK RPMGFPFDRPIPDLR YGGEFPARPDN DGFAPHTTYK VFKVQNQHGQVVK DTDVAHQQQAIN EAAETWNPR DTDVAHQQQAINR YGGEFPARPDNK DTDVAHQQQAINR DTDVAHQQQAINRLL YK SVAIPDRVSFPQLIHD ADEAVAN SSESSVAIPDRVSFPQL IH DTDVAHQQQAINR QEVSVKEAPTE AAPIEEKFEIPKITLK AGEKI GESQYEDTVLKMQY DAVVI Oxidation (M) Amidated (Cterm) (N-term E) 0.004000000000000.0 0.00000000000.1 446 510 510 hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] ecdysteroid receptor [Carcinus maenas] ecdysteroid receptor [Carcinus maenas] Dpahnia pulex DNMT3 [Daphnia pulex] Dpahnia pulex DNMT3 [Daphnia pulex] Dpahnia pulex DNMT3 [Daphnia pulex] Dpahnia pulex DNMT3 [Daphnia pulex] crustin-like peptide type 5 [Marsupenaeus japonicus] crustin-like peptide type 5 [Marsupenaeus japonicus] crustin-like peptide type 4 [Marsupenaeus japonicus] crustin-like peptide type 4 [Marsupenaeus japonicus] crustin-like peptide type 2 [Marsupenaeus japonicus] crustin-like peptide type 2 [Marsupenaeus japonicus] crustin-like peptide [Marsupenaeus japonicus] crustin-like peptide [Marsupenaeus japonicus] chymotrypsin-like protein [Daphnia pulex] chymotrypsin-like protein [Daphnia pulex] Chain A, X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A, X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A, X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A, X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A, The 2.1a Crystal Structure Of Copgfp Chain A, The 2.1a Crystal Structure Of Copgfp caspase 6, partial [Daphnia pulex] gi|62701385|dbj|BAD95643.1| gi|40748295|gb|AAR89628.1| gi|40748295|gb|AAR89628.1| gi|321467881|gb|EFX78869.1| gi|321467881|gb|EFX78869.1| gi|321467881|gb|EFX78869.1| gi|321467881|gb|EFX78869.1| gi|402715413|pdb|1PDZ|A gi|114793939|pdb|2G3O|A gi|114793939|pdb|2G3O|A gi|385048340|gb|AFI39931.1| gi|402715413|pdb|1PDZ|A gi|402715413|pdb|1PDZ|A gi|402715413|pdb|1PDZ|A gi|46240812|dbj|BAD15063.1| gi|46240810|dbj|BAD15062.1| gi|46240810|dbj|BAD15062.1| gi|321468614|gb|EFX79598.1| gi|321468614|gb|EFX79598.1| gi|46240812|dbj|BAD15063.1| gi|46240816|dbj|BAD15065.1| gi|46240816|dbj|BAD15065.1| gi|46240818|dbj|BAD15066.1| gi|46240818|dbj|BAD15066.1| gi|62701385|dbj|BAD95643.1| 510 hemocyanin [Pacifastacus leniusculus] 62 20 20 25 62 62 62 34 34 34 21 21 34 34 34 34 34 38 38 27 27 38 38 25 25 510 hemocyanin [Pacifastacus leniusculus] gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 19.58 21.94 17.24 24.72 19.72 39.53 47.01 19.03 33.72 19.03 21.28 18.9 33.72 19.03 33.72 19.03 33.72 17.43 19.12 26.99 16.1 38.1 23.26 24.93 26 15.42 16.22 17.6 17.63 IEEELGSGA MFFYHFGT SHMHFKSAIHPSI QSKATQTDGP DDLTVTNPK FAGKNFR GNPTVEVDLYTSK GFGGVQGGGV GGGFGGGFGGPQGGG GFGGVQGGGV QSISPVLMKVT SELFPRA GGGFGGGFGGPQGGG GFGGVQGGGV GGGFGGGFGGPQGGG GFGGVQGGGV GGGFGGGFGGPQGGG EADGLL FTDVGNMSRTD TVDGPSAKDWR DYPSHGSPIAEEKAIPT SPM HITEMTILTVQLI SLFDGIGT SLFDGIGT GGRGAAQNIIPSS NMPPGV FNMPPGVMEHFETAT RDPAFF NEHGIDILGDIIE AETWNPR Oxidation (M) Amidated (Cterm); Oxidation (M) Glu->pyro-Glu (N-term E) Oxidation (M) Oxidation (M) Oxidation (M) Oxidation (M) 0.00000004000.0 3.000000.0 0.00000040000.1 0.000000000000000000 04.0 0.0000400000000.0 0.040000.0 0.004000000000000000 000.0 447 caspase 3, partial [Daphnia pulex] caspase 3, partial [Daphnia pulex] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus gi|385048368|gb|AFI39945.1| gi|385048368|gb|AFI39945.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| caspase 6, partial [Daphnia pulex] caspase 3, partial [Daphnia pulex] gi|385048340|gb|AFI39931.1| gi|385048368|gb|AFI39945.1| 477 477 477 477 477 477 477 477 477 477 477 477 477 477 477 477 477 477 477 59 59 23 59 59 59 23 25 23 19.87 19.51 27.92 28.57 28.83 29.7 33.62 34.68 35.69 40.05 42.86 43.92 44.03 65 71.62 76.95 94.4 95.08 116.08 25.33 25.07 16.23 38.3 35.75 25.56 19.19 16.29 23.04 MGLTEFQAVK QTDKHPNKDF GTYFPLTGMSK LIDDHFLFK RMGLTEFQAVK ATLLDVIQSGVENLDS GVGIYAPDA ASVHIKLPK RMGLTEFQAV ATLLDVIQSGVENLDS GVGIYAPDAEAY RMGLTEFQAVK LFDPIIEDYH FDPIIEDYH ATLLDVIQSGVENL ATLLDVIQSGVENLD LIDDHFLFKEGDR APLFDPIIEDYH PFSSXXCATLAGKP AVFPSIVGR DAYVGDEAQSK FAGDDAPRA AGFAGDDAPRAVFPSI VG HQGVMVGMGQ ATLLDVIQSGVENLDS GVGIYAPDA ATLLDVIQSGVENLDS GVG EVQQKLIDDHFLFKE GDR TLAGKP EIKTIVNDL SNSST Oxidation (M) Gln->pyro-Glu Oxidation (M) 2 Oxidation (M) Amidated (Cterm) Amidated (Cterm) 0.4000000000.0 2.0000000000.0 0.04000000000.0 0.0000400400.0 0.00000000000000.1 0.000000.1 448 Eyestal k2 ABC protein, subfamily ABCH [Daphnia pulex] ABC protein, subfamily ABCH [Daphnia pulex] ABC protein, subfamily ABCH [Daphnia pulex] ABC protein, subfamily ABCH [Daphnia pulex] ABC protein, subfamily ABCH [Daphnia pulex] ABC protein, subfamily ABCH [Daphnia pulex] gi|551380|gb|AAB31477.1| gi|321473807|gb|EFX84774.1| gi|321473807|gb|EFX84774.1| gi|321473807|gb|EFX84774.1| gi|321473807|gb|EFX84774.1| gi|321473807|gb|EFX84774.1| gi|321473807|gb|EFX84774.1| gi|321473807|gb|EFX84774.1| Protein Description I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] Protein Accession # gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] ABC protein, subfamily ABCH [Daphnia pulex] 37 37 37 37 37 37 37 37 37 37 37 37 37 37 37 37 37 37 37 28 28 Protein Mascot Score 28 28 28 28 477 28 477 477 20.26 19.82 18.69 20.42 20.28 22.34 21.71 21.44 21.44 21.42 22.58 25.41 26.39 26.27 26.27 25.86 28.33 26.79 32.88 16.46 16.1 Peptide Mascot Score 17.07 17.78 21.13 19.33 15.3 28.33 15.51 18.05 IELTYE KNLSNLEEGEIA AAPIEEKF KITPPSKPNSL KEEITEEVIIKKPE EKIEEVSEE KILDKPSQP LEVSETIVL MSGEPTKPKF KIVGKTATLTVNAQ LERAKPN KPITIGSRFKTY LEAQVQPA IVIKPEE IVIKPEE IVIKPEE KPFEKPKP PEREKVPDQVPKPEK E AAPIEEKFEIPK Peptide Sequence QANAIGLMRSGRL GASGCGKTTLLSCLV G NGIDMSVKKGT QANAIGLMRSGRL QQMELNLQH VEFITPGIILT FAPLFDPIIE ATLLDVIQSGVENLDS GVGIYAPDAEAY VEFITPGIILT DDHFLFK 0.00000000.1 Amidated (Cterm) 0.000000000.1 0.4000000000.0 0.00000000000.1 0.000000.1 Oxidation (M) Amidated (Cterm) Amidated (Cterm) 0.0000000.1 0.000000000000.1 Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) 0.00000000.1 Position of Variable Modifications 0.0000000000000000.1 0.00004000000.0 0.0000000000000.1 0.0000000000000.1 2.004000000.0 Oxidation (M) Peptide Variable Modifications Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) Gln->pyro-Glu (N-term Q); Oxidation (M) (N-term Q) 449 I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|249511|gb|AAB22190.1| gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| gi|249511|gb|AAB22190.1| 32 32 32 32 32 37 37 37 37 37 37 37 37 37 37 37 37 37 37 37 586 586 I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| 37 hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] I-connectin [Procambarus clarkii] gi|15425681|dbj|BAB64297.1| 37 32 I-connectin [Procambarus clarkii] gi|15425681|dbj|BAB64297.1| 37 37 37 37 37 hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster, Peptide, 661 aa] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|249511|gb|AAB22190.1| I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| gi|15425681|dbj|BAB64297.1| 103.33 83.53 17.36 19.76 20.09 29.39 30.96 35.29 15.05 15.61 15.39 16.04 16.02 16.01 15.78 15.62 16.18 17.4 17.2 16.83 16.69 16.63 16.23 17.67 17.76 17.83 17.77 18.19 18.09 17.89 DTDVAHQQQAINR SSESSVAIPDRVSFPQL QYPDKRPHGYPLDR LPKGQAQG VVLPPL LMMHRVLMN YGGYFPSRPDN DPSFFR QGVQRVQGPTQ QQAPHEAVEP LGLSDALEVS KPEEKIE PEEKVVEV ILDKPSQP DAEPPAFNPPLTP LEEHYP EEKVKVTPKP QMINQGIETEE KIAEEVTEEIVVKKA KTEQAKSILSPRE ENEEFLDEFIET REWSPVP KLKKSTRMLEPKEP KITPPSKPNSL LEVSSELAPD KAKKVMPQQESI SVHEISVKDAP KSIISPHESLTVE EEVKPKTEQ VTIKQE 2.00000000000.1 Amidated (Cterm); Gln>pyro-Glu (Nterm Q) 0.040000040.1 2.0000000000.0 Gln->pyro-Glu (N-term Q) Amidated (Cterm); 2 Oxidation (M) 0.0000000.1 0.0000000000.1 0.04000000000.1 0.00000000000.1 0.0000000000.1 0.000000000000.1 Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) Amidated (Cterm) Amidated (Cterm); Oxidation (M) Amidated (Cterm) 450 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 AF522504_1 586 586 586 586 586 586 586 586 586 586 586 586 586 586 586 586 586 586 586 586 586 586 hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] 586 hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] 28.96 29.92 29.97 30.37 30.88 32.87 32.93 33.04 33.55 34.05 35.21 37.06 37.5 39.14 41.65 43.36 44.18 44.97 52.96 53.58 62.05 64.41 83.49 MGFPFDRPIPDL VFKVQNQHGQVVKIF HH VSFPQLIHDA DPAFFR RPMGFPFDRPIPDLR YGGEFPARPDNK VFKVQNQHGQVVK DTDVAHQQQAINRLL YK RPMGFPFDRPIPDL VSFPQLIHDADEAVA NGAELPH DGFAPHTTYK RPMGFPFDRPIPDL DTDVAHQQQAIN FNMPPGVMEHFETAT RDPAFF YPDKRPMGFPFDRPIP DLR FNMPPGVMEHFE KFNMPPGVMEHFETA TRDPAFFR EAAETWNPR YGGEFPARPDNK DTDVAHQQQAINRLL YK FNMPPGVMEHFETAT RDPAFF IHDADEAVANGAELP HK FNMPPGVMEHFETAT RDPAFFR SSESSVAIPDRVSFPQL IHDADEAVANGAELP HKESR Oxidation (M) 0.004000000000000000 000.0 451 586 586 586 586 586 586 586 586 586 586 586 586 586 586 hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] Dpahnia pulex DNMT3 [Daphnia pulex] Dpahnia pulex DNMT3 [Daphnia pulex] Dpahnia pulex DNMT3 [Daphnia pulex] Chain A, X-Ray Structure And Catalytic Mechanism Of Lobster Enolase gi|62701385|dbj|BAD95643.1| gi|321467881|gb|EFX78869.1| gi|321467881|gb|EFX78869.1| gi|321467881|gb|EFX78869.1| gi|402715413|pdb|1PDZ|A gi|62701385|dbj|BAD95643.1| gi|62701385|dbj|BAD95643.1| 21 42 42 22 42 22 22 22 586 hemocyanin [Pacifastacus leniusculus] gi|62701385|dbj|BAD95643.1| 586 hemocyanin [Pacifastacus leniusculus] gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 gi|21914372|gb|AAM81357.1| AF522504_1 21 19.21 21.93 15.28 41.93 19.65 22.31 29.19 16.68 16.9 19.76 19.96 20.09 20.63 21.49 21.55 22.33 22.79 22.85 23.17 23.3 23.61 28.09 28.26 AGAAELGIPLYR FTDVGNMSRTD FTDVGNMSRTD HKGEVKAEDGC SLFDGIGT DGPSAKDWR TVDGPSAKDWR QGGAKKVI LPKGKEQ DTDVAHQQQAINRLL Y FNMPPGVMEHFETAT RDPAFFR VVLPPL FNMPPGVMEHFETAT RDPAFF DLKELESR TATRDPAFF YGGEFPARPDNKEF VQNQHGQVVKIFHH FNMPPGVMEHFETAT R SSESSVAIPDRVSFPQL IHDA VSFPQLIHDADEAVA NGAELPHKESR YPDKRPMGFPFDRPIP DL PMGFPFDRPIPDL YGGEFPARPD SSESSVAIPDRVSFPQL IH Amidated (Cterm); Oxidation (M) Amidated (Cterm); Oxidation (M) Amidated (Cterm) Oxidation (M) Amidated (Cterm) 2 Oxidation (M) Amidated (Cterm) 0.00000040000.1 0.00000040000.1 0.00000000000.1 0.00000000.1 0.000000040000000000 0000.0 0.0000000.1 0.004000040000000000 000.0 452 Chain A, X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A, X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A, X-Ray Structure And Catalytic Mechanism Of Lobster Enolase caspase 5, partial [Daphnia pulex] caspase 4, partial [Daphnia parvula] caspase 4, partial [Daphnia parvula] caspase 4, partial [Daphnia parvula] caspase 4, partial [Daphnia parvula] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus gi|402715413|pdb|1PDZ|A gi|385048476|gb|AFI39999.1| gi|385048398|gb|AFI39960.1| gi|385048412|gb|AFI39967.1| gi|385048398|gb|AFI39960.1| gi|385048412|gb|AFI39967.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|254832574|gb|ACT82962.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|402715413|pdb|1PDZ|A gi|402715413|pdb|1PDZ|A 454 454 454 454 454 454 454 454 454 454 454 77 15 77 77 77 77 77 77 77 15 15 15 21 27 21 21 42.85 40.67 45.1 46.82 49.71 51.69 56.59 67.67 76.88 92.39 98.71 16.82 15.41 50.65 45.14 34.38 30.51 28.48 18.14 17.81 15.41 16.74 16.74 15.08 27.29 15.59 17.79 DPIIEDYH ATLLDVIQSGVENL ATLLDVIQSGVENLD ASVHIKLPK LIDDHFLFKEGDR LIDDHFLFKEGDR ATLLDVIQSGVENLDS GVGIYAPDA EVQQKLIDDHFLFK EVQQKLIDDHFLFK APLFDPIIEDYH LRVAPEE EVQQKLIDDHFLFKE GDR DCFMVV HQGVMVGMGQK FAGDDAPR AVFPSIVGR GFAGDDAPR AGFAGDDAPR AGFAGDDAPR AGFAGDDAPRA DCFMVV DCFMVV DCFMVV SKMTSGTTIQIVG IANVTVSNF DDLTVTNPK DMYMEFCKDFPIVSIE DPFD 0.0000000.1 Amidated (Cterm) 0.000400.1 0.000400.1 0.000400.1 0.000400.1 0.040400000000000000 00.0 Amidated (Cterm); Oxidation (M) Amidated (Cterm); Oxidation (M) Amidated (Cterm); Oxidation (M) Amidated (Cterm); Oxidation (M) 2 Oxidation (M) 453 gi|225714196|gb|ACO12944.1| gi|225714196|gb|ACO12944.1| gi|225714196|gb|ACO12944.1| gi|148613137|gb|ABQ96193.1| gi|321467477|gb|EFX78467.1| gi|290562041|gb|ADD38417.1| gi|290562041|gb|ADD38417.1| gi|148613137|gb|ABQ96193.1| gi|148613141|gb|ABQ96195.1| gi|148613141|gb|ABQ96195.1| gi|148613143|gb|ABQ96196.1| gi|148613143|gb|ABQ96196.