Periodontal Disease and Diabetes Mellitus
Transcription
Periodontal Disease and Diabetes Mellitus
{i} C Under the aegis of EPHORB MEDLINE INTERNATIONAL Pvt. Ltd. ® Registered Office – 12/44, Vasundhara, Ghaziabad, Delhi NCR, India 201012 Website: http://www.idjsr.com Email:[email protected] Publication, Subscription, Printing, Distribution and Advertisement handle by Innovative Publication: NNOVATIVE PUBLICATION H - 2 / 94, Bengali Colony, Mahavir Enclave, Part - 1, New Delhi - 110045, India Ph.: 91-11-25052216, Telefax: 011-25051061 Mo: +91-8826373757, 8527826746, 8826859373 Website: www.innovativepublication.comEmail: editor@innovativepublic ation.com, [email protected] I Dr. Ankur Rustagi, MDS (OMFS) (AIIMS) - India. Dr. Pankaj Dhingra, BDS, DDS - USA Dr. Anil Chandra, BDS, MDS (KGMU) - India Dr. Zeeshan Sheikh, BDS, Dip Dental Hygiene, M.Sc (Dental Materials), PhD - Canada Dr. Ameet Vaman Revankar, BDS, MDS (Ortho) - India Dr. Jitendra Sharan, BDS, MDS (Ortho), PhD (Ortho) (AIIMS) India Dr. Pravesh Bizenia, BDS, MDS (Prostho) (AIIMS) - India Dr. Sumeet Grover, BDS, MDS (Ortho) - Russia Subscription Information: A subscription to International Dental Journal of Student’s Researchcomprises four issues per year. Prices include postage. Annual Subscription rate for Institutional is INR 5000/- and Individual INR 3000/- and International Institutional Price US$ 200 and Individual US$ 125 including all postal exp. Free online access with print subscription. The amount shall be remitted as Cheque/DD/online transfer in favour of “INNOVATIVE PUBLICATION” Kotak Mahindra Bank, Branch: Harpool Singh Market, Main Dwarka Janakpuri Road, New Delhi – 110045, Account No: 7911500308, IFSC Code: KKBK0000177, MICR Code: 110485008 Dr. Mahesh Sadhnani, BDS, MPH, DMD - USA Founders Dr. Kumar Anshul – Manipal College of Dental Sciences, India Dr. Harsh Rajvanshi - I.T.S Dental College, India Dr. Ayesha Zaka - Margalla College of Dentistry, Pakistan Dr. Joharia Azhar Saadat, BDS, MSc (Oral Path) (London), Mphill (NUST) – Pakistan Indexed In Index Copernicus Genamics Journal Seek Ulrichsweb Global Serial Dictionary Directory of Open Access Journals (DOAJ) DRJI (Directory of Research Journals Indexing) IIFS (International Impact Factor Services) ResearchGate.net GIGA (German Institute of Global and Area Studies) The Hong Kong Polytechnic University Scientific Indexing Services Dr. Kamran Habib Awan, BDS, Ph.D (Oral Medicine) - UK Editorial Board Dr. Kumar Anshul - Editor In Chief Dr. Harsh Rajvanshi - Executive Editor Dr. Ayesha Zaka - Executive Editor Communications and Associate Editor Dr. Paridhi Kalia, University of Louisville School of Dentistry, USA Cover Art By Dr. Ebadullah Shafi, Design and Graphics Incharge- RAK College of Dental Sciences, UAE Reviewers Panel Dr. Gregori Kurtzman, DDS, Fellow in the AGD, AAIP, ACD, ICOI, PFA, ADI. Mastership - AGD and ICOI. Diplomat- ICOI, ADIA – USA Dr. Saurabh Lall, BDS, MDS (Perio), Fellow PFA - India Dr. Rajeev Chitguppi, BDS, MDS (Perio) - India Dr. Thomas Thong Nguyen, DMD, AEGD, MSc Dental SciencesCanada Dr. Ashith B. Acharya, BDS, GDFO, Forensic Odontologist - India Dr. Mohamed-Nur Abdallah, BDS, M.Sc Dental Sciences - Canada Dr Swati Pradeep Patel, BDS, MDS (Perio), PhD (Perio) - India Dr. Emmanuel J. N. L. Silva, M.Sc (Endo), PhD (Endo) - Brazil Dr. Jaime Díaz, DDS, M.Sc - Chile Dr. Malay Kumar, BDS, MDS (Oral Path) - India Dr. Fahim Ahmed Vora , MDS (Prostho), M Clin Dent (Prostho), MFDS, RCPSG - UK Dr Haby Mathew Somson, BDS. MDS (Prostho), Dip. in Laser (Geneva) - India Dr. Suresh Shenvi, BDS, MDS (Endo) - India Dr. Satpreet Singh, BDS, MPH (USA) - India Dr. Ramesh Kumaresan, BDS, MDS (OMFS) - Malaysia Dr. Ala' Ersheidat, BSc, BDS, JB (Perio) – Kingdom of Jordan Dr. Meena Jain, BDS, MDS (PHD), PhD (Dent. Sci.) India Dr. Zohaib Khurshid, BDS, MRes in Biomaterials - UK Dr. Shivani Kohli, BDS (MAHE), MDS (Prostho) - Malaysia Dr. Naresh Sharma, BDS, MDS (Pedo), MADA (USA) - India Dr. Christopher Majdi, MSHCA, CHBC, CBA - USA Dr. Mohammed Jasim Al-juboori, BDS, M.Sc (Implant Specialist), Fellow ICOI - Iraq Dr. Stefan Theodor Serban, DMD, MPH- Netherlands Dr. Qian Wang, DMD, Ph.D (China), Post-Doctoral Fellow- USA Dr. Juan Carlos Munévar, MSc (Oral Biology) - Colombia Dr. Shivlal Vishnoi, BDS, MDA (Perio) - India Dr. Manu Dhillon, BDS, MDS (OMDR) - India Dr. Shaveta Kaushal, MDS, MIPS (Prostho) - India Dr. Narayan H Gandedkar, MDS (Ortho), FCFO - Taiwan Dr. Ujjwal Pyakurel, BDS, MDS (Ortho) - Nepal Dr. Ricardo Machado, Masters in Endo, PhD (Endo) - Brazil Dr. Muhammad Sohail Zafar, BDS, PhD (UK), MSc (UK), F.I.C.D (USA), F.A.D.I (USA) – Kingdom of Saudi Arabia Dr. Mohamed El-Massry, MBBCh, MSc (Gen Surgery), BDS, PhD (Oral, Plastic and Maxillofacial Surgery) - Egypt International Dental Journal of Student’s Research, January – March 2015:3(2) ii VOLUME III ISSUE II SPECIAL EDITION ON PERIODONTOLOGY INDEX S.No. 1. 