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| gi|551380|gb|AAB31477.1| japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] arginine kinase, AK [Penaeus japonicus=shrimps, tail muscle, Peptide, 355 aa] anti-lipopolysaccharide factor isoform 4 [Farfantepenaeus paulensis] anti-lipopolysaccharide factor isoform 4 [Farfantepenaeus paulensis] anti-lipopolysaccharide factor isoform 3 [Farfantepenaeus paulensis] anti-lipopolysaccharide factor isoform 3 [Farfantepenaeus paulensis] anti-lipopolysaccharide factor isoform 1 [Farfantepenaeus paulensis] anti-lipopolysaccharide factor isoform 1 [Farfantepenaeus paulensis] ABC protein, subfamily ABCH [Daphnia pulex] 14-3-3 protein zeta [Lepeophtheirus salmonis] 14-3-3 protein zeta [Lepeophtheirus salmonis] Calcitonin gene-related peptide type 1 receptor precursor [Lepeophtheirus salmonis] Calcitonin gene-related peptide type 1 receptor precursor [Lepeophtheirus salmonis] Calcitonin gene-related peptide type 1 receptor precursor [Lepeophtheirus salmonis] 15 15 15 35 22 28 28 35 35 35 35 35 454 454 454 454 454 454 454 454 454 454 15.41 15.6 16.74 15.56 22.09 27.78 15.55 35.13 15.56 35.13 15.56 35.13 16.22 17.12 19.78 21.06 21.23 22.98 28.5 37.36 38.24 38.36 SKFMCP AENSCPDFP SKFMCP IRGEA LLSLHGLSTL LLETNMT EAFDEAINDL CPGWTPIRGEA IRGEA CPGWTPIRGEA IRGEA CPGWTPIRGEA QTDKHPNKDF ATLLDVIQSGVENLDS GVGIYAPDA MGLTEFQAVK QTDKHPNKDF ASVHIKLP SVHIKLPK FDPIIEDYH GVENLDSGVGIYAPD AEAY GVENLDSGVGIYAPD A ATLLDVIQSGVENLDS GVG Oxidation (M) Oxidation (M) Amidated (Cterm) Oxidation (M) Oxidation (M) Gln->pyro-Glu (N-term Q) Gln->pyro-Glu (N-term Q) 0.000400.0 0.000000000.1 0.000400.0 0.0000040.0 2.0000000000.0 0.4000000000.0 2.0000000000.0 454 Proteins found in O. rusticus, PEAKS Brain 1 Accession gi|380003174 14-3-3 protein [Scylla paramamosain] gi|321452467 ABC protein_ subfamily ABCC [Daphnia pulex] gi|321467478 ABC protein_ subfamily ABCH [Daphnia pulex] gi|395863637 antimicrobial peptide type 2 precursor [Pandalopsis japonica] gi|551380 arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] gi|551380 arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] gi|551380 arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] gi|551380 arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] gi|551380 arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] gi|551380 arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] gi|551380 arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] gi|551380 arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] gi|254832574 beta-actin [Penaeus monodon] gi|254832574 beta-actin [Penaeus monodon] gi|254832574 beta-actin [Penaeus monodon] gi|254832574 beta-actin [Penaeus monodon] gi|254832574 beta-actin [Penaeus monodon] gi|254832574 beta-actin [Penaeus monodon] gi|254832574 beta-actin [Penaeus monodon] gi|254832574 beta-actin [Penaeus monodon] gi|254832574 beta-actin [Penaeus monodon] gi|254832574 beta-actin [Penaeus monodon] gi|254832574 beta-actin [Penaeus monodon] gi|254832574 beta-actin [Penaeus monodon] gi|254832574 beta-actin [Penaeus monodon] gi|254832574 beta-actin [Penaeus monodon] gi|254832574 beta-actin [Penaeus monodon] gi|254832574 beta-actin [Penaeus monodon] gi|254832574 beta-actin [Penaeus monodon] gi|254832574 beta-actin [Penaeus monodon] gi|254832574 beta-actin [Penaeus monodon] gi|254832574 beta-actin [Penaeus monodon] gi|254832574 beta-actin [Penaeus monodon] gi|254832574 beta-actin [Penaeus monodon] gi|211947526 beta-actin [Procambarus clarkii] gi|211947526 beta-actin [Procambarus clarkii] gi|225713794 Carboxypeptidase A2 precursor [Lepeophtheirus salmonis] gi|121078585 caspase [Litopenaeus vannamei] gi|296933602 caspase [Marsupenaeus japonicus] gi|296933420 caspase [Marsupenaeus japonicus] gi|99032198 Chain A_ 1.2a Resolution Structure Of A Crayfish Trypsin Complexed With A Peptide Inhibitor_ Sgti gi|99032198 Chain A_ 1.2a Resolution Structure Of A Crayfish Trypsin Complexed With A Peptide Inhibitor_ Sgti gi|402715413 Chain A_ X-Ray Structure And Catalytic Mechanism Of Lobster Enolase gi|402715413 Chain A_ X-Ray Structure And Catalytic Mechanism Of Lobster Enolase gi|402715413 Chain A_ X-Ray Structure And Catalytic Mechanism Of Lobster Enolase gi|402715413 Chain A_ X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Peptide YYEILNSPDRACHLA AVIVTFLAHIG GGPPGTPDSGIPGP(-.98) RPFGSGGR APLFDPIIED ATLLDVIQSGVENL ATLLDVIQSGVENLDSGVG DVIQSGVENLDSGVG FAPLFDPIIED FAPLFDPIIEDY LFAPLFDPIIED LIDDHFLFK AGFAGDDAPR AGFAGDDAPRAVF AGFAGDDAPRAVFPS AGFAGDDAPRAVFPSIVG APLNPK DDAPRAVFP DDAPRAVFPSIVG DEAQSKRGILT DEAQSKRGILT EAPLNPKANR FAGDDAPR FAGDDAPRAVFPS FAGDDAPRAVFPSIVG GDDAPRAVFPSIVG HQGVM(+15.99)VGM(+15.99)GQ LKYPIEHGIIT LPHAILR LTEAPLNPK RGILTLK TEAPLNPK TEAPLNPK TEAPLNPKANR DSGDGVSHTVPIYEG VSHTVPIYEG PCEVIIAN KNVPWGR IGSALIR IGSALLR AGELDMSVNEGS(-.98) IVGGTDATLGEFPYQL AGAAELGIPLYR DEGGFAPNIL DEGGFAPNILN DEGGFAPNILN -10lgP 17.15 17.4 15.7 16.51 26.43 44.98 49.85 34.61 44.46 54.83 36.67 22.77 46.67 46.07 26.71 46.8 21.07 20.56 41.73 46.25 39.49 23.04 41.27 31.45 58.25 27.61 48.2 18.58 24.76 45.91 22.58 39.45 39.27 61.29 51.78 23.66 20.87 16.48 15.54 15.54 18.21 73.7 76.09 16.96 41.61 27.46 455 gi|402715413 gi|402715413 gi|402715413 gi|402715413 gi|402715413 gi|402715413 gi|402715413 gi|118175954 gi|290562063 gi|62701385 gi|62701385 gi|62701385 gi|62701385 gi|62701385 gi|62701385 gi|62701385 gi|62701385 gi|62701385 gi|310743896 gi|310743896 gi|310743896 gi|310743896 gi|310743896 gi|310743896 gi|310743896 gi|310743896 gi|310743896 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 Chain A_ X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A_ X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A_ X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A_ X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A_ X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A_ X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A_ X-Ray Structure And Catalytic Mechanism Of Lobster Enolase corticotropin-releasing hormone binding protein [Tigriopus japonicus] Geranylgeranyl transferase type-2 subunit beta [Lepeophtheirus salmonis] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] DEGGFAPNILNNK EVILPVP EVILPVPAF EVILPVPAF M(+15.99)GTEVYHHLK NVINGGSHAGNK NVINGGSHAGNKLA QAKRSEGGPH(-.98) WLAERQLPSGG(-.98) DGPSAKDWR GGRGAAQNIIPS GGRGAAQNIIPSS GGRGAAQNIIPSSTG GPSAKDWR GPSAKDWR TVDGPSAKDWR TVDGPSAKDWR TVDGPSAKDWR AM(+15.99)PYDNPIPGYK FVPRTVM(+15.99)IGGK GGKAAP ILPRHLQI LPAPHEP LPAPHEP LPRHLQ VLYPNDNFFEGK VVHLDQL AERLSNFLPAVDEL AETWNPRDHTDK AINRLLYK ALHNQAHRVLG APHTTYK ATRDPAF ATRDPAFFR DADEAVANGAELPH DAERLSNFLPAVDEL DEAVANGAELPH DEAVANGAELPHK DGFAPHTTY DGFAPHTTYK DGFAPHTTYKYG DPAFFR DTDVAHQQQAINR DTDVAHQQQAINR DTDVAHQQQAINRL DTDVAHQQQAINRLL DTDVAHQQQAINRLL DTDVAHQQQAINRLLY 73.62 35.26 50.48 44.17 23.16 18.32 56.12 18.99 31.64 27.14 22.05 41.19 33.54 53.36 52.28 42.98 42.54 29.25 18.94 26.52 19.78 25.52 43 37.13 18.34 25.07 18.72 30.5 48.86 17.05 67.96 33.87 20.3 46 58.33 65.39 46.27 74.58 24.61 73.44 48.27 38.95 93.48 76.15 74.36 108.77 53.35 43.99 456 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] DTDVAHQQQAINRLLYK E(-18.01)VTPHLF EAAETWNPR EAAETWNPRDHTDK ETWNPRDHTD ETWNPRDHTDK EVTPHLF EVTPHLFTNSE EVTPHLFTNSEVIDQ(-.98) FAPHTTYK FNM(+15.99)PPGVM(+15.99)EHFET FNMPPGVMEHFET FPFDRPIPD GAELPHKESR GEFPARPDNK GEFPARPDNK GFAPHTTYK GFPFDRPIPD GFPFDRPIPD KFNM(+15.99)PPGVM(+15.99) KFNM(+15.99)PPGVM(+15.99)EHFET LLLPKGK LPPLYEVTPHLF LSNFLPAVDEL M(+15.99)PPGVM(+15.99)EHFET M(+15.99)TQTPGNFK NEHGIDILGDIIE NEHGIDILGDIIE NEHGIDILGDIIES NEHGIDILGDIIESS NEHGIDILGDIIESS NQAHRVL NQAHRVLG PARPDNKEF PHTTYK SSESSVAIPDR SSESSVAIPDRVSFPQ SSESSVAIPDRVSFPQL TDINNEHGIDILG TWNPRDHTDK VFKVQNQHGQVVK VLGAQSDPK VM(+15.99)EHFETATR VQNQHGQVVKIFHH VSFPQLIHDA VSFPQLIHDADEAVAN VTPHLFTNS VVLPPLYEVTPH 112.14 39.28 18.98 19.97 34.77 59.17 31.81 41.42 60.72 38.74 54.78 39.48 27.7 45.08 49.92 39.16 35.48 42.62 27.8 21.79 51.53 28.64 55.88 41.53 51.21 47.15 44.78 19.14 22.47 51.97 40.19 18 19.87 15.8 39.13 28.08 42.62 67.26 15.09 38.52 79.99 18.75 68.94 83.88 27.7 51.4 49.69 22.55 457 gi|290462393 gi|225710178 gi|225717830 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|249511 gi|249511 gi|249511 gi|249511 gi|249511 gi|249511 gi|249511 gi|249511 gi|249511 gi|33087166 gi|387811963 gi|114386730 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|133754273 gi|326579691 gi|326579693 gi|290562011 gi|290562255 gi|310743902 gi|17223042 gi|321458499 gi|321458458 gi|321479308 gi|225709352 gi|74785595 gi|257096767 hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin subunit c [Panulirus interruptus=spiny lobster_ Peptide_ 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster_ Peptide_ 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster_ Peptide_ 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster_ Peptide_ 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster_ Peptide_ 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster_ Peptide_ 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster_ Peptide_ 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster_ Peptide_ 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster_ Peptide_ 661 aa] hepatopancreas kazal-type proteinase inhibitor [Penaeus monodon] hormone receptor-like 97a [Daphnia magna] hyasin [Hyas araneus] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] inhibitor of apoptosis protein [Penaeus monodon] JHE-like carboxylesterase 1 [Pandalopsis japonica] JHE-like carboxylesterase 2 [Pandalopsis japonica] Peptide deformylase_ mitochondrial [Lepeophtheirus salmonis] Peptidyl-tRNA hydrolase 2_ mitochondrial [Lepeophtheirus salmonis] phosphoenolpyruvate carboxykinase [Marsupenaeus japonicus] putative antimicrobial peptide [Litopenaeus setiferus] putative DH31 receptor [Daphnia pulex] putative diuretic hormone 44 receptor [Daphnia pulex] putative PDF receptor variant 2 [Daphnia pulex] Pyrazinamidase/nicotinamidase [Caligus rogercresseyi] RecName: Full=Armadillidin; Flags: Precursor RecName: Full=Succinate dehydrogenase assembly factor 2_ mitochondrial; Short=SDH assembly factor 2; AltName: Full=Succinate dehydrogenase subunit 5_ mitochondrial; Flags: Precursor Serine/threonine-protein phosphatase 2A regulatory subunit B [Lepeophtheirus salmonis] Signal recognition particle 54 kDa protein [Caligus rogercresseyi] takeout precursor [Caligus clemensi] VDEIPLE NNALSSLSKA(-.98) RLM(+15.99)PVGIPE WAHHQLTAR WNPRDHTD YAAKM(+15.99)TQT YDAERLSNFLPAVDEL YGGEFPARPD YGGEFPARPDNK DPSFFR DPSFFRLH LHNTAHIM(+15.99)LG LPNRFLLP LPNRFLLPK NTAHIM(+15.99)LGR RVDDERIF TAHIM(+15.99)LGR TRDPSFFR QEITVAYP(-.98) APLEIK PRPYRP APVDIKE E(-18.01)HAEVI EAPVDIK EPITTP KPSEVQP LKVTAPQAPT M(+15.99)LKNNVRVNEIWSMFRAGE(-.98) Q(-17.03)SLEGLK QSAHVECRL RETKAQSPI RVWTPPP(-.98) TATPTPKID(-.98) VTVTENLQRADQEDV EALKPPSSGPP(-.98) LGAVLMGMN(-.98) VTANEGAALT QTLPPYE(-.98) VVGIGPGP KAKVIM(+15.99)HDPFA(-.98) PGGFPGGRP(-.98) LPAVVT VIAGPT M(+15.99)IKAPLD(-.98) PAGENGEEVVPPI GFHSGGS(-.98) IPPYEERKNE(-.98) 29.95 18.62 22.39 20.81 24.91 18.87 77.39 43.87 81.07 31.57 42.05 18.95 22.48 22.83 59.21 22.21 18.37 43.85 15.05 15.66 15.94 19.72 15.16 23.49 16.28 27.9 17.81 21.09 15.45 15.9 20.16 15.02 18.61 15.33 16.06 18.04 18.01 26.39 27.2 16.53 16.18 28.11 15.97 30.86 15.68 16.15 19.27 458 Brain 2 gi|304360632 gi|225908473 gi|321465377 gi|321465377 Accession gi|225710814 gi|66774602 gi|321460334 gi|321468387 gi|321473806 gi|381216270 gi|77168456 gi|155964257 gi|114386732 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|450 gi|450 gi|254832574 gi|254832574 gi|254832574 gi|254832574 gi|254832574 gi|254832574 gi|254832574 gi|254832574 gi|254832574 gi|254832574 gi|254832574 gi|254832574 gi|254832574 gi|254832574 gi|254832574 gi|254832574 gi|254832574 gi|254832574 gi|254832574 gi|254832574 gi|254832574 gi|254832574 gi|254832574 gi|211947526 gi|211947526 gi|225714196 tumor necrosis factor receptor-associated factor 6 [Litopenaeus vannamei] ubiquitin carboxyl-terminal esterase L3 [Scylla paramamosain] vitellogenin fused with superoxide dismutase [Daphnia pulex] vitellogenin fused with superoxide dismutase [Daphnia pulex] Description 14-3-3 protein beta/alpha [Caligus rogercresseyi] 14-3-3-like protein [Penaeus monodon] ABC protein_ subfamily ABCH [Daphnia pulex] ABC protein_ subfamily ABCH [Daphnia pulex] ABC protein_ subfamily ABCH [Daphnia pulex] androgenic hormone_ partial [Oniscus asellus] antimicrobial peptide PEN2-4 [Litopenaeus vannamei] anti-microbial Scy2 precursor [Scylla serrata] arasin-1 [Hyas araneus] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] AVLLPKKTEKK AVLLPKKTEKK beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Procambarus clarkii] beta-actin [Procambarus clarkii] Calcitonin gene-related peptide type 1 receptor precursor [Lepeophtheirus salmonis] TVDLMYIEH(-.98) VRWLPLESNPAVM(+15.99)NK E(-18.01)AEPKLL EAVEAEP Peptide ISSIGQKT ASNPESKVFYLK PSIPVEF(-.98) LLDRNLVAGVKM(+15.01) ILVLGHEP(-.98) KVDLMFPLLE(-.98) TGPIPRPPP YTAVMS SPGRPR ATLLDVIQSGVENLDSGVG ATLLDVIQSGVENL FAPLFDPIIEDY LFAPLFDPIIED ASVHIKLP APLFDPIIED AVLLPKKTEKK AVLLPK HQGVMVGMGQK EAPLNPKANR AGFAGDDAPR AGFAGDDAPR AGFAGDDAPR AGFAGDDAPR APLNPKANR DEAQSKRGILT TEAPLNPK TEAPLNPK LTEAPLNPK FAGDDAPRAVFP VVDNGSGMVK FAGDDAPR FAGDDAPR AGFAGDDAPRAVFPS GFALPHAILR M(+15.99)GQKDAYVGDEAQSK DDAPRAVFPS E(-18.01)APLNPK AGFAGDDAPRA HQGVM(+15.99)VGM(+15.99)GQK RGILTLK VSHTVPIYEG SGDGVSHTVPIYEG VSDEYL 21.05 26.28 16.35 20.71 -10lgP 15.47 36.19 18.57 17.84 16.31 17.23 19.56 16.05 17.59 54.02 46.63 42.15 38.21 30.29 15.02 42 29.3 67.73 53.1 50.84 46.1 39.57 34.42 50.68 48.45 43.4 34.95 41.25 39.6 36.92 36.42 19.02 35.69 28.58 28.07 27.31 23.32 19.93 19.3 16 48.29 15.74 19.06 459 gi|225714196 gi|225717916 gi|225708792 gi|290562677 gi|296933280 gi|296933378 gi|296933480 gi|296933780 gi|296933780 gi|99032198 gi|402715413 gi|402715413 gi|402715413 gi|402715413 gi|402715413 gi|402715413 gi|402715413 gi|238244 gi|121615296 gi|210076885 gi|254351320 gi|162945363 gi|45385712 gi|193878327 gi|448 gi|154936860 gi|133754271 gi|133754271 gi|8698612 gi|225709118 gi|62701385 gi|62701385 gi|62701385 gi|62701385 gi|62701385 gi|62701385 gi|310743896 gi|310743896 gi|310743896 gi|310743896 gi|310743896 gi|310743896 gi|310743898 gi|318104933 gi|21914372 gi|21914372 gi|21914372 gi|21914372 Calcitonin gene-related peptide type 1 receptor precursor [Lepeophtheirus salmonis] Carboxypeptidase B [Caligus clemensi] Carboxypeptidase B [Caligus rogercresseyi] Carboxypeptidase B [Lepeophtheirus salmonis] caspase [Marsupenaeus japonicus] caspase [Marsupenaeus japonicus] caspase [Marsupenaeus japonicus] caspase [Marsupenaeus japonicus] caspase [Marsupenaeus japonicus] Chain A_ 1.2a Resolution Structure Of A Crayfish Trypsin Complexed With A Peptide Inhibitor_ Sgti Chain A_ X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A_ X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A_ X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A_ X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A_ X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A_ X-Ray Structure And Catalytic Mechanism Of Lobster Enolase Chain A_ X-Ray Structure And Catalytic Mechanism Of Lobster Enolase chymotrypsin Pm1 {EC 3.4.4.5} [Penaeus monodon=shrimps_ midgut_ Peptide Partial_ 31 aa] crustin [Litopenaeus schmitti] crustin [Paralithodes camtschaticus] crustin antimicrobial peptide [Portunus trituberculatus] crustin antimicrobial peptide [Scylla paramamosain] crustin I [Litopenaeus vannamei] crustin-2 [Eriocheir sinensis] DLPKVDTALK ecdysone receptor [Marsupenaeus japonicus] effector caspase [Penaeus monodon] effector caspase [Penaeus monodon] eyestalk peptide [Jasus edwardsii] GDP-fucose protein O-fucosyltransferase 1 precursor [Caligus rogercresseyi] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] glycogen synthase [Marsupenaeus japonicus] glycogen synthase [Marsupenaeus japonicus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] LDPLRE(-.98) IHTFM(+15.99)DQI TPLM(+15.99)SLTLH(-.98) LKLLRIEKAGSN(-.98) PTGIKDCLF ISAIQNKFEMTVE IIGFFVGP LPKTRYP KYLEVVPP(-.98) GTDATLGEFPYQ DEGGFAPNILNNK DEGGFAPNILNNK NVINGGSHAGNKL DEGGFAPNILN DEGGFAPNIL PIVSIEDPFDQ NAVKNVN GVEAVPHSWPYQAALFII TPVGTKLLD NDPLTPAQVA(-.98) E(-18.01)ASLVLPYSGL(-.98) KHHVCKT TPVGTKILD FSGGPP(-.98) DLPKVDTALK VM(+15.99)IGKLLNVLT DALVVVFMSHGEK TGLGERN TPARQR SFSAFVK TVDGPSAKDWR TVDGPSAKDWR TVDGPSAKDWR DGPSAKDWR GVNLEKYSK GGRGAAQNIIPSSTG NNVVNTMRL LPAPHEP RDIFKDF PAPHEP SPNNFNLK TNGITPR LTTAINNIR SELINSLLGLP(-.98) VQNQHGQVVKIFHH DTDVAHQQQAINR DTDVAHQQQAINR DTDVAHQQQAINR 17.81 20.09 17.4 15.41 20.15 15.4 16.37 19.69 15.78 28.75 65.54 50.53 58.27 40.25 28.56 27.02 17.47 18.85 21.6 28.82 20.29 16.78 21.6 18.6 43.41 17.69 22.38 19.82 18.55 23.58 53.22 22.69 15.78 43.88 32.54 23.81 34.42 31.48 30.63 24.93 19.61 17.06 15.96 21.08 80.87 79.45 66.87 56.91 460 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] DTDVAHQQQAINRLLY YGGEFPARPDNK KEAAETWNPRDHTDK KEAAETWNPRDHTDK EAAETWNPRDHTDK EAAETWNPRDHTDK DTDVAHQQQAINRL YGGEFPARPDNKEF LIHDADEAVANGAELPHK AETWNPRDHTDK AETWNPRDHTDK DTDVAHQQQAINRLL DTDVAHQQQAINRLL ETWNPRDHTDK VMEHFETATR MPPGVMEHFET LPPLYEVTPHLFTNS VFKVQNQHGQVVK GGEFPARPDNK DGFAPHTTYK VMEHFET YGGEFPARPD EVTPHLFTNSEVID DPAFFR EVTPHLFTNSE FKVQNQHGQVVKIFHH GEFPARPDNK E(-18.01)VTPHLF DPAFFRLH FPQLIHDADEAVAN GEFPARPDNKEF APHTTYK EVTPHLFTNS NQHGQVVKIFHH EVTPHLF NEHGIDILGDIIES GFAPHTTYK DVAHQQQAINR KVQNQHGQVVK PHTTYK AINRLLYK RPIPDLR LLLPKGK AERLSNFLPAVDEL ATRDPAFFRL EVTPHLFTNSEVID(-.98) SSESSVAIPDR GFPFDRPIPDL 74.51 71.86 63.47 25.67 61.5 25.3 60.89 59.35 59.18 58.71 29.82 57.66 51.5 54.09 49.97 47.76 47.66 45.97 45.48 44 43.59 43.51 43.32 42.93 42.3 42.05 42.03 41.03 40.06 38.17 38.01 37.96 37.6 35.47 34.54 33.93 33.14 32.68 32.62 31.86 30.46 30.26 29.87 29.6 28.2 27.87 27.64 27.58 461 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|249511 gi|249511 gi|249511 gi|249511 gi|249511 gi|249511 gi|249511 gi|249511 gi|249511 gi|249511 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|133754273 gi|46398237 hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin subunit c [Panulirus interruptus=spiny lobster_ Peptide_ 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster_ Peptide_ 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster_ Peptide_ 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster_ Peptide_ 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster_ Peptide_ 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster_ Peptide_ 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster_ Peptide_ 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster_ Peptide_ 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster_ Peptide_ 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster_ Peptide_ 661 aa] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] inhibitor of apoptosis protein [Penaeus monodon] intestinal trypsin 5 precursor [Lepeophtheirus salmonis] YGGEFPARP IDRKGELF EVTPHLFTNSEVIDQ VTPHLFT TRDPAFFR E(-18.01)VTPHLFT VM(+15.99)EHFET LLVDVLLH DGFAPHTTY SSESSVAIPDRVSFPQ EVTPHLFTNSEVIDQ(-.98) VSFPQLIHDADEAVAN DAERLSNFLPAVDEL E(-18.01)VTPHLFTNS FPFDRPIPD FPFDRP FPARPDN DPAFFRLHK PAFFRLH ATRDPAFF RLLLPK EVTPHL PARPDNK EAAETWNPR QYPDKRPHGYPLDR NTAHIM(+15.99)LGR DPSFFR YMDNIFR LPPGVLEHFET RDPSFFR LPPGVLEHF SALGLPNR ALELDRF EVIHEAYKA(-.98) ADVAPSIAPLE(-.98) TPTPVLSWQKDGV(-.98) DPNPLP(-.98) TVEVVGPRPT QADVAPSIAP(-.98) DVTSSSL KLVSQGKT(-.98) TATPTPKID KDIPEREPVELE(-.98) KPKDKPEP(-.98) IPGVKCFVK(-.98) EGLEKIRQLE(-.98) TFGSSDFVV IVGGTEVSP(-.98) 27.13 26.99 26.92 26.82 26.53 26.42 25.47 24.57 24.55 23.74 23.61 23.22 22.5 21.74 19.92 19.36 19.27 17.99 17.91 17.91 17.87 16.83 16.16 15.53 48.22 44.69 37.07 34.67 31.61 31.04 30.33 29.22 22.46 18.74 29.33 23.03 23.02 21.65 17.83 17.21 17.11 16.66 15.41 15.37 15.35 15.15 15.49 18.23 462 Eyestalk 1 14-3-3 protein epsilon [Lepeophtheirus salmonis] ABC protein_ subfamily ABCH [Daphnia pulex] ABC protein_ subfamily ABCH [Daphnia pulex] Abl interactor 2 [Lepeophtheirus salmonis] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] astacidin 2 [Pacifastacus leniusculus] gi|290562958 gi|321467477 gi|321460334 gi|290462835 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|111118804 gi|290462393 gi|225710178 gi|225908473 gi|290561503 gi|148589360 gi|148589360 gi|95113635 gi|399936386 Accession gi|257096767 putative antimicrobial peptide [Litopenaeus vannamei] putative G-protein coupled receptor_ orphan [Daphnia pulex] putative UDP-N-acetylglucosamine--peptide N-acetylglucosaminyltransferase [Mesocyclops edax] RecName: Full=NAD-dependent protein deacylase; AltName: Full=Regulatory protein SIR2 homolog 5; Flags: Precursor RecName: Full=Succinate dehydrogenase assembly factor 2_ mitochondrial; Short=SDH assembly factor 2; AltName: Full=Succinate dehydrogenase subunit 5_ mitochondrial; Flags: Precursor Serine/threonine-protein phosphatase 2A regulatory subunit B [Lepeophtheirus salmonis] Signal recognition particle 54 kDa protein [Caligus rogercresseyi] ubiquitin carboxyl-terminal esterase L3 [Scylla paramamosain] Ubiquitin carboxyl-terminal hydrolase isozyme L3 [Lepeophtheirus salmonis] unnamed protein product [Cypridina noctiluca] unnamed protein product [Cypridina noctiluca] vitellogenin fused with superoxide dismutase [Daphnia magna] voltage-gated calcium channel beta subunit transcript variant 3 [Scylla paramamosain] Description gi|17223030 gi|321477804 gi|157812760 gi|387935383 NKASEDKLNLIKN VYLRYVSY EEARQAGTI PTQRPPSPP(-.98) SVHIKLPK SVHIKLPK SVHIKLP RMGLTEFQAVK RMGLTEFQAV RM(+15.99)GLTEFQAVK RM(+15.99)GLTEFQAVK RM(+15.99)GLTEFQAV Q(-17.03)TDKHPNKDFG Q(-17.03)TDKHPNKDF Q(-17.03)TDKHPNKDF LIDDHFLFK LIDDHFLF LIDDHFL LFDPIIEDYH IDDHFLFK GTYFPLTGMSK GTYFPL FAPLFDPIIE DDHFLFK ATLLDVIQSGVENLDSGVGIYAPDAEAY ATLLDVIQSGVENLDSGVGIYAPDA ATLLDVIQSGVENLDSGVG ATLLDVIQSGVENLD ATLLDVIQSGVENL APLFDPIIEDYH VLVALM(+15.99)AAVP KYMDLCRKIQR(-.98) KGLFKGGDMS(-.98) VRWLPLESNPAVM GPINRGTT NFKQLL TVIEFFKLIVIDI KNILSEKLNE(-.98) PPLPPP(-.98) Peptide RARLLYQ IKAVILCGL AALLDRL E(-18.01)FAAAH(-.98) IAEFEHRM(+15.99)T 18.26 15.43 23.76 15.3 42.39 36.22 29.98 53.64 54.99 50.89 31.03 20.99 16.91 37.52 19.5 35.09 24.89 29.53 48.07 34.98 41.97 24.68 29.65 34.49 22.72 100.64 84.56 70.28 57.79 87.59 15.66 19.44 15.32 51.97 27.29 19.16 16.65 18.12 20.52 -10lgP 18.35 16.57 27.98 20.83 47.29 463 gi|254832574 gi|254832574 gi|254832574 gi|254832574 gi|321478601 gi|290561028 gi|304569876 gi|296933604 gi|296933318 gi|321474388 gi|159163115 gi|402715413 gi|121615293 gi|262385516 gi|254351320 gi|229459067 gi|258618631 gi|46240814 gi|46240814 gi|46240814 gi|321470051 gi|154936860 gi|226934556 gi|226934556 gi|284981903 gi|290462553 gi|62701385 gi|310743896 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|249511 beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] Btk family kinase at 29A-like protein [Daphnia pulex] Carboxypeptidase B [Lepeophtheirus salmonis] caspase [Eriocheir sinensis] caspase [Marsupenaeus japonicus] caspase [Marsupenaeus japonicus] CG13830-PA-like protein [Daphnia pulex] Chain A_ Solution Structure