2. 3. TITLE Title Page A Foreword GUEST EDITORIALS – Dr. Zeeshan Sheikh & Dr. Zohaib Khurshid Page No. 4. EDITORIAL – FOUNDERS 42 5. ORIGINAL RESEARCH In-vitro properties of calcium phosphate cement as a bone grafting material 43-45 40 41 6. Dr. Ashar Jamelle, Professor Robert Hill, Dr. David Gillam SHORT COMMUNICATION Natural and synthetic hydrogels for periodontal tissue regeneration 46-48 7. Dr. Marco Laurenti CLINICAL CASE REPORT Bone fenestration: A case report of management of a lower anterior buccal bone fenestration 49-51 8. Dr. Murai Khalifa CLINICAL DIAGNOSIS & GUIDELINES A Histological and Clinical Evaluation of Shallow and Deep Probing Depths 52-55 9. Siavash Hassanpour, Shlomi Tamam , Timur Shigapov CLINICAL DIAGNOSIS & GUIDELINES Prevention of mandibular nerve injury associated with dental implant placement: preoperative and intraoperative recommendations 56-57 10. Thomas T. Nguyen CLINICAL CASE REPORT Periodontal Disease and Diabetes Mellitus: Case Report 58-65 11. H Marie Keeling, Tamara L. Wright SYSTEMATIC REVIEW Sinus lift grafting materials and immediate implant placement: A systematic review 66-71 12. Dr. Kashif Hafeez, Dr. Aiyesha Wahaj, Dr. Muhammad Sohail Zafar , Dr. Sana Shahab SYSTEMATIC REVIEW Laboratorial and clinical impacts of tobacco on periodontal health: A systematic review 72-78 13. Fahad Sikander Khan, Aisha Aziz , Dr. Sana Shahab, ,Dr. Muhammad Sohail Zafar SHORT COMMUNICATION A review of the use of laser in periodontal therapy 79-82 14. Dr. Amir Manzoor Shah , Dr. Khurram Khan , Dr.Fahd Ahmed , Dr.Nida Amir ORIGINAL RESEARCH Prevalence of bleeding gums while tooth brushing among betel nut chewers vs non betel nut chewers in school going children 83-87 15. Dr. Syed Misbahdduin, Dr. Mansoor Ul Aziz, Dr. Asma Fazal, Dr. Tayyaba Khairuddin, Dr. Safia Khairuddin SHORT COMMUNICATION Short communication: Stem Cells for Periodontal Tissue Regeneration 88-92 16. Dr. Mohamed-Nur Abdallah, Dr. Mai S. Ali CLINICAL DIAGNOSIS & GUIDELINES Periodontal Disease in Immunodeficient Patients: Clinical Guidelines for Diagnosis and Management. 93-104 17. Morvarid Oviesi, Oriyah Barzilay, Ahmed A. Hanafi CLINICAL CASE REPORT Autogenous Connective Tissue Graft for the Treatment of Localized Gingival Recession: A Case Report 105-108 Dr Zohaib Akram, Dr Haroon Rashid, Dr FahimVohra International Dental Journal Of Student’s Research, April-June 2015;3(2) {40} Dr. Nader Hamdan BDS, MSc Resident - Division of Periodontics Department of Diagnostic and Surgical Sciences College of Dentistry, Faculty of Health Sciences University of Manitoba, Canada Email: [email protected] It is very hard to find somebody who does not agree that periodontology and its related sciences were the focus of dental research in the last few decades thanks to its unique and special mix of biology, basic sciences, medicine, and surgery. Interest in periodontology as a science and periodontics as a clinical practice was heightened even more after the proven and suggested links between periodontal disease and a host of relatively common systemic conditions, such as cardiovascular diseases, diabetes, arthritis, pneumonia, Alzheimer’s disease and gestation outcome complications among others. Therefore, it was not surprising to dedicate this issue of the International Dental Journal of Students' Research (IDJSR) to periodontology in order to shed more light on what do university academics and their trainees contribute to this interesting and quickly progressing field. This periodontology issue, which is the first special edition of IDJSR, includes studies originating from reputable university research centers in the east and the west (Malaysia, Pakistan, Ireland, UK, USA, and Canada) offering different perspectives and providing different levels of evidence ranging