Of The [t8a]-Penaeidin-3 Chain A_ X-Ray Structure And Catalytic Mechanism Of Lobster Enolase crustin [Farfantepenaeus paulensis] crustin 1 [Procambarus clarkii] crustin antimicrobial peptide [Portunus trituberculatus] crustin Pm4 antimicrobial peptide [Penaeus monodon] crustin-like antimicrobial peptide [Fenneropenaeus indicus] crustin-like peptide type 3 [Marsupenaeus japonicus] crustin-like peptide type 3 [Marsupenaeus japonicus] crustin-like peptide type 3 [Marsupenaeus japonicus] dissatisfaction-like protein [Daphnia pulex] ecdysone receptor [Marsupenaeus japonicus] ecdysteroid receptor isoform [Neomysis integer] ecdysteroid receptor isoform [Neomysis integer] fortilin binding protein [Penaeus monodon] Geranylgeranyl transferase type-2 subunit beta [Lepeophtheirus salmonis] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin subunit c [Panulirus interruptus=spiny lobster_ Peptide_ 661 aa] TEAPLNPK DAYVGDEAQSK AVFPSIVGR AGFAGDDAPRAVFPSIVG Q(-17.03)TVKVVEAVDPA LKEQDISAEVI Q(-17.03)CREVREM(+15.99)A PEYQFYSSFTCW(-.98) FFFTTHADT DSVNQLDD PLPGGPI FAGKNFR FGGGGVGGGF PCLSLN(-.98) E(-18.01)ASLVLPYSGL(-.98) GGGGVNGGGL PGHGGIAPGFE(-.98) GVQGGGVGGV GGGFGGGFGGPQGGG GFGGVQGGGV FGGGGGGGGGG(-.98) FGDLLREDQ YFDNEPY(-.98) AHQLRVSSLD PPCPYSK(-.98) IQIAAIYDSM(+15.99) TVDGPSAKDWR PAADLSEQIST YGGEFPARPDNK YGGEFPARPDN VSFPQLIHDADEAVAN VSFPQLIHDA VFKVQNQHGQVVK VFKVQNQHGQVV PMGFPFDRPIPDL PGNFKM(+15.99) LLLPKGK GGEFPARPDNK GFPFDRPIPDL FPFDRPIPDL EAAETWNPR DTDVAHQQQAINR DTDVAHQQQAINR DTDVAHQQQAIN DGFAPHTTYK DEAVANGAELPH AETWNPR KELGETFNPQGD(-.98) 34.19 43.65 36.52 56.23 16.05 25.52 21.14 26.58 25.89 17.5 20.04 30.99 19.22 17.17 16.55 19.96 18.27 20.65 25.24 17.65 43.96 19.16 21.28 15.8 15.08 16.91 33.18 20.19 85.4 65 26.76 30.25 65.63 59.42 32.78 22.94 32.41 16.15 43.5 33.55 43.12 105.32 83.35 56.92 49.7 22.65 19.87 16.03 464 Eyestalk 2 14-3-3 protein beta/alpha [Caligus rogercresseyi] ABC protein_ subfamily ABCH [Daphnia pulex] antimicrobial peptide [Penaeus monodon] antimicrobial peptide type 1 precursor [Pandalopsis japonica] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] gi|225710814 gi|321473806 gi|317383206 gi|395863635 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|321465377 Accession gi|14285771 gi|225710178 gi|2655270 gi|158266420 gi|251354 gi|257096767 gi|6015045 hemocyanin subunit c [Panulirus interruptus=spiny lobster_ Peptide_ 661 aa] hormone receptor-like 97a [Daphnia magna] hormone receptor-like 97a [Daphnia magna] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] phantom [Marsupenaeus japonicus] putative DH31 receptor [Daphnia pulex] putative PDF receptor variant 2 [Daphnia pulex] Pyrazinamidase/nicotinamidase [Caligus clemensi] RecName: Full=Nuclear hormone receptor E75; AltName: Full=Nuclear receptor subfamily 1 group D member 3 RecName: Full=Nuclear hormone receptor E75; AltName: Full=Nuclear receptor subfamily 1 group D member 3 RecName: Full=Succinate dehydrogenase assembly factor 2_ mitochondrial; Short=SDH assembly factor 2; AltName: Full=Succinate dehydrogenase subunit 5_ mitochondrial; Flags: Precursor RecName: Full=Superoxide dismutase [Mn]_ mitochondrial; Flags: Precursor Signal recognition particle 54 kDa protein [Caligus rogercresseyi] small heat shock/alpha-crystallin protein precursor [Artemia franciscana] stylicine 2 [Litopenaeus stylirostris] vacuolar H(+)-ATPase proteolipid subunit homolog [Nephrops norvegicus_ hepatopancreas_ Peptide Partial_ 151 aa] vitellogenin fused with superoxide dismutase [Daphnia pulex] Description gi|249511 gi|387811963 gi|387811963 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|224471277 gi|321458499 gi|321479308 gi|225717724 gi|6015045 E(-18.01)KVEGELR IYLLPPFLST VPGVGVPGVGGG SPRSPIP(-.98) APLFDPIIEDYH ASVHIKLP ASVHIKLP ASVHIKLPK ATLLDVIQSGVENL ATLLDVIQSGVENLD ATLLDVIQSGVENLDSGVG ATLLDVIQSGVENLDSGVGIYAPDA DPIIEDYH AM(+15.99)GPWFTKLMEF(-.98) Peptide DPLEATTGLVP AGGAGVM(+15.99)EM(+15.99) TPARTTRSGG(-.98) RLPGWEPP TLYQGFVHM(+15.99)GAGL IPPYEERKNE(-.98) KAKILAAMQS DPSFFR QPPTSQDTNNNQP NEASAPAENTYHP(-.98) VRTTEDDEGMYS(-.98) SPPTAPLEVRPT RFKTSYDF QTELLDDVTKPMPVVA PIEEKF PDGVTEPVKP(-.98) KPRFLT KLKKKPEPKI IIEPEPEPEPELIILRPVE E(-18.01)VSDKIPELA(-.98) E(-18.01)PQATGKFR ALTGPSQIM(+15.99)E AAPIEEKF KHPAFMPFQ YFLVSNYF(-.98) M(+15.99)IKAPLD(-.98) TAKDSM(+15.99)ELGF(-.98) KMTEHTAA 16.55 15.13 15.37 15.69 87.17 36.88 19.31 59.85 46.56 65.5 40.82 55.77 52.02 15.8 -10lgP 27.45 19.76 16.8 20.47 16.19 33.16 18.84 33.81 17.24 18.99 16.84 16.04 15.77 20.6 20.65 17.16 19.01 17.53 70.08 21.22 22.14 16.21 41.66 18.61 15.64 46.42 20.75 18.17 465 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|551380 gi|254832574 gi|254832574 gi|254832574 gi|254832574 gi|254832574 gi|254832574 gi|290561028 gi|296933458 gi|296933666 gi|296933806 gi|385048424 gi|157881028 gi|402715413 gi|238245 gi|118175954 gi|46240814 gi|46240814 gi|73426824 gi|448 gi|448 gi|290784917 gi|402169217 gi|402169217 gi|402169217 gi|290562063 gi|290562063 gi|62701385 gi|62701385 gi|310743896 gi|321458864 gi|321458864 gi|21914372 gi|21914372 gi|21914372 arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] arginine kinase_ AK [Penaeus japonicus=shrimps_ tail muscle_ Peptide_ 355 aa] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] beta-actin [Penaeus monodon] Carboxypeptidase B [Lepeophtheirus salmonis] caspase [Marsupenaeus japonicus] caspase [Marsupenaeus japonicus] caspase [Marsupenaeus japonicus] caspase 8_ partial [Daphnia pulex] Chain A_ Nmr Structure Of The Synthetic Penaeidin 4 Chain A_ X-Ray Structure And Catalytic Mechanism Of Lobster Enolase chymotrypsin Pm2 {EC 3.4.4.5} [Penaeus monodon=shrimps_ midgut_ Peptide Partial_ 31 aa] corticotropin-releasing hormone binding protein [Tigriopus japonicus] crustin-like peptide type 3 [Marsupenaeus japonicus] crustin-like peptide type 3 [Marsupenaeus japonicus] crustin-like protein fc-2 [Fenneropenaeus chinensis] DLPKVDTALK DLPKVDTALK duplex-specific nuclease [Chionoecetes opilio] ecdysone phosphate phosphatase [Daphnia magna] ecdysone phosphate phosphatase [Daphnia magna] ecdysone phosphate phosphatase [Daphnia magna] Geranylgeranyl transferase type-2 subunit beta [Lepeophtheirus salmonis] Geranylgeranyl transferase type-2 subunit beta [Lepeophtheirus salmonis] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glyceraldehyde 3-phosphate dehydrogenase [Marsupenaeus japonicus] glycogen phosphorylase [Marsupenaeus japonicus] GPCR-like protein_ family B [Daphnia pulex] GPCR-like protein_ family B [Daphnia pulex] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] EVQQKLIDDHFLFK FDPIIEDYH GVENLDSGVGIYAPDA GVENLDSGVGIYAPDAEAY HIKLPK IDDHFLFK LIDDHFLF LIDDHFLF LIDDHFLF M(+15.99)GLTEFQAVK Q(-17.03)TDKHPNKDF Q(-17.03)TDKHPNKDF SVHIKLPK TEAQYKEM(+15.99)QQ(-.98) AGFAGDDAPR AGFAGDDAPRAVFPSIVG AVFPSIVGR FAGDDAPR GFAGDDAPR HQGVMVGMGQK LKEQDISAEVI LLLATTVALHN(-.98) QLPPATTVALHN TPKKKAKLFF(-.98) LKQEDF RPIFIRP(-.98) AGAAELGIPLYR VGGVEAVPGV TTLLFPANFE(-.98) GFGGVQGGGV GPQGGGFGG VPGVGGGFVP DLPKVDTALK PKVDTALK IVRIAAGATV(-.98) ILGETQAGLLGQAL(-.98) LIGHASTLD LVGGLP ILYTLSA IQIAAIYDSM DGPSAKDWR TVDGPSAKDWR Q(-17.03)GKKWVDTQIVFAM(+15.99) ITVKYLRLT M(+15.99)FISILLNSILVIVF AVTHSDLTPH(-.98) DGFAPHTTY DGFAPHTTYK 75.5 35.31 23.53 59.8 17.38 23.88 32.51 23.62 23.35 39.45 26.92 26.12 46.58 15.12 30.29 23.98 47.75 40.04 27.39 77.03 36.42 15.76 23.1 15.24 18.01 18.66 60.66 18.98 18.57 25.64 20.44 16.91 22.66 40.14 16.97 15.52 21.13 20.05 21.65 16.17 21.15 23.8 16.79 15.5 21.24 15.03 26.96 60.13 466 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|21914372 gi|249511 gi|249511 gi|387811965 gi|387811965 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|15425681 gi|46398233 gi|326579693 gi|298684192 gi|225717516 gi|76786564 gi|119655552 gi|356483023 gi|321458458 gi|321458458 gi|321479308 gi|225717724 gi|74785595 gi|74785595 gi|74836523 hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin [Pacifastacus leniusculus] hemocyanin subunit c [Panulirus interruptus=spiny lobster_ Peptide_ 661 aa] hemocyanin subunit c [Panulirus interruptus=spiny lobster_ Peptide_ 661 aa] hormone receptor-like 97b [Daphnia magna] hormone receptor-like 97b [Daphnia magna] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] I-connectin [Procambarus clarkii] intestinal trypsin 3 precursor [Lepeophtheirus salmonis] JHE-like carboxylesterase 2 [Pandalopsis japonica] Kazal-type serine proteinase inhibitor 2 [Procambarus clarkii] Mitogen-activated protein kinase kinase kinase 9 [Caligus clemensi] PEN2-1 [Litopenaeus vannamei] peritrophin 3 precursor [Penaeus monodon] putative crustin-like antimicrobial peptide [Nephrops norvegicus] putative diuretic hormone 44 receptor [Daphnia pulex] putative diuretic hormone 44 receptor [Daphnia pulex] putative PDF receptor variant 2 [Daphnia pulex] Pyrazinamidase/nicotinamidase [Caligus clemensi] RecName: Full=Armadillidin; Flags: Precursor RecName: Full=Armadillidin; Flags: Precursor RecName: Full=Scygonadin; Flags: Precursor DLKELESR DPAFFR DTDVAHQQQAIN DTDVAHQQQAINR EAAETWNPR FNMPPGVMEHFE GEFPARPDNK GFPFDRPIPDL MGFPFDRPIPDL MTQTPGNFK PMGFPFDRPIPDL RPMGFPFDRPIPDL TATRDPAFF VAHGYIINADGT VSFPQLIHDA VVLPPL YGGEFPARPD YGGEFPARPDNK YGGEFPARPDNKEF DPSFFR YGGYFPSRPDN IESIVTKYM TSDCGTEQDAC(-.98) AAPIEEKF APIEEKF E(-18.01)AKATVTEELK ISQDTVQV KNHITLKVTAP KPIETSSRINRTH LEGRIEPV(-.98) LEPLKPLK LSKANLYFDT QTHQKEKEFL SEEVTIKKPEEKI ISALFFLV LEAPVIST PVGRLCGRY(-.98) KIRPDVLIDWSIQMAR IPRPPPI YSKLREK TVVTVALGSGH LIPLLGLTYVL(-.98) WGIPLPVVIVWAVV(-.98) LLCWPPT LKLAENSIIIHK GFNRGGGF GGFNRGGGF KLMPKIVSAII 29.2 27.99 60.99 110.22 33.46 58.8 33.94 18.01 45.57 24.34 36.67 31.38 27.93 20.01 50.3 43.54 53.47 89.07 45.81 29.56 40.91 17.34 15.2 27.55 15.21 15.81 17.21 17 16.67 22.96 16.77 23.25 15.01 19.36 22.28 25.83 15.65 16.66 16.79 15.32 23.22 24.66 17.89 15.84 19.03 33.19 18.48 20.32 467 gi|71040960 gi|71040960 gi|290462393 gi|290462393 gi|2655270 gi|2655270 gi|304360632 gi|304360632 gi|95113635 gi|95113635 gi|225711432 RXRd nuclear hormone receptor [Gecarcinus lateralis] RXRd nuclear hormone receptor [Gecarcinus lateralis] Serine/threonine-protein phosphatase 2A regulatory subunit B [Lepeophtheirus salmonis] Serine/threonine-protein phosphatase 2A regulatory subunit B [Lepeophtheirus salmonis] small heat shock/alpha-crystallin protein precursor [Artemia franciscana] small heat shock/alpha-crystallin protein precursor [Artemia franciscana] tumor necrosis factor receptor-associated factor 6 [Litopenaeus vannamei] tumor necrosis factor receptor-associated factor 6 [Litopenaeus vannamei] vitellogenin fused with superoxide dismutase [Daphnia magna] vitellogenin fused with superoxide dismutase [Daphnia magna] Zinc finger MYND domain-containing protein 19 [Caligus rogercresseyi] ASEVPCANP(-.98) TM(+15.99)GMKREAA(-.98) VDEIPLEEQSQ VDEIPLEEQSQR APAVGRIEGGTTGTT FGFGGFG(-.98) KGGADSGIE LSNLAKRIEEVDAL(-.98) DPSSKYTSDKIN E(-18.01)TVAIGKEE(-.98) RVLLRYYN(-.98) 19.7 18.39 21.82 15.48 15.35 19.95 18.38 15.89 19.86 18.43 16.68 468 Protein Identifications from O. rusticus, Hypothetical Proteins from Daphnia pulex genome, Mascot Protein Mascot Brain 1 Protein Accession # Protein Description Score hypothetical protein gi|321455882|gb|EFX67003.1| DAPPUDRAFT_189558 [Daphnia pulex] hypothetical protein gi|321455882|gb|EFX67003.1| DAPPUDRAFT_189558 [Daphnia pulex] hypothetical protein gi|321477436|gb|EFX88395.1| DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein gi|321477436|gb|EFX88395.1| DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein gi|321477436|gb|EFX88395.1| DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein gi|321477436|gb|EFX88395.1| DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein gi|321477436|gb|EFX88395.1| DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein gi|321477436|gb|EFX88395.1| DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein gi|321477436|gb|EFX88395.1| DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein gi|321477436|gb|EFX88395.1| DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein gi|321477436|gb|EFX88395.1| DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein gi|321477436|gb|EFX88395.1| DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein gi|321477436|gb|EFX88395.1| DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein gi|321477436|gb|EFX88395.1| DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein gi|321477714|gb|EFX88672.1| DAPPUDRAFT_206384 [Daphnia pulex] hypothetical protein gi|321477714|gb|EFX88672.1| DAPPUDRAFT_206384 [Daphnia pulex] hypothetical protein gi|321477714|gb|EFX88672.1| DAPPUDRAFT_206384 [Daphnia pulex] hypothetical protein gi|321477714|gb|EFX88672.1| DAPPUDRAFT_206384 [Daphnia pulex] hypothetical protein gi|321477714|gb|EFX88672.1| DAPPUDRAFT_206384 [Daphnia pulex] hypothetical protein gi|321477714|gb|EFX88672.1| DAPPUDRAFT_206384 [Daphnia pulex] hypothetical protein gi|321477714|gb|EFX88672.1| DAPPUDRAFT_206384 [Daphnia pulex] hypothetical protein gi|321473719|gb|EFX84686.1| DAPPUDRAFT_209520 [Daphnia pulex] 20.13 15.95 27.85 24.99 23.24 21.18 20.67 20.2 18.97 18.04 17.24 15.66 15.42 15.12 22.31 19.3 18.82 17.44 17.31 16.5 15.8 29.53 20 20 25 25 25 25 25 25 25 25 25 25 25 25 19 19 19 19 19 19 19 30 Peptide Mascot Score EAQDIL KLMAATRGTKLE DLKKIEDLGAILEE KALDE YVKKIEPGLS QTATILN QGDNVEALIKK QEIEN ICQQKLMQSYA VISLI RYPDGIHYG PLNAPF RIVLDR LKISSEMR REELQR RIVLDR LETPPAFPI EQILEASPLLE RIVLDR CNQTWRNEDPEA VA QINKANQY EMMSSTGRTTPSS LT Peptide Sequence Glu->pyro-Glu (N-term E) Amidated (C-term) Amidated (C-term) Amidated (C-term) Oxidation (M) Amidated (C-term) Amidated (C-term) Amidated (C-term); Oxidation (M) Amidated (C-term) Glu->pyro-Glu (N-term E) Amidated (C-term) Glu->pyro-Glu (N-term E); 2 Oxidation (M) Peptide Variable Modifications 3.000000.0 0.0000000.1 0.00000000000.1 0.00000.1 0.00000040000.0 0.000000.1 0.000000.1 0.00000040.1 0.000000.1 3.00000000000.0 0.000000.1 3.044000000000000. 0 Position of Variable Modifications 469 gi|321469429|gb|EFX80409.1| gi|321469429|gb|EFX80409.1| gi|321469652|gb|EFX80631.1| gi|321469652|gb|EFX80631.1| gi|321478348|gb|EFX89305.1| gi|321478348|gb|EFX89305.1| gi|321478348|gb|EFX89305.1| gi|321478348|gb|EFX89305.1| gi|321478348|gb|EFX89305.1| gi|321478348|gb|EFX89305.1| gi|321478348|gb|EFX89305.1| gi|321478348|gb|EFX89305.1| gi|321478348|gb|EFX89305.1| gi|321478348|gb|EFX89305.1| gi|321473719|gb|EFX84686.1| gi|321473719|gb|EFX84686.1| gi|321473719|gb|EFX84686.1| gi|321473719|gb|EFX84686.1| gi|321473719|gb|EFX84686.1| gi|321473719|gb|EFX84686.1| gi|321473719|gb|EFX84686.1| gi|321473719|gb|EFX84686.1| gi|321473719|gb|EFX84686.1| gi|321473719|gb|EFX84686.1| hypothetical protein DAPPUDRAFT_209520 [Daphnia pulex] hypothetical protein DAPPUDRAFT_209520 [Daphnia pulex] hypothetical protein DAPPUDRAFT_209520 [Daphnia pulex] hypothetical protein DAPPUDRAFT_209520 [Daphnia pulex] hypothetical protein DAPPUDRAFT_209520 [Daphnia pulex] hypothetical protein DAPPUDRAFT_209520 [Daphnia pulex] hypothetical protein DAPPUDRAFT_209520 [Daphnia pulex] hypothetical protein DAPPUDRAFT_209520 [Daphnia pulex] hypothetical protein DAPPUDRAFT_209520 [Daphnia pulex] hypothetical protein DAPPUDRAFT_209520 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303153 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303153 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303153 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303153 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303153 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303153 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303153 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303153 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303153 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303153 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303899 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303899 [Daphnia pulex] hypothetical protein DAPPUDRAFT_304111 [Daphnia pulex] hypothetical protein DAPPUDRAFT_304111 [Daphnia pulex] 45 45 23 23 27 27 27 27 27 27 27 27 27 27 30 30 30 30 30 30 30 30 30 30 30.63 46.25 18.33 22.89 15.28 15.36 15.92 16.23 18.07 18.59 21.25 24.04 29.08 31.18 15.2 15.66 15.74 17.21 18.56 20.12 20.57 22.23 25.61 27.99 APPRPPPP APPRPPPP KEIAA GAPVD QPGDFILAIN DMNAKNASG QSQPIEI EAVRTLLDLGASP AGNLE GADREALNYA DDATILENHK KTMTGMKD EVEIPP SLQLIR QEAFLNNDDL GQMME EQLMAAGHFA EKLVAAGEIH KDNLNAGEAAR LELDTYSGLINEM KDLPQV QDIDQMLD QEIEI QGLEN Amidated (C-term) Amidated (C-term) Oxidation (M) Amidated (C-term) Amidated (C-term) Amidated (C-term) Oxidation (M) Amidated (C-term) Oxidation (M) Oxidation (M) Amidated (C-term) Amidated (C-term) 0.00000.1 0.0000000000.1 0.040000000.0 0.0000000000000.1 0.0000000000.1 0.0000000000.1 0.00000400.0 0.00000.1 0.0004000000.0 0.0000000000004.0 0.00000000.1 0.00000.1 470 gi|321476878|gb|EFX87838.1| gi|321476878|gb|EFX87838.1| gi|321476878|gb|EFX87838.1| gi|321476878|gb|EFX87838.1| gi|321460846|gb|EFX71884.1| gi|321460846|gb|EFX71884.1| gi|321460846|gb|EFX71884.1| gi|321475268|gb|EFX86231.1| gi|321475268|gb|EFX86231.1| gi|321475268|gb|EFX86231.1| gi|321475268|gb|EFX86231.1| gi|321475268|gb|EFX86231.1| gi|321461797|gb|EFX72825.1| gi|321461797|gb|EFX72825.1| gi|321461797|gb|EFX72825.1| gi|321461797|gb|EFX72825.1| gi|321463228|gb|EFX74245.1| gi|321463228|gb|EFX74245.1| gi|321476821|gb|EFX87781.1| gi|321476821|gb|EFX87781.1| gi|321469429|gb|EFX80409.1| gi|321469429|gb|EFX80409.1| gi|321469429|gb|EFX80409.1| gi|321469429|gb|EFX80409.1| hypothetical protein DAPPUDRAFT_304111 [Daphnia pulex] hypothetical protein DAPPUDRAFT_304111 [Daphnia pulex] hypothetical protein DAPPUDRAFT_304111 [Daphnia pulex] hypothetical protein DAPPUDRAFT_304111 [Daphnia pulex] hypothetical protein DAPPUDRAFT_306505 [Daphnia pulex] hypothetical protein DAPPUDRAFT_306505 [Daphnia pulex] hypothetical protein DAPPUDRAFT_307391 [Daphnia pulex] hypothetical protein DAPPUDRAFT_307391 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308055 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308055 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308055 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308055 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308511 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308511 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308511 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308511 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308511 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308707 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308707 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308707 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] 23 23 23 23 45 45 45 20 20 20 20 20 18 18 18 18 56 56 24 24 45 45 45 45 19.28 20.2 23.18 30.75 15.67 30.63 46.25 15.04 15.84 16.06 18.9 20.42 15.69 17.31 17.63 25.61 19.94 55.65 17.99 23.52 15.3 15.48 19.36 23.6 LARDPDE SIIGGQDAEAFT AVLLLL EQIIGQELKSGGLE KI KPVPPPKPKN APPRPPPP APPRPPPP LTPLPP KNIESGAIRS EVPKIT LQYNSLLE ETTTAAPSSETKP LDIRK KADLE SIPPPPPPL KEIEI AVVDDSQK ASNPESKVFYLK AVQKLMVHNW SSRAPPR LSGRINFQIQYQLS F APDLLI SSRAPPR SSRAPPR Amidated (C-term) Amidated (C-term) Amidated (C-term) Amidated (C-term) Amidated (C-term); Glu>pyro-Glu (N-term E) Amidated (C-term) Amidated (C-term) Amidated (C-term) Amidated (C-term) Amidated (C-term) 0.0000000.1 0.000000.1 0.000000.1 0.00000000.1 3.0000000000000.1 0.000000000.1 0.00000000.1 0.0000000000.1 0.000000000000000. 1 0.000000.1 471 gi|321470568|gb|EFX81544.1| gi|321470568|gb|EFX81544.1| gi|321470568|gb|EFX81544.1| gi|321470568|gb|EFX81544.1| gi|321470568|gb|EFX81544.1| gi|321470568|gb|EFX81544.1| gi|321470568|gb|EFX81544.1| gi|321470568|gb|EFX81544.1| gi|321470568|gb|EFX81544.1| gi|321470568|gb|EFX81544.1| gi|321470568|gb|EFX81544.1| gi|321470568|gb|EFX81544.1| gi|321470568|gb|EFX81544.1| gi|321473952|gb|EFX84918.1| gi|321473952|gb|EFX84918.1| gi|321473952|gb|EFX84918.1| gi|321474567|gb|EFX85532.1| gi|321476878|gb|EFX87838.1| gi|321476878|gb|EFX87838.1| gi|321476878|gb|EFX87838.1| gi|321476878|gb|EFX87838.1| gi|321476878|gb|EFX87838.1| gi|321476878|gb|EFX87838.1| gi|321476878|gb|EFX87838.1| hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_314077 [Daphnia pulex] hypothetical protein DAPPUDRAFT_314509 [Daphnia pulex] hypothetical protein DAPPUDRAFT_314509 [Daphnia pulex] hypothetical protein DAPPUDRAFT_314509 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] 22 22 22 22 22 22 22 22 22 22 22 22 22 27 27 27 23 23 23 23 23 23 23 23 16.62 16.82 17.54 17.82 18.36 18.43 18.8 19.48 20.27 21.33 22.97 24.23 30.75 15.73 18.78 27.39 22.89 15 15.27 15.37 15.68 16.03 16.17 17.93 SLSIR TIAFKLESPQ QPLTTLLE KPRDSSN ISAETTCSDF KEPSPTKL HLDLP PAMQAEGIPI PIRCSLVVGAKLG DQLEPF PTTCALPLH ADVVQMAS GATAGEPKNA AVLLLL WLDLEKPMNRQ QLELSLTDP KDPEGVPLN GAPVD RLVLNNFR QPCPATW NPIVA QTMYTANV GDVAVVQP QPATGAIIT AGEPMP Amidated (C-term) Amidated (C-term) Amidated (C-term) Amidated (C-term) Amidated (C-term) Amidated (C-term); Oxidation (M) Amidated (C-term) Amidated (C-term) Amidated (C-term) Amidated (C-term) Amidated (C-term) Oxidation (M) 0.0000000000.1 0.00000000.1 0.0000000.1 0.0000000000.1 0.00000400.1 0.0000000000.1 0.000000.1 0.00000000000.1 0.000000000.1 0.00000.1 0.00000000.1 0.000040.0 472 gi|321462130|gb|EFX73155.1| gi|321462130|gb|EFX73155.1| gi|321470448|gb|EFX81424.1| gi|321470448|gb|EFX81424.1| gi|321470448|gb|EFX81424.1| gi|321470448|gb|EFX81424.1| gi|321470448|gb|EFX81424.1| gi|321470448|gb|EFX81424.1| gi|321470448|gb|EFX81424.1| gi|321470448|gb|EFX81424.1| gi|321470448|gb|EFX81424.1| gi|321470448|gb|EFX81424.1| gi|321470448|gb|EFX81424.1| gi|321470448|gb|EFX81424.1| gi|321470448|gb|EFX81424.1| gi|321470448|gb|EFX81424.1| gi|321470448|gb|EFX81424.1| gi|321470448|gb|EFX81424.1| gi|321470448|gb|EFX81424.1| gi|321470568|gb|EFX81544.1| gi|321470568|gb|EFX81544.1| gi|321470568|gb|EFX81544.1| gi|321470568|gb|EFX81544.1| gi|321470568|gb|EFX81544.1| hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_325390 [Daphnia pulex] hypothetical protein DAPPUDRAFT_325390 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] 24 24 22 22 22 22 22 22 22 22 22 22 22 22 22 22 22 22 22 22 22 22 22 22 27.32 30.73 15.07 15.24 15.28 15.28 15.42 16 16.29 16.41 16.58 17.1 17.45 17.93 18.66 20.14 21.01 21.57 22.49 15.26 15.28 15.58 15.75 16.58 IAQEGVDMF ISKLLGL QPTMQPTMQ LIATSPPP QLIMIR MQPMMQPMMQP MMQPMMQ MISPP GRLLVL QPMMQPV QPMVQPI LSDIDMDGRL QTGLFP PAMQPTMQPTMQ PAMQPM ETADLSSLSP EQEERERK PMVQPIMSSNLP RGILVQ LIATSPPPT KAVCTHAVLEL EESSSPILGQGEIV DN LEVLAKIPV QAVVSL WIQEVAISVQR TGLLNKLMH Oxidation (M) Oxidation (M) 4 Oxidation (M) Amidated (C-term); Gln>pyro-Glu (N-term Q); Oxidation (M) Oxidation (M) Oxidation (M) Oxidation (M) 2 Oxidation (M) Amidated (C-term) Amidated (C-term) Amidated (C-term) Amidated (C-term) Gln->pyro-Glu (N-term Q) Oxidation (M) 0.000000040.0 0.000000040.0 2.000400.1 0.400440000000000 040.0 0.0004000.0 0.0040000.0 0.0000040000.0 0.000000.1 0.000000400000000 004.0 0.00000000.1 0.000000.1 0.000000000000000 0.1 2.000000.0 0.000000040.0 473 Brain 2 gi|321468653|gb|EFX79637.1| gi|321468653|gb|EFX79637.1| gi|321468653|gb|EFX79637.1| gi|321468653|gb|EFX79637.1| gi|321468030|gb|EFX79017.1| gi|321473999|gb|EFX84965.1| gi|321477436|gb|EFX88395.1| gi|321477436|gb|EFX88395.1| gi|321477436|gb|EFX88395.1| gi|321477436|gb|EFX88395.1| gi|321477436|gb|EFX88395.1| gi|321477436|gb|EFX88395.1| gi|321470602|gb|EFX81578.1| gi|321470602|gb|EFX81578.1| gi|321470602|gb|EFX81578.1| gi|321470602|gb|EFX81578.1| gi|321470602|gb|EFX81578.1| gi|321470602|gb|EFX81578.1| Protein Accession # gi|321454896|gb|EFX66047.1| gi|321454896|gb|EFX66047.1| gi|321462130|gb|EFX73155.1| gi|321462130|gb|EFX73155.1| gi|321462130|gb|EFX73155.1| Protein Description hypothetical protein DAPPUDRAFT_102305 [Daphnia pulex] hypothetical protein DAPPUDRAFT_102305 [Daphnia pulex] hypothetical protein DAPPUDRAFT_102305 [Daphnia pulex] hypothetical protein DAPPUDRAFT_102305 [Daphnia pulex] hypothetical protein DAPPUDRAFT_102305 [Daphnia pulex] hypothetical protein DAPPUDRAFT_102305 [Daphnia pulex] hypothetical protein DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein DAPPUDRAFT_194294 [Daphnia pulex] hypothetical protein DAPPUDRAFT_225308 [Daphnia pulex] hypothetical protein DAPPUDRAFT_30197 [Daphnia pulex] hypothetical protein DAPPUDRAFT_30197 [Daphnia pulex] hypothetical protein DAPPUDRAFT_30197 [Daphnia pulex] hypothetical protein DAPPUDRAFT_30197 [Daphnia pulex] hypothetical protein DAPPUDRAFT_325390 [Daphnia pulex] hypothetical protein DAPPUDRAFT_325390 [Daphnia pulex] hypothetical protein DAPPUDRAFT_325390 [Daphnia pulex] hypothetical protein DAPPUDRAFT_332604 [Daphnia pulex] hypothetical protein DAPPUDRAFT_332604 [Daphnia pulex] Protein Mascot Score 34 34 34 34 21 24 24 24 24 24 24 24 35 35 35 35 35 35 19 19 24 24 24 15.14 17.68 17.92 33.66 20.63 23.97 15.22 15.33 16.08 16.1 17.26 24.4 15.08 15.59 17.2 17.4 18.04 34.9 18.94 Peptide Mascot Score 22.27 16.08 18.48 24.37 RQPPM ELLKPGHGRDR LAGGGGGELALG RQPPM EQILE ESEAVAIRDSLAA NSGFVKA PFRVGIFCVLDQL DR ISSEMRAKLE KASPGSIGKLKIS EPMSRIEARK AEKEPLE ELKPVEKK CNQTWRNEDPEA VA PPPPSRV KELKPVEK DPPNRS APPPPPPPPPPP APPPPPPPPPPP Peptide Sequence ISPSSCTNK AGGLNK SLKGSPEAVLNSL N GLGDIEGLIDK GAGGLGNLMGM GG Oxidation (M) Amidated (C-term); Glu>pyro-Glu (N-term E) Oxidation (M) Amidated (C-term) Amidated (C-term) Amidated (C-term); Oxidation (M) Oxidation (M) Amidated (C-term) Amidated (C-term) Amidated (C-term) Amidated (C-term) Peptide Variable Modifications Amidated (C-term) Amidated (C-term); 2 Oxidation (M) 0.00004.0 3.00000000000.1 0.00004.0 0.000000000000000 00000.1 0.000000000000000. 1 0.0000400000.1 0.0040000000.0 0.0000000.1 0.00000000.1 0.0000000.1 0.000000.1 Position of Variable Modifications 0.00000000000000.1 0.0000000040400.1 474 gi|321470568|gb|EFX81544.1| gi|321470568|gb|EFX81544.1| gi|321470568|gb|EFX81544.1| gi|321470568|gb|EFX81544.1| gi|321470568|gb|EFX81544.1| gi|321470568|gb|EFX81544.1| gi|321470568|gb|EFX81544.1| gi|321476878|gb|EFX87838.1| gi|321476878|gb|EFX87838.1| gi|321476878|gb|EFX87838.1| gi|321476878|gb|EFX87838.1| gi|321476878|gb|EFX87838.1| gi|321476878|gb|EFX87838.1| gi|321476878|gb|EFX87838.1| gi|321476878|gb|EFX87838.1| gi|321477347|gb|EFX88306.1| gi|321477347|gb|EFX88306.1| gi|321463228|gb|EFX74245.1| gi|321463228|gb|EFX74245.1| gi|321478348|gb|EFX89305.1| gi|321478348|gb|EFX89305.1| gi|321478348|gb|EFX89305.1| gi|321478348|gb|EFX89305.1| gi|321478348|gb|EFX89305.1| hypothetical protein DAPPUDRAFT_303153 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303153 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303153 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303153 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303153 [Daphnia pulex] hypothetical protein DAPPUDRAFT_307391 [Daphnia pulex] hypothetical protein DAPPUDRAFT_307391 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311561 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311561 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] 25 25 25 25 25 25 25 31 31 31 31 31 31 31 31 17 17 106 106 29 29 29 29 29 18.17 20.53 21.83 21.94 26.23 28.05 30.52 15.28 15.41 15.59 15.59 16.47 19.78 24.21 30.52 15.13 17.44 64.1 66.45 15.39 17.19 18.97 21.29 29.33 ISAETTCSDF ISAETTCSDFIRLL T HPSCPPIAL LILPNDL PAMQAEGIPI LILPNDL AVLLLL IKGAWS GDVAVVQP PPPPDIL EVVTPN NDNAPRF GLMPT QPDGPAIK AVLLLL MPPKSPDLIE DTESPTPTNSAVTI D ASNPESKVFYLK ASNPESKVFYLK EDVVAFKETP QKTSAEL KDFNP QVKSAKNEEPRVS VTVNSTPLIPDPD GGALIDY Amidated (C-term) Amidated (C-term) Amidated (C-term) Amidated (C-term) Amidated (C-term) Oxidation (M) Amidated (C-term) Amidated (C-term) Amidated (C-term) Amidated (C-term); Glu>pyro-Glu (N-term E) Amidated (C-term) Amidated (C-term) Amidated (C-term) 0.0000000.1 0.0000000000.1 0.0000000.1 0.000000.1 0.000000.1 0.00400.0 0.000000.1 0.0000000000.1 0.000000000000000. 1 3.0000000000.1 0.00000.1 0.000000000000000 00000000000.1 0.0000000.1 475 Eyestalk 1 gi|321462615|gb|EFX73637.1| gi|321462615|gb|EFX73637.1| gi|321462615|gb|EFX73637.1| gi|321462615|gb|EFX73637.1| gi|321462615|gb|EFX73637.1| gi|321462615|gb|EFX73637.1| Protein Accession # gi|321470570|gb|EFX81546.1| gi|321470570|gb|EFX81546.1| gi|321470570|gb|EFX81546.1| gi|321464766|gb|EFX75772.1| gi|321464766|gb|EFX75772.1| gi|321464766|gb|EFX75772.1| gi|321465056|gb|EFX76060.1| gi|321465056|gb|EFX76060.1| gi|321470448|gb|EFX81424.1| gi|321470448|gb|EFX81424.1| gi|321470448|gb|EFX81424.1| gi|321470448|gb|EFX81424.1| gi|321470568|gb|EFX81544.1| gi|321470568|gb|EFX81544.1| gi|321470568|gb|EFX81544.1| gi|321470568|gb|EFX81544.1| gi|321470568|gb|EFX81544.1| Protein Description hypothetical protein DAPPUDRAFT_57919 [Daphnia pulex] hypothetical protein DAPPUDRAFT_57919 [Daphnia pulex] hypothetical protein DAPPUDRAFT_57919 [Daphnia pulex] hypothetical protein DAPPUDRAFT_57919 [Daphnia pulex] hypothetical protein DAPPUDRAFT_57919 [Daphnia pulex] hypothetical protein DAPPUDRAFT_57919 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_322731 [Daphnia pulex] hypothetical protein DAPPUDRAFT_322731 [Daphnia pulex] hypothetical protein DAPPUDRAFT_323032 [Daphnia pulex] hypothetical protein DAPPUDRAFT_323032 [Daphnia pulex] hypothetical protein DAPPUDRAFT_323032 [Daphnia pulex] hypothetical protein DAPPUDRAFT_50104 [Daphnia pulex] hypothetical protein DAPPUDRAFT_50104 [Daphnia pulex] hypothetical protein DAPPUDRAFT_50104 [Daphnia pulex] Protein Mascot Score 31 31 31 31 31 31 35 35 35 26 26 26 19 19 15 15 15 15 25 25 25 25 25 17.4 16.31 16.39 20.36 20.53 21.64 31.06 Peptide Mascot Score 18.04 34.9 19.74 22.84 26.41 15.63 18.56 15.12 19.42 22.47 23.09 15.33 15.6 16.47 16.54 16.85 QIAEMQA SSPVPVAD EFPGPTESAPLPPP P ENEELEVLLAF GAGAGLIQSGKQ KIVRQNKL Peptide Sequence DPPNRS APPPPPPPPPPP APPPPPPPPPPP GQVSISN GQVSISN QGSVDSVVING GQVVEIL KILMLLDE QPMVQPI QPLVQPMMQPLM QPMMQP EGIQELPENVS LMATNMQS SHSEELAD EQLTTLLRM QPIRCSLVVGAK ISAETTCSDFIRLL T TQPLTTLLEE Amidated (C-term) Glu->pyro-Glu (N-term E) Amidated (C-term) Amidated (C-term) Peptide Variable Modifications Amidated (C-term) Amidated (C-term) Amidated (C-term) Gln->pyro-Glu (N-term Q) Oxidation (M) Amidated (C-term); Oxidation (M) Oxidation (M) Amidated (C-term) Amidated (C-term) Amidated (C-term); 2 Oxidation (M) Amidated (C-term) Amidated (C-term); Gln>pyro-Glu (N-term Q) 0.0000000.1 0.00000000.1 3.000000000000000. 0 0.00000000000.1 Position of Variable Modifications 0.000000.1 0.0000000.1 0.0000000.1 2.00000000000.0 0.00040000.1 0.0040000.0 0.000000000004000 000.0 0.00000000000.1 0.04000400.1 0.00000000.1 2.000000000000.1 0.0000000000.1 476 Eyestalk 2 Protein Accession # gi|321474231|gb|EFX85196.1| gi|321457271|gb|EFX68361.1| gi|321457271|gb|EFX68361.1| gi|321478348|gb|EFX89305.1| gi|321478348|gb|EFX89305.1| gi|321478348|gb|EFX89305.1| gi|321475268|gb|EFX86231.1| gi|321475268|gb|EFX86231.1| gi|321475268|gb|EFX86231.1| gi|321475268|gb|EFX86231.1| gi|321476878|gb|EFX87838.1| gi|321476878|gb|EFX87838.1| gi|321476878|gb|EFX87838.1| gi|321476878|gb|EFX87838.1| gi|321476878|gb|EFX87838.1| gi|321476878|gb|EFX87838.1| gi|321476878|gb|EFX87838.1| gi|321475051|gb|EFX86015.1| gi|321475051|gb|EFX86015.1| gi|321475051|gb|EFX86015.1| gi|321475051|gb|EFX86015.1| gi|321475051|gb|EFX86015.1| Protein Description hypothetical protein DAPPUDRAFT_301481 [Daphnia pulex] hypothetical protein DAPPUDRAFT_300356 [Daphnia pulex] hypothetical protein DAPPUDRAFT_313508 [Daphnia pulex] hypothetical protein DAPPUDRAFT_313508 [Daphnia pulex] hypothetical protein DAPPUDRAFT_313508 [Daphnia pulex] hypothetical protein DAPPUDRAFT_313508 [Daphnia pulex] hypothetical protein DAPPUDRAFT_313508 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308511 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308511 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308511 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308511 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303153 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303153 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303153 [Daphnia pulex] hypothetical protein DAPPUDRAFT_301481 [Daphnia pulex] Protein Mascot Score 26 15 15 31 31 31 29 29 29 29 15 15 15 15 15 15 15 20 20 20 20 20 25.83 Peptide Mascot Score 15.12 15.78 15.97 30.63 31.05 16.67 16.87 17.18 28.59 15.12 15.14 15.7 15.8 16.33 19.47 21.01 15.11 17.78 18.65 20.15 23.83 Peptide Sequence SSPDDNASPR SLFQSQNG SELEASYAKGLAK LASKLLKTTGELQ GPHGTVTAGWSA VAMKM LPGMTVVCTEA SLQLIR EMVVTT TSSSPGTRV KSILL TRVAA YSLIGGNTQ MDSLRPIRRETRD L LSGDVAVVQPLD NSVTLRLGDMDQ FITMAREPT CLRKE QLGGLP SCLRKE DPFVGPLVSVML AVRNFVCR KNVEVTVRVCN HLGSG KDLVAKH Peptide Variable Modifications 2 Oxidation (M) Glu->pyro-Glu (N-term E) Amidated (C-term) Oxidation (M) Amidated (C-term) Amidated (C-term) Position of Variable Modifications 0.000000000000000 00000000000000000 00000000404.0 3.000000.0 0.00000.1 0.000000000400.0 0.00000000000.1 0.0000000.1 477 gi|321478626|gb|EFX89583.1| gi|321478626|gb|EFX89583.1| gi|321478626|gb|EFX89583.1| gi|321478626|gb|EFX89583.1| gi|321478626|gb|EFX89583.1| gi|321478626|gb|EFX89583.1| gi|321478626|gb|EFX89583.1| gi|321474528|gb|EFX85493.1| gi|321474528|gb|EFX85493.1| gi|321470892|gb|EFX81866.1| gi|321470448|gb|EFX81424.1| gi|321470448|gb|EFX81424.1| gi|321469585|gb|EFX80565.1| gi|321469585|gb|EFX80565.1| gi|321469585|gb|EFX80565.1| gi|321464134|gb|EFX75144.1| gi|321464134|gb|EFX75144.1| gi|321464134|gb|EFX75144.1| gi|321474937|gb|EFX85901.1| gi|321474937|gb|EFX85901.1| gi|321474937|gb|EFX85901.1| gi|321470625|gb|EFX81600.1| gi|321470625|gb|EFX81600.1| gi|321470625|gb|EFX81600.1| hypothetical protein DAPPUDRAFT_314171 [Daphnia pulex] hypothetical protein DAPPUDRAFT_314171 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303213 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303213 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303213 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303213 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303213 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303213 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303213 [Daphnia pulex] hypothetical protein DAPPUDRAFT_49826 [Daphnia pulex] hypothetical protein DAPPUDRAFT_49826 [Daphnia pulex] hypothetical protein DAPPUDRAFT_49826 [Daphnia pulex] hypothetical protein DAPPUDRAFT_45411 [Daphnia pulex] hypothetical protein DAPPUDRAFT_45411 [Daphnia pulex] hypothetical protein DAPPUDRAFT_45411 [Daphnia pulex] hypothetical protein DAPPUDRAFT_323685 [Daphnia pulex] hypothetical protein DAPPUDRAFT_323685 [Daphnia pulex] hypothetical protein DAPPUDRAFT_323685 [Daphnia pulex] hypothetical protein DAPPUDRAFT_318494 [Daphnia pulex] hypothetical protein DAPPUDRAFT_318494 [Daphnia pulex] hypothetical protein DAPPUDRAFT_318494 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317013 [Daphnia pulex] 15 15 15 15 15 15 15 23 23 44 34 34 27 27 27 25 25 25 22 22 22 20 20 20 15.01 15.41 15.47 15.94 16.01 16.74 17.09 18.36 23.34 44.45 20 34.23 15.05 20.87 26.86 15.66 21.37 25.29 17.15 18.64 22.48 16.16 18.78 20.19 KSDLAWVAGKR QHPMCP SVLLMAI KGFTCMPA TTLRPTTT QHPMCP LTGRIADI LLTGLL FSPPAST GAELLSFSMALTI VCVWILTGHWLL M EGIQELPENVS QPAMQPTMQ PMMAPPLSM LVQQPKG LPVPMMAPP DLIKACTT LNLEDLITLP LIRIDRA ALSMIKTNFIVPLS S LRDSIIFPSSEH LVCNLGMAD WMMGTVLRT GDLEKKAAMEQK RTLTGDEANGH GDLEKKAAMEQK RTLTGDEANGH Oxidation (M) Oxidation (M) Amidated (C-term) Amidated (C-term); Oxidation (M) Amidated (C-term) Oxidation (M) Amidated (C-term) Amidated (C-term); 2 Oxidation (M) Amidated (C-term) Amidated (C-term) Amidated (C-term); Oxidation (M) Amidated (C-term) Oxidation (M) Amidated (C-term) 2 Oxidation (M) Amidated (C-term) 0.000400.0 0.000400.0 0.00000000.1 0.000000000000000 00000000004.1 0.00000000000.1 0.000400000.0 0.0000000.1 0.000044000.1 0.0000000000.1 0.0000000.1 0.000400000000000. 