from case reports to systematic reviews in addition to reports of original research and short communications. The material presented covers appealing topics and addresses important questions of clinical relevance adding value to this issue through offering knowledge that could be directly translated into clinical practice. Since it was first established, IDJSR took big strides towards achieving its goal as the students’ committed researchdissemination hub and their exclusive vehicle to present scientific work and share intellectual output in the most pertinent topics. This special edition on periodontology is just another example of that commitment and will mark an important step forward for the IDJSR. I hope you enjoy this issue as much as I enjoyed going over its versatile material. I would like to extend special thanks to the authors who participated. My thanks also go to the guest editor Dr. Zeeshan Sheikh who devoted this edition of the IDJSR to periodontology and honored me with the kind invitation to provide the “foreword section” as my humble contribution! Best Wishes! International Dental Journal of Student’s Research, April - June 2015;3(2):40 {41} GUEST EDITORIAL Dr. Zeeshan Sheikh – Guest Editor Matrix Dynamics Group. Faculty of Dentistry, University of Toronto. 150 College Street, Room 222.Toronto, ON, Canada. M5S 3E2 Website: http://matrixdynamics.ca E-Mail: [email protected] Dear Colleagues, It is an honor for me to be the guest editor for the periodontology special edition issue by the IDJSR. The primary purpose of this special edition is to present the research and clinical case reports in the field of periodontology in order to stimulate a critical analysis and a discussion on the future of the periodontal practice and research. The initiatives being taken by this journal paves the way for dental students/clinicians/researchers from all over the world to contribute and share their research. I can only see this journal go from strength to strength and I wish the whole team of IDJSR the very best and look forward to being associated with them in the future. We know from the current data from USA alone that about 46% of the population has some form of periodontitis. This when coupled with an increase in life-expectancy leading to an aging population, means that the importance of managing and treating periodontal disease is more important than ever. There is a need to develop more effective strategies for periodontal therapy to achieve optimal outcomes with high levels of clinical success. Most of the activities within the field of periodontology are largely focused on the development of implant placement techniques, implant-related materials and state-of-the-art implant systems. As dentists, we treat patients by acknowledging the importance of prevention by maintenance of oral hygiene, early diagnosis and intervention for appropriate management of periodontal health. The same approach must be adopted for implant patients and a risk assessment should be performed. Presence of different medical conditions, susceptibility to periodontal disease and use of certain drugs can limit the success of implants. Implant loss, mucositis, marginal bone loss and periimplantitis are the more common types of complications reported in implant therapy. With the increasing number of implants being placed currently, it can be expected that the frequency with which we encounter peri-implant lesions will increase as well. There are constant advances being made in periodontal regenerative therapy, immunoregulation, vaccination and community based approach to prevention. The knowledge and technology that is available today makes it an exciting time in the field of periodontology. It is imperative that we decide wisely how these professionals aids are going to be of benefit to our patients. Dr Zohaib Khurshid – Co Guest Editor B.D.S, MRes in Biomaterials (Student) Cert. in Aesthetic Dentistry (Kings College London) Cert. in Laser Dentistry (BACD, UK) Senior Lecturer Department of Dental Materials and Oral Biology Altamash Institute of Dental Medicine Karachi, Pakistan Consultant Dental Advisor Paramount Book Publisher, Pakistan Email: [email protected] It is extremely difficult to ignore the research done on periodontal tissue in relation to its physiology and pathology, diagnosis, epidemiology and prevention and therapy of periodontal disease. In present scenario where almost half of the population is effected by some level of periodontal problem it is difficult to its importance. Therefore special volume of this journal emphasises on publishing the original contributions of high merit from different part of world. International Dental Journal of Student’s Research, April - June 2015;3(2):41 {42} EDITORIAL Dr. Kumar Anshul Dr. Harsh Rajvanshi Dr. Ayesha Zaka Founder & Editor In Chief Manipal College of Dental Sciences, India Founder & Executive Editor I.T.S Centre for Dental Studies & Research, India Founder &Executive Editor Margalla College of Dentistry Pakistan “My family is like a sanctuary to me.I turn to them for support and strength. I take comfort in knowing no matter which path I choose, my family stands behind me.” Anonymous Family is our support system. We rely on our family for everything – they are our pillars of strength. Same way for the tooth – the periodontal structures are its family. No matter how much stress the tooth bears, family backs it up. The care of these periodontal structures is necessary, for without it; the tooth cannot stand alone. This is the era of Periodontology and as promised, the editorial board of IDJSR brings to you the dessert from the Editorial Kitchen – The Special Issue on Periodontology. This delicacy is topped with creamy articles with ingenious stuffing. With modernization, focus is being shifting to more prophylactic and conservative approaches &Periodontics deals with the very same concept. In this issue, we have two great researchers – Dr. Zeeshan Sheikh as the Guest Editor and Dr. Zohaib Khurshidas the Co-Guest Editor who have kindly worked with us on this issue. We have articles from around the world - Malaysia, Pakistan, Saudi, Canada, Egypt, Ireland, UK, USA with the stalwarts sharing their experiences with us. Readers, authors and peers – We shall be coming up with another regular issue hot off the press in the month of August, 2015. The call for papers is open! At IDJSR, we have always strived to bring something fresh and new to the field of Dental Literature. We hope that this issue helps both the clinicians and researchers at large. Happy Reading! International Dental Journal of Student’s Research, April – June 2015;3(2):42 {43} ORIGINAL RESEARCH In-vitro properties of calcium phosphate cement as a bone grafting material Dr. Ashar Jamelle1, Prof. Robert Hill2, Dr. David Gillam3, 1 Masters clinical Periodontology, Queen Mary University London, UK and Department of Periodontology, Fatima Jinnah Dental Hospital, Karachi, Pakistan 2 Professor of Physical Sciences in Relation to Dentistry Institute of Dentistry Dental Physical Sciences Unit 3 Clinical Senior Lecturer in Periodontology (Specialist in Periodontology) Queen Mary University London, UK Corresponding author: Dr. Ashar Jamelle Email: [email protected] Access this Article Online www.idjsr.com Use the QR Code scanner to access this article online in our database Article Code: IDJSR SE 0160 Quick Response Code Abstract In 1980’s researchers discovered CPCs (calcium phosphate crystals) which are a bioactive and biodegradable bone grafting material. Phases form after mixing in different compositions with different end products which are mainly two types; Brushite, and a Apatite. Bioactive glass can undergo dissolution in physiological solutions and form a hydroxycarbonated apatite like phase (this includes Octacalcium or Flouroapatite). Novel material can be made by mixing bioglass and Ca(H2PO4)2 and have cements set to form hydroxyapatite or brushite produce HAP, brushite and fluorapatite forming cements. The aims and objectives of this study were to investigate the influence of storage media on the Calcium Phosphate Cements combined with bioactive glass, with respect to properties and phase formed and strength of development. Would the outcomes of storing in a media enriched in calcium and phosphate (that more closely mimics the in vivo conditions, Simulated body fluid) and Tris buffer solution. To See histological and structural that do invivo implanted cements show the formation of more hydroxyapatite and higher mineral contents. To determine mechanical properties does it result in higher compressive strength. Functionally does it aid the conversion of Octacalcium phosphate to Hydroxyapatite. Calcium phosphate was measured 0.98 gms and mixed with bioactive glass 1.02 gms and placed in a 6 by 4 Cylinder, placed in aoven. Cements were immersed into both TRIS buffer and Simulated body fluid solution for 1hour, 1day, 7days and 28 days .The compressive strength was determined by a Instron machine. Characterization of phases was seen by analysis through FTIR(spectoscopy) and X-ray diffraction microtomography (determine quantitative measurements of mineral concentration in hard tissue). It was seen that the storage media does have a influence in properties and phases formed. Keywords: Calcium phosphate; Brushite; Apatite; Bioactive glass; TRIS buffer; Simulated body fluid. Introduction One of the prerequisites of periodontal regeneration is the formation of bone. Bone grafting is possible because the bone tissue, unlike many other tissues, has the ability to regenerate completely if there is sufficient space to grow into. As bone grows, it will generally replace the graft material completely, and result in a totally integrated region of new bone. The biological mechanisms providing a rationale for bone grafting are the following osteoconduction, osteoinduction, osteopromotion and osteogenesis.1 Osteoconduction phenomena occurred when the bone graft material served as a scaffold for new bone growth and therefore it was perpetuated by the native bone. Osteoblast from the margin of the defect being grafted utilized the bone graft material as a framework upon which to spread and to synthesize new bone.1 Bone grafting is a surgical procedure that was performed to replace the missing bone with a material from a patient's own body, an artificial, synthetic, or a natural substitute. Bone grafting was possible when the bone tissue had the ability to regenerate completely if sufficient space was International Dental Journal of Student’s Research, April-June 2015;3(2):43-45 {44} provided into which the bone can grow. As natural bone grows, it can generally replace the graft material completely, and result in a fully integrated region of new bone. Classification of the bone grafts based on various material groups are2 Allograft-based bone graft entails allograft bone, can be used alone or incombination with other materials (e.g., Grafton®, OrthoBlast®). Factor-based bone graft are natural and recombinant growth factors, which are used alone or in either in merging with other materials, such as transforming growth factor-beta (TGF-beta), Platelet-derived growth factor (PDGF), fibroblast growth factors (FGF), and bone morphogenetic protein (BMP). Cellbased bone grafts use cells to generate new tissue alone or are added onto a support matrix, for example, haemapoetic stem cells. Ceramic-based bone graft substitutes which include material likes calcium phosphate, calcium sulphate, and Bioglass used alone or in combination; for example, OsteoGraf®, ProOsteon®, OsteoSet®. Polymerbased bone graft used degradable and nondegradable polymers alone or in combination with other materials, for example, open porosity polylactic acid polymers Flexible hydrogel-hydroxyapatite (HA) composite has a mineral to organic matrix ratio, which approximates that of the human bone. Artificial bone can be created from ceramics, like calcium phosphates (e.g., HA and tricalcium phosphate), bioglass, and calciumsulphate are biologically active depending on the solubility in a physiological environment 3Alloplastic grafts can be manufactured from hydroxyapatite, and was a naturally occurring mineral (a main mineral component of bone), made from bioactive glass. Hydroxyapatite was a synthetic bone graft, which was commonly used now due to its properties e.g., osteoconduction, hardness, and acceptability by bone. Calcium orthophosphates have been studied as bone repair materials for the last 80 years. Calcium phosphates are part of a group of bioactive synthetic materials and the most frequently used are the hydroxyapatite and the tricalcium phosphate materials. They are commonly used due to their osteoconductivity, crystallographic structures, and chemical composition similar to the skeletal tissue. They are therefore classified according to their 'resorbability' which was that extent of degradation in vivo. Hydroxyapatite in turn hasbeen described as “non resorbable” and tricalcium phosphate has been described as “resorbable”4,5. Calcium phosphate materials demonstrate a positive interaction with living tissue that included also the differentiation of the immature cells towards bone cells5,6. Calcium Orthophosphate cements (CPC) have been reported to form two major end products: a precipitated poorly crystalline HA or CDHA and DCPD (also called ‘‘brushite’’) and apatite cements. The final setting product of the cements was of the paramount importance as this would determine the solubility and, therefore, the in vivo bioresorbability. The main difference between the two cement types was the solubility of the end-product: brushite was 1–2 orders of magnitude more soluble than apatite’s at a physiological pH and therefore brushite CPCs normally resorb faster than that of apatite. It has numerous properties which include osteotransductive, e.g., after implantation calcium orthophosphate cements may be replaced by new bone tissue and was osteo-conductive. Calcium phosphate [Ca(H2PO4)2] cements (CPC) have been used for the treatment of non-weight bearing bone fractures or defects. Bioactive glass has also been used in dentistry as a bone substitute and a number of therapeutic agents may be incorporated into the glass structure, for example fluoride, strontium, chlorine etc. Bioactive glass (BG) has been reported to form hydroxycarbonate apatite (HCA). These materials also exhibit excellent Osseo-integration with bone and therefore were originally developed for applications in bone regeneration. Strontium has been reported in vitro and in vivo to enhance the replication of pre-osteoblastic cells and decreases the activity and the number of osteoclasts. The intake of these strontium containing drugs lead to a greater deposition of calcium in bone and DNA and bone collagen synthesis are enhanced7. Novel materials may also be formulated by mixing a bioglass composition with CPC in order to improve the physical properties. The ideal outcome of the new product was to utilize the bioactivity and resorbability of a bioactive glass with the added clinical advantages of in situ setting and extrudability of CPCs. Strontium can be added to bioglass to some samples in 25% ratio as that enhances bone formation. The aim of the present study was to investigate the influence of a storage media (Tris buffer solution and Simulated Body Fluid (SBF) on a modified CPC combined with a Bioactive glass composition, with respect to both its properties (e.g., compressive strength) and the phase formed (e.g., conversion of Octacalcium phosphate [OCP] to Hydroxyapatite [HA]) and compressive strength Materials and Methods To investigate the influence of storage media on the Calcium Phosphate Cements, with respect to properties and phase formed andstrength of development).What would be the outcomes of storing in a media enriched in calcium and phosphate (that more closely mimics the in vivo conditions, e.g., Simulated body fluid) Structural; Do in-vivo implanted cements show the formation of more International Dental Journal of Student’s Research, April-June 2015;3(2):43-45 {45} hydroxyapatite and higher mineral contents. Mechanical; does it result in a higher compressive strength of the new material calcium phosphate and bioactive glass.Functional; does it aid the conversion of Octacalcium phosphate to Hydroxyapatite. The CPC/Bioglass composition was formulated by measuring 0.98 gm of calcium phosphate and mixing with 1.02 gm. of bioactive glass. The cement paste was mixed and packed into 6 by 4 cylindrical steel moulds and placed in an incubator at 370c for 120 minutes. The cylinders were removed from the moulds and immersed in 50 ml of either TRIS buffer solution or SBF at 370c for 1hour, 24 hours, 7 days and 28 days The testing of the compressive strength (Mpa) of the samples (n=8) was by an Instron universal testing machine type 5567 and characterization of the different phases of the samples was by FTIR spectrum and X-rayDiffraction in order to determine the quantitative measurements of the mineral concentration in hard tissue. Fig 2: XRD of the 25% Sr cement after immersion in SBF Very similar behaviour was observed as for the 0% Sr glass. However the OCP peak at 4.7o two theta had now completely disappeared after four weeks of immersion and the diffraction lines for apatite were slightly sharper. This indicated that the 25% Sr cement was converting from OCP to hydroxyapatite more rapidly. Results Fig 1: X-Ray Diffraction(XRD) of the 0% Sr cement after immersion in SBF : shows as a function of time had a XRD pattern that matched that of hydroxyapatite but in addition it also had a sharp diffraction peak at 4.7 degrees two theta which corresponded to the water layer in octacalcium phosphate (Ca8(PO4)6 H2.5H2O (OCP) which was thought to be a precursor to hydroxyapatite(HA) formation in the biomineralisation of tooth and bone 8 .The characteristic diffraction lines for apatite increased for longer immersion times and the diffraction line at 4.7 degree two theta characteristic of OCP decreased with immersion time. Fig 3: XRD of the 0% Sr cement after immersion in Tris The phase development and phases present in the 25%Sr cement immersed in Tris buffer were similar to the phases observed on immersion in SBF. Fig 4: XRD of the 25% Sr cement after immersion in Tris: The phase development and phases present in the 25%Sr cement immersed in Tris buffer were similar to the phases observed on immersion in SBF. International Dental Journal of Student’s Research, April-June 2015;3(2):43-45 {46} Fig 5: Shows the compressive strength for the cements after immersion in SBF. The cements exhibit an increasing compressive strength on immersion in SBF from 1 hour to 24 hours but then a marked decrease from 24 hours to 168 hours. This contrasted markedly with the behaviour found on immersion in Tris buffer. The reduction in the compressive strength has been associated with the conversion of OCP to HA for conventional; calcium phosphate cements. The 4.7o two theta line of OCP was elucidating and the results would suggest that immersion in Tris buffer compared to immersion in SBF favoured the conversion of OCP to hydroxyapatite. It was also observed that the presence of calcium in SBF does not accelerate the conversion process as expected and in fact the conversion was slower in SBF compared to Tris buffer. In the compressive strength data cements exhibited an increasing compressive strength on immersion in SBF from 1 hour to 24 hours but then a marked decrease from 24 hours to 168 hours. This contrasted markedly with the behaviour observed on immersion in Tris buffer. The reduction in the compressive strength has been associated with the conversion of OCP to HA for conventional calcium phosphate cements. Table 1: Presence of OCP as a function of time Discussion There are limited data with regard to the novel materials used in the present study although a recent study by Sadiasaet al.9 in which the investigators used injectable bone substitutes modified by placing bioactive glass powders (synthesized via a ultrasonic energy-assisted hydrothermal method) to the calcium phosphate-based bone cement in order to improve its biocompatibility. The present study did not use this particular methodology (e.g., using the ultrasonic energy assisted hydrothermal method) and therefore it would be interesting to speculate on the differences between the methods. For example in the Sadiasaet al. 8 study the injectable bone substitutes were initially composed of a powder component (tetracalcium phosphate, dicalcium phosphate dihydrate and calcium sulfate dehydrate) and a liquid component (citric acid, chitosan and hydroxylpropyl-methyl-cellulose) to which was added various concentrations of bioactive glass: 0%, 10%, 20% and 30%. By way of comparison in the present study the liquid and powder ratio was different and the bioactive glass content was with strontium 0% and Strontium 25%. Furthermore in the Sadiasaet al. 9 study the setting time and compressive strength of the injectable bone substitutes was evaluated and it was reported that the bone substitute improved (in terms of compressive strength) with the increased bioactive glass content. Another difference between the two studies was that the surface morphologies of the material was not evaluated by scanning electron microscope (SEM) before and after placing the samples into simulated body fluid in the present study. There was however agreement between the two studies with regard to the observation that there was an increase in the apatite formation as shown by xray diffraction. The in vitro biocompatibility of the injectable bone substitutes would therefore appear to improve with the placement of bioactive glass as the proliferation/adhesion behaviour of cells on the material increased as reported by Sadiasaet al9. Another element to the Sadiasaet al9study was that human gene markers were expressed by real timepolymerase chain reaction and the samples were reported to promote cell viability which appeared to demonstrate an improved biocompatible as the concentration of bioactive glass was increased. This aspect was not explored in the present study. In addition the In vivo biocompatibility of the various samples containing 0% and 30% bioactive glass was also evaluated using a Micro-CT and histological staining after 3 months of implantation in male rabbits' femurs. An interesting observation from the that there was no inflammatory reaction and significant bone .Another study of interest by Yu et al10 also evaluated a novel injectable bioactive cement in order to determine its composition, microstructure, setting time, injectability, compressive strength and to observe the behaviour of the material in simulated body fluid an aspect which was similar to the present study. The in vitro cellular International Dental Journal of Student’s Research, April-June 2015;3(2):43-45 {47} responses of the osteoblasts and the in vivo tissue responses following the implantation of calcium phosphate cement and bioglass in the femoral condyle defects of rabbits was also investigated by Yu et al.10 As mentioned previously the present study did not undertake any similar procedures in an animal model to determine whether the product would be suitable in terms of biocompatible which in retrospect would have been an important component to the investigation. In the Yu et al. study CPC-BG was observed to have a retarded setting time and also an improved injectability and mechanical properties than CPC alone. It was also observed that a new Cadeficient apatite layer was deposited on the composite surface after it was placed in SBF for 7 days. It was also observed that the CPC-BG samples demonstrated a significantly improved degradability and bioactivity compared to CPC in the simulated body fluid (SBF). The improvement in cell attachment, proliferation and differentiation on CPCBG were superior to cells observed on CPC. Macroscopic evaluation, histological evaluation, and micro-computed tomography (micro-CT) analysis observations also demonstrated that CPC-BG enhanced the efficiency of new bone formation in comparison with CPC alone. No histological evaluation or proliferation studies were undertaken in the present study. The Yu et al.10study concluded that a novel.HydroSet represented the next generation in bone substitute technology and was reported to be an excellent bone substitute solution for a number of clinical applications and surgical specialties. HydroSet was a self-setting calcium phosphate cement and contained apatite which converted to hydroxyapatite (the principal mineral component of bone). The crystalline structure and porosity of hydroSet indicated that it was an effective osteoconductive and osteointegrative material, with good biocompatibility An Ovine Implant study in Britain was undertaken by Hill et al.11 on bioactive glass (with three kinds of glasses) plus Calcium phosphate and Hydroset. The research group implanted the material into femur sites both right and left sides distal and proximal. The implantation was placed in one animal for six weeks and in six animals for twelve weeks. Scattered SEMs demonstrated that for the 6 weeks ovine implanted there was relatively little resorbtion of the cement for all cements including Hydroset. No thermal emissions (isothermic) were observed during the hardening phase at 6 weeks and three months. Analysis was done using XMT, Histology, Peripheral quantitative computed tomography (pQCTBack). It was observed that there was excellent osseointegration with bioglass cements and that HydroSet was more radio opaque due to higher density at 6 weeks. New bone growth surrounded all thecements and interdigitation of cements with the host bone The novel cements were observed to set invivo and ‘wash out’ of the cement was not witnessed and excellent osseointegration of all cement compositions was evident. New bone formation surrounding implanted cement high level of resorption and remodeling at twelve weeks octacalcium phosphate & hydroxyapatite forming cements