1 0.000000000000.1 0.000000400.0 0.044000000.0 0.000000000000000 00000000.1 0.000000000000000 00000000.1 478 gi|321477436|gb|EFX88395.1| gi|321477436|gb|EFX88395.1| gi|321477436|gb|EFX88395.1| gi|321477436|gb|EFX88395.1| gi|321477436|gb|EFX88395.1| gi|321477436|gb|EFX88395.1| gi|321477436|gb|EFX88395.1| gi|321477436|gb|EFX88395.1| gi|321477436|gb|EFX88395.1| gi|321470311|gb|EFX81288.1| gi|321470311|gb|EFX81288.1| hypothetical protein DAPPUDRAFT_242543 [Daphnia pulex] hypothetical protein DAPPUDRAFT_242543 [Daphnia pulex] hypothetical protein DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein DAPPUDRAFT_191611 [Daphnia pulex] 26 26 26 26 26 26 26 26 26 17 17 15.27 15.62 15.83 16.73 17.17 18.15 18.34 21.52 26.35 15.24 17.36 QDSSNALSIF IEEITAI ILEASPLL ILEASPLL QQQQPN GFLDKNRDT LLSFHKGD EELQRREE IGRIS AADLDHMPTMKE TKYL ELQEMNGMNLMG PP Amidated (C-term) Amidated (C-term) Amidated (C-term) Amidated (C-term) Amidated (C-term); Glu>pyro-Glu (N-term E) 0.0000000.1 0.000000.1 0.00000000.1 0.000000000000000 0.1 3.00000000000000.1 479 Protein Identifications from O. rusticus, Hypothetical Proteins from Daphnia pulex genome, PEAKS Brain 1 Accession gi|321472102 hypothetical protein DAPPUDRAFT_101007 [Daphnia pulex] gi|321470602 hypothetical protein DAPPUDRAFT_102305 [Daphnia pulex] gi|321466199 hypothetical protein DAPPUDRAFT_106344 [Daphnia pulex] gi|321466199 hypothetical protein DAPPUDRAFT_106344 [Daphnia pulex] gi|321464736 hypothetical protein DAPPUDRAFT_11648 [Daphnia pulex] gi|321465242 hypothetical protein DAPPUDRAFT_188571 [Daphnia pulex] gi|321477436 hypothetical protein DAPPUDRAFT_191611 [Daphnia pulex] gi|321477436 hypothetical protein DAPPUDRAFT_191611 [Daphnia pulex] gi|321477436 hypothetical protein DAPPUDRAFT_191611 [Daphnia pulex] gi|321473723 hypothetical protein DAPPUDRAFT_194402 [Daphnia pulex] gi|321472278 hypothetical protein DAPPUDRAFT_195011 [Daphnia pulex] gi|321471029 hypothetical protein DAPPUDRAFT_195819 [Daphnia pulex] gi|321467013 hypothetical protein DAPPUDRAFT_198141 [Daphnia pulex] gi|321460747 hypothetical protein DAPPUDRAFT_201492 [Daphnia pulex] gi|321477714 hypothetical protein DAPPUDRAFT_206384 [Daphnia pulex] gi|321457437 hypothetical protein DAPPUDRAFT_301440 [Daphnia pulex] gi|321478348 hypothetical protein DAPPUDRAFT_303153 [Daphnia pulex] gi|321478348 hypothetical protein DAPPUDRAFT_303153 [Daphnia pulex] gi|321478626 hypothetical protein DAPPUDRAFT_303213 [Daphnia pulex] gi|321478626 hypothetical protein DAPPUDRAFT_303213 [Daphnia pulex] gi|321468823 hypothetical protein DAPPUDRAFT_304375 [Daphnia pulex] gi|321468823 hypothetical protein DAPPUDRAFT_304375 [Daphnia pulex] gi|321468289 hypothetical protein DAPPUDRAFT_304927 [Daphnia pulex] gi|321467722 hypothetical protein DAPPUDRAFT_305142 [Daphnia pulex] gi|321477408 hypothetical protein DAPPUDRAFT_305633 [Daphnia pulex] gi|321466003 hypothetical protein DAPPUDRAFT_305989 [Daphnia pulex] gi|321464495 hypothetical protein DAPPUDRAFT_306711 [Daphnia pulex] gi|321460846 hypothetical protein DAPPUDRAFT_308707 [Daphnia pulex] gi|321460846 hypothetical protein DAPPUDRAFT_308707 [Daphnia pulex] gi|321460846 hypothetical protein DAPPUDRAFT_308707 [Daphnia pulex] gi|321478464 hypothetical protein DAPPUDRAFT_310476 [Daphnia pulex] gi|321475865 hypothetical protein DAPPUDRAFT_312745 [Daphnia pulex] gi|321475864 hypothetical protein DAPPUDRAFT_312746 [Daphnia pulex] gi|321475051 hypothetical protein DAPPUDRAFT_313508 [Daphnia pulex] gi|321475051 hypothetical protein DAPPUDRAFT_313508 [Daphnia pulex] gi|321475051 hypothetical protein DAPPUDRAFT_313508 [Daphnia pulex] gi|321474567 hypothetical protein DAPPUDRAFT_314077 [Daphnia pulex] gi|321470892 hypothetical protein DAPPUDRAFT_317013 [Daphnia pulex] gi|321470892 hypothetical protein DAPPUDRAFT_317013 [Daphnia pulex] gi|321470892 hypothetical protein DAPPUDRAFT_317013 [Daphnia pulex] gi|321470568 hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] gi|321470568 hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] Peptide FSSVGPS(-.98) SSPSLPPT RQPPPPPP TPSAAP IPAVVT AVLLLFPLNDKFESY DPGSPTQPN(-.98) NQPVISLIAKKPI RSPNPPGP ENLKPVYPCT TQYTIGSST VLVGSE TFTCVAKNSAGF PIPPLPDK TM(+15.99)EALEDTWRN TVPELNIT NGEDLAKA TDCGLLEGWLR HDGRKKNM(+15.99)FDAPIE SLGCNGSADQQTTV(-.98) DLMSIGMGKPA VGPSWGSPAA ELWINGKLTDY(-.98) KKWAEGDFK(-.98) VPEPARSPSPP PDSVAPLDM(-.98) SIFDSIK ADGEDAPPRP APPRPPPP APPRPPPP E(-18.01)SIASEVDEVY(-.98) QGVDLNR KKANLIKLYLTL HVFELN(-.98) LTPPDQEIM(+15.99)D(-.98) SIGGRG(-.98) VVLSHGENGM(+15.99)IY(-.98) GELIKHTENDH TVLAKHDEGG(-.98) VGNEPIVGM(+15.99)DN HGPIFRGIGP(-.98) LFQSGLL -10lgP 19.92 19.24 17.22 15.65 28.11 15.48 16.14 18.18 20.33 21.42 18.61 16.79 16.46 19.27 20.59 21.02 15.26 18.73 17.38 21.17 18.03 17.02 18.97 15.05 19.77 19.89 18.44 17.68 45.54 27.73 25.22 16.14 28.07 15.35 19.34 15.29 18.04 15.5 23.93 20.57 17.66 17.8 480 Brain 2 gi|321470568 gi|321470448 gi|321470448 gi|321470448 gi|321469585 gi|321477671 gi|321477671 gi|321477671 gi|321477376 gi|321470910 gi|321462615 gi|321477575 gi|321474610 Accession gi|321472102 gi|321470602 gi|321470602 gi|321470602 gi|321455473 gi|321477436 gi|321475531 gi|321473999 gi|321473723 gi|321473723 gi|321461543 gi|321473719 gi|321473719 gi|321461113 gi|321479055 gi|321461456 gi|321471172 gi|321471172 gi|321478348 gi|321478626 gi|321469744 gi|321469429 gi|321468905 gi|321477300 gi|321477408 gi|321466003 gi|321462341 gi|321461745 gi|321460934 gi|321460846 hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_318494 [Daphnia pulex] hypothetical protein DAPPUDRAFT_41019 [Daphnia pulex] hypothetical protein DAPPUDRAFT_41019 [Daphnia pulex] hypothetical protein DAPPUDRAFT_41019 [Daphnia pulex] hypothetical protein DAPPUDRAFT_4688 [Daphnia pulex] hypothetical protein DAPPUDRAFT_49564 [Daphnia pulex] hypothetical protein DAPPUDRAFT_57919 [Daphnia pulex] hypothetical protein DAPPUDRAFT_95322 [Daphnia pulex] hypothetical protein DAPPUDRAFT_98778 [Daphnia pulex] Description hypothetical protein DAPPUDRAFT_101007 [Daphnia pulex] hypothetical protein DAPPUDRAFT_102305 [Daphnia pulex] hypothetical protein DAPPUDRAFT_102305 [Daphnia pulex] hypothetical protein DAPPUDRAFT_102305 [Daphnia pulex] hypothetical protein DAPPUDRAFT_116167 [Daphnia pulex] hypothetical protein DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein DAPPUDRAFT_192770 [Daphnia pulex] hypothetical protein DAPPUDRAFT_194294 [Daphnia pulex] hypothetical protein DAPPUDRAFT_194402 [Daphnia pulex] hypothetical protein DAPPUDRAFT_194402 [Daphnia pulex] hypothetical protein DAPPUDRAFT_201000 [Daphnia pulex] hypothetical protein DAPPUDRAFT_209520 [Daphnia pulex] hypothetical protein DAPPUDRAFT_209520 [Daphnia pulex] hypothetical protein DAPPUDRAFT_216285 [Daphnia pulex] hypothetical protein DAPPUDRAFT_232322 [Daphnia pulex] hypothetical protein DAPPUDRAFT_290667 [Daphnia pulex] hypothetical protein DAPPUDRAFT_302612 [Daphnia pulex] hypothetical protein DAPPUDRAFT_302612 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303153 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303213 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303875 [Daphnia pulex] hypothetical protein DAPPUDRAFT_304111 [Daphnia pulex] hypothetical protein DAPPUDRAFT_304348 [Daphnia pulex] hypothetical protein DAPPUDRAFT_305524 [Daphnia pulex] hypothetical protein DAPPUDRAFT_305633 [Daphnia pulex] hypothetical protein DAPPUDRAFT_305989 [Daphnia pulex] hypothetical protein DAPPUDRAFT_307720 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308094 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308626 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308707 [Daphnia pulex] QASSFASS(-.98) LIATSPPP LVNAASGSLT TVVPGENHP PPM(+15.99)ALGLPVP ASALGDLI LLEKPNEP(-.98) TASVITLTSGPP DAGIFSC LSKIRQ(-.98) EFLQDAEGLVTLGLM(-.98) APAKDD(-.98) PKLPEPP Peptide QPWRGPTDVT APPPPPPPPPPP APPPPPPPPPPP QPDVRGAL NDPIYTR CNQTWRNEDPEAVA TPLGNSDIL(-.98) ESEAVAIRDSLAANSGFVKA(-.98) NADKRIT DGKPIPGSS(-.98) PLEAIDLYN DPTNLQSKLQ WINEHMITASSED(-.98) TAGIVGGLST SIPSVK(-.98) EVFLPLKNIK RIVIKMAAD RNSLLLAPMP M(+15.99)EDRLQSQQCDMA LLPGLK TPLFLALVRGDIKTLF PPPPKASAQLKAVQNA VVSAEGKRIQ(-.98) PATLKILGAH(-.98) SPVITEPVIR(-.98) VPIPDPM(+15.99)P RNSPASSPRLRT CRHNKSLSIVYK KDELM(+15.99)ILMQSM(+15.99) PPRPPPP 20.11 25.32 15.33 16.14 21.34 17.88 16.68 16.99 15.03 15.87 18.39 19.14 19.51 -10lgP 15.71 37.75 23.24 15.22 21.15 18.64 15.77 16.34 17.82 15.7 15.54 24.06 19.07 27.49 19.68 20.1 16.43 15.89 18.13 17.1 29.63 15.97 15.32 15.09 26.47 18.51 24.76 16.42 16.48 44.42 481 Eyestalk 1 gi|321460846 gi|321460846 gi|321460846 gi|321460846 gi|321460846 gi|321460261 gi|321468635 gi|321477347 gi|321476878 gi|321476878 gi|321476878 gi|321476878 gi|321475864 gi|321475051 gi|321475051 gi|321475051 gi|321474202 gi|321470892 gi|321470892 gi|321470892 gi|321470892 gi|321470568 gi|321470568 gi|321470568 gi|321470568 gi|321470448 gi|321470448 gi|321470448 gi|321470448 gi|321469372 gi|321466438 gi|321466438 gi|321465056 gi|321462130 gi|321458311 gi|321443655 gi|321477671 gi|321473455 gi|321470625 gi|321462615 gi|321460687 gi|321478212 gi|321474610 Accession hypothetical protein DAPPUDRAFT_308707 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308707 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308707 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308707 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308707 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308950 [Daphnia pulex] hypothetical protein DAPPUDRAFT_31061 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311561 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_312746 [Daphnia pulex] hypothetical protein DAPPUDRAFT_313508 [Daphnia pulex] hypothetical protein DAPPUDRAFT_313508 [Daphnia pulex] hypothetical protein DAPPUDRAFT_313508 [Daphnia pulex] hypothetical protein DAPPUDRAFT_313770 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317013 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317013 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317013 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317013 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_318481 [Daphnia pulex] hypothetical protein DAPPUDRAFT_321304 [Daphnia pulex] hypothetical protein DAPPUDRAFT_321304 [Daphnia pulex] hypothetical protein DAPPUDRAFT_322731 [Daphnia pulex] hypothetical protein DAPPUDRAFT_325390 [Daphnia pulex] hypothetical protein DAPPUDRAFT_329158 [Daphnia pulex] hypothetical protein DAPPUDRAFT_345055 [Daphnia pulex] hypothetical protein DAPPUDRAFT_41019 [Daphnia pulex] hypothetical protein DAPPUDRAFT_46980 [Daphnia pulex] hypothetical protein DAPPUDRAFT_49826 [Daphnia pulex] hypothetical protein DAPPUDRAFT_57919 [Daphnia pulex] hypothetical protein DAPPUDRAFT_59834 [Daphnia pulex] hypothetical protein DAPPUDRAFT_95253 [Daphnia pulex] hypothetical protein DAPPUDRAFT_98778 [Daphnia pulex] PPRPPPP APPRPPPP TPGAVDKLN VTAVQPGSPAA PPTLDPR SVVAVPHERIVG GIYLLCLGERDI(-.98) PAVPPRVSPSM(+15.99) EGYGNLR NNYVKNPVLFD(-.98) PAWSSQLP YAIYRTAGEILP(-.98) LPVSNILPV(-.98) PDSASVVQL DLMSNRSKMLT Q(-17.03)LESFT PPGTLALGDGS(-.98) VVIIPATT PEQLYMVTGRCEI(-.98) KTSLGM(+15.99)APT(-.98) SLGMAPTT VPPHGP YTSSTSVVAPPP(-.98) HVELPH QLLLACLAVFT M(+15.99)MQPLMQPMM(+15.99) AGTPPSSMMGAPL KGQTGLFP(-.98) PHM(+15.99)SPYIRFCSRQ DCPLTILGET IFTMIPYT LGVIPP IEIDFR ISKLLGL VDILLSM(+15.99)GISRN(-.98) GIQRLIISGIR(-.98) KTANADPSAP DM(+15.99)ISVIDM(+15.99)PPP(-.98) PAVLSATT QTLQGMKESGLK(-.98) SLGAAAIEAT IKGVFEGF EPIVGA(-.98) Peptide 26.36 40.76 21.66 15.79 15.31 15.4 22.32 18.55 22.15 16.47 16.35 15.87 16.97 21.79 20.49 16.83 18.02 31.31 17.6 16.8 15.66 25.21 21.56 21.26 19.77 25.75 19.63 17.61 15.51 17.8 28.74 19.5 18.09 24.57 19.57 20.31 17.16 18.96 18.41 18.74 19.16 17.25 17.78 -10lgP 482 gi|321472102 gi|321469722 gi|321466199 gi|321464736 gi|321477436 gi|321476638 gi|321471029 gi|321468642 gi|321467013 gi|321464553 gi|321461543 gi|321460747 gi|321460747 gi|321473719 gi|321468920 gi|321475130 gi|321472279 gi|321460227 gi|321461456 gi|321479333 gi|321458870 gi|321457271 gi|321471172 gi|321478626 gi|321478626 gi|321468905 gi|321468905 gi|321468289 gi|321464495 gi|321464495 gi|321464495 gi|321463123 gi|321461797 gi|321461745 gi|321461745 gi|321460934 gi|321460846 gi|321460846 gi|321460729 gi|321478464 gi|321476878 gi|321476878 gi|321476878 gi|321475051 hypothetical protein DAPPUDRAFT_101007 [Daphnia pulex] hypothetical protein DAPPUDRAFT_103233 [Daphnia pulex] hypothetical protein DAPPUDRAFT_106344 [Daphnia pulex] hypothetical protein DAPPUDRAFT_11648 [Daphnia pulex] hypothetical protein DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein DAPPUDRAFT_192312 [Daphnia pulex] hypothetical protein DAPPUDRAFT_195819 [Daphnia pulex] hypothetical protein DAPPUDRAFT_197424 [Daphnia pulex] hypothetical protein DAPPUDRAFT_198141 [Daphnia pulex] hypothetical protein DAPPUDRAFT_199303 [Daphnia pulex] hypothetical protein DAPPUDRAFT_201000 [Daphnia pulex] hypothetical protein DAPPUDRAFT_201492 [Daphnia pulex] hypothetical protein DAPPUDRAFT_201492 [Daphnia pulex] hypothetical protein DAPPUDRAFT_209520 [Daphnia pulex] hypothetical protein DAPPUDRAFT_224893 [Daphnia pulex] hypothetical protein DAPPUDRAFT_236642 [Daphnia pulex] hypothetical protein DAPPUDRAFT_239963 [Daphnia pulex] hypothetical protein DAPPUDRAFT_290661 [Daphnia pulex] hypothetical protein DAPPUDRAFT_290667 [Daphnia pulex] hypothetical protein DAPPUDRAFT_300115 [Daphnia pulex] hypothetical protein DAPPUDRAFT_300640 [Daphnia pulex] hypothetical protein DAPPUDRAFT_301481 [Daphnia pulex] hypothetical protein DAPPUDRAFT_302612 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303213 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303213 [Daphnia pulex] hypothetical protein DAPPUDRAFT_304348 [Daphnia pulex] hypothetical protein DAPPUDRAFT_304348 [Daphnia pulex] hypothetical protein DAPPUDRAFT_304927 [Daphnia pulex] hypothetical protein DAPPUDRAFT_306711 [Daphnia pulex] hypothetical protein DAPPUDRAFT_306711 [Daphnia pulex] hypothetical protein DAPPUDRAFT_306711 [Daphnia pulex] hypothetical protein DAPPUDRAFT_307445 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308055 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308094 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308094 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308626 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308707 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308707 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308744 [Daphnia pulex] hypothetical protein DAPPUDRAFT_310476 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311854 [Daphnia pulex] hypothetical protein DAPPUDRAFT_313508 [Daphnia pulex] QPIINSPHELEN(-.98) E(-18.01)SAAAQVNGRP(-.98) Q(-17.03)TTLAPQP(-.98) ERHLPGY PDAAQKRPAT E(-18.01)ADIVNR QGKMCVLDCSPN(-.98) DSAEGGAGPTTPA EIKKKPKVRK DYNDMKDL SGGAAGGP VSEANQGSNP NVAPAIQQQQ IDAASSSHE(-.98) APNEKPLQV QQDDELCFCKH(-.98) KAIFVDGGI HGQHVLSVPLTL(-.98) SLAVADM(+15.99)LM(+15.01) DPLGAPSLS(-.98) GPVPQGLV E(-18.01)ELVKPLK RPGAFAIY TPGGIK DKPTDEDIYNY(-.98) GDM(+15.99)PEPKY(-.98) EQLRDEVYCQ(-.98) PARVKDIPLQDM(+15.01) SIFDSIK SIFDSIK Q(-17.03)QPVANM(+15.99)TAS TDIM(+15.99)ESKS(-.98) PLNLILEPT SYVSVARSF DPGVAH(-.98) VSPAGVVNQSD NGHHPSQRDP KGGPAEG PQIGM(+15.99)VHP KNGGDIQMIKPSAFK PSPTPPHNMM(+15.01) E(-18.01)VM(+15.99)AGDGRSQPR DNVPVLND(-.98) TQWIPTPEKD(-.98) 19.73 16.82 16.74 16.14 15.79 19.21 20.4 21.41 15.54 21.12 17.15 15.24 19.99 19.97 18.4 17.68 16.34 34 20.39 17.99 25.02 22.55 19.22 26.31 15.37 16.55 15.99 16.55 26.72 19.72 18.2 16.65 17.45 16.39 18.62 18.95 15.63 22.16 15.03 15.43 15.95 16.74 17 16.39 483 Eyestalk 2 gi|321475051 gi|321475051 gi|321474567 gi|321470892 gi|321470706 gi|321470568 gi|321470448 gi|321469478 gi|321465056 gi|321465056 gi|321465056 gi|321465056 gi|321477671 gi|321477671 gi|321470910 gi|321470625 gi|321453462 Accession gi|321472102 gi|321472102 gi|321472102 gi|321466199 gi|321466199 gi|321464736 gi|321460217 gi|321455882 gi|321477436 gi|321477436 gi|321473999 gi|321477714 gi|321473719 gi|321473719 gi|321470311 gi|321470311 gi|321455555 gi|321460227 gi|321460227 gi|321460227 gi|321461456 gi|321458600 gi|321478348 gi|321478733 gi|321468905 gi|321468823 hypothetical protein DAPPUDRAFT_313508 [Daphnia pulex] hypothetical protein DAPPUDRAFT_313508 [Daphnia pulex] hypothetical protein DAPPUDRAFT_314077 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317013 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317361 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317679 [Daphnia pulex] hypothetical protein DAPPUDRAFT_318657 [Daphnia pulex] hypothetical protein DAPPUDRAFT_322731 [Daphnia pulex] hypothetical protein DAPPUDRAFT_322731 [Daphnia pulex] hypothetical protein DAPPUDRAFT_322731 [Daphnia pulex] hypothetical protein DAPPUDRAFT_322731 [Daphnia pulex] hypothetical protein DAPPUDRAFT_41019 [Daphnia pulex] hypothetical protein DAPPUDRAFT_41019 [Daphnia pulex] hypothetical protein DAPPUDRAFT_49564 [Daphnia pulex] hypothetical protein DAPPUDRAFT_49826 [Daphnia pulex] hypothetical protein DAPPUDRAFT_93775 [Daphnia pulex] Description hypothetical protein DAPPUDRAFT_101007 [Daphnia pulex] hypothetical protein DAPPUDRAFT_101007 [Daphnia pulex] hypothetical protein DAPPUDRAFT_101007 [Daphnia pulex] hypothetical protein DAPPUDRAFT_106344 [Daphnia pulex] hypothetical protein DAPPUDRAFT_106344 [Daphnia pulex] hypothetical protein DAPPUDRAFT_11648 [Daphnia pulex] hypothetical protein DAPPUDRAFT_15026 [Daphnia pulex] hypothetical protein DAPPUDRAFT_189558 [Daphnia pulex] hypothetical protein DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein DAPPUDRAFT_191611 [Daphnia pulex] hypothetical protein DAPPUDRAFT_194294 [Daphnia pulex] hypothetical protein DAPPUDRAFT_206384 [Daphnia pulex] hypothetical protein DAPPUDRAFT_209520 [Daphnia pulex] hypothetical protein DAPPUDRAFT_209520 [Daphnia pulex] hypothetical protein DAPPUDRAFT_242543 [Daphnia pulex] hypothetical protein DAPPUDRAFT_242543 [Daphnia pulex] hypothetical protein DAPPUDRAFT_262733 [Daphnia pulex] hypothetical protein DAPPUDRAFT_290661 [Daphnia pulex] hypothetical protein DAPPUDRAFT_290661 [Daphnia pulex] hypothetical protein DAPPUDRAFT_290661 [Daphnia pulex] hypothetical protein DAPPUDRAFT_290667 [Daphnia pulex] hypothetical protein DAPPUDRAFT_300857 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303153 [Daphnia pulex] hypothetical protein DAPPUDRAFT_303168 [Daphnia pulex] hypothetical protein DAPPUDRAFT_304348 [Daphnia pulex] hypothetical protein DAPPUDRAFT_304375 [Daphnia pulex] QSPVVQEVTSVLRE(-.98) PAILSALM(+15.99)K NIIAWISS VLAKHDEGGN PPPPGPPSF(-.98) GVQGLDE(-.98) LSFVAGEM(+15.99)VTII PSSGGPGAA(-.98) VLIAEPIEN(-.98) SGGGTPTNPTP LAGVALGGDIRYSGVTMLHPS GGGSGGGSGG(-.98) VM(+15.99)VTNVT QM(+15.99)MM(+15.99)LDQSKT ESPVPSLP QTCSMALNM(+15.99)KT KPIAWFDCNIH(-.98) Peptide LGLQNASLSTAT LPQPVTIRF(-.98) LPQPVTIRF(-.98) KEAIAQER QPPPPPPAPE(-.98) RDAHNLVLCF(-.98) IIQACRGNSP(-.98) E(-18.01)RKTDVTY AADLDHMPT IAKKPIGVLHLLD(-.98) QEMASNNSLV(-.98) IVDPNRDGY(-.98) LEEETRKAE(-.98) LVAASLD IVGPLSD(-.98) VDNEGRKV(-.98) LSRECPKPST HGQHVLSVPLTL(-.98) HGQHVLSVPLTL(-.98) LDLSQTAITN(-.98) LPKKKDGPCGSGSF(-.98) FLPLSDT(-.98) LLALVGGGAL(-.98) LLTASGGSF(-.98) TITAAHA E(-18.01)KLTGTQQQQ 15.9 17.11 15.55 17.43 23.52 26.79 17.82 19.76 19.18 16.08 17.8 16.53 18.87 26.94 16.47 18.96 18.08 -10lgP 16.8 22.57 16.22 17.36 19.15 19.48 21.01 18.14 16.46 24.4 15.34 20.54 15.6 20.67 18.05 18.61 34.5 21.24 16.61 15.36 19.98 15.25 15.55 17.42 15.54 21.05 484 gi|321468823 gi|321468823 gi|321468823 gi|321468220 gi|321468220 gi|321477408 gi|321464495 gi|321462341 gi|321461745 gi|321461569 gi|321475268 gi|321460846 gi|321460846 gi|321460846 gi|321460729 gi|321478464 gi|321478573 gi|321468635 gi|321477347 gi|321475051 gi|321470706 gi|321470568 gi|321470568 gi|321465056 gi|321465056 gi|321464134 gi|321458311 gi|321454896 gi|321477671 gi|321477671 gi|321477671 gi|321474247 gi|321470910 gi|321470570 gi|321470570 gi|321468494 gi|321460687 hypothetical protein DAPPUDRAFT_304375 [Daphnia pulex] hypothetical protein DAPPUDRAFT_304375 [Daphnia pulex] hypothetical protein DAPPUDRAFT_304375 [Daphnia pulex] hypothetical protein DAPPUDRAFT_304918 [Daphnia pulex] hypothetical protein DAPPUDRAFT_304918 [Daphnia pulex] hypothetical protein DAPPUDRAFT_305633 [Daphnia pulex] hypothetical protein DAPPUDRAFT_306711 [Daphnia pulex] hypothetical protein DAPPUDRAFT_307720 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308094 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308200 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308511 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308707 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308707 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308707 [Daphnia pulex] hypothetical protein DAPPUDRAFT_308744 [Daphnia pulex] hypothetical protein DAPPUDRAFT_310476 [Daphnia pulex] hypothetical protein DAPPUDRAFT_310476 [Daphnia pulex] hypothetical protein DAPPUDRAFT_31061 [Daphnia pulex] hypothetical protein DAPPUDRAFT_311561 [Daphnia pulex] hypothetical protein DAPPUDRAFT_313508 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317361 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_317519 [Daphnia pulex] hypothetical protein DAPPUDRAFT_322731 [Daphnia pulex] hypothetical protein DAPPUDRAFT_322731 [Daphnia pulex] hypothetical protein DAPPUDRAFT_323685 [Daphnia pulex] hypothetical protein DAPPUDRAFT_329158 [Daphnia pulex] hypothetical protein DAPPUDRAFT_332604 [Daphnia pulex] hypothetical protein DAPPUDRAFT_41019 [Daphnia pulex] hypothetical protein DAPPUDRAFT_41019 [Daphnia pulex] hypothetical protein DAPPUDRAFT_41019 [Daphnia pulex] hypothetical protein DAPPUDRAFT_45861 [Daphnia pulex] hypothetical protein DAPPUDRAFT_49564 [Daphnia pulex] hypothetical protein DAPPUDRAFT_50104 [Daphnia pulex] hypothetical protein DAPPUDRAFT_50104 [Daphnia pulex] hypothetical protein DAPPUDRAFT_52222 [Daphnia pulex] hypothetical protein DAPPUDRAFT_59834 [Daphnia pulex] LSQSNGSNPR QCISTVSGQQP(-.98) SSPARPVV GVQALQNTYG HDLPAEYDEM(+15.99)LRVM(+15.01) VPAQTQQHIA(-.98) SIFDSIK CNILKARA(-.98) KAGCEPDEE(-.98) ETAINALAKDVV PTATASGPPSAT IVIISHRAAA(-.98) KNEKKKKK LLGAVGGSIGGPGQAG(-.98) M(+15.99)ARAIGT HQM(+15.99)EMYIDMSPALKSF(-.98) LLYTLSA LAVSTSEVSLL(-.98) SPTGSVASPG SGGGGGLGV LTERIGIFPI LNKLMHNM(+15.99)GSP(-.98) RLTITGR AAAAAAPTPKKR(-.98) AAAAAQSGHPSSL LNLEDLITLP(-.98) E(-18.01)IAATWIFD(-.98) SVVVRSEKGRKC(-.98) LPTLQGLATVAH(-.98) LSADGSSFV(-.98) VM(+15.99)VTNVT LQM(+15.99)YAH(-.98) E(-18.01)APSDTM(+15.99)SE(-.98) ISPVEN(-.98) LPPTPAQP IGDKNHPKF LGGPVD(-.98) 21.76 23.48 15.05 15.38 19.71 17.7 29.26 15.05 16.15 15.03 15.27 16.22 15.54 17.56 19.28 17.06 21.65 17.61 16.99 35.28 20.26 21.12 18.29 21.66 20.24 19.16 18.2 15.69 19.81 17.14 33.17 15.39 23.45 16.68 18.03 17.36 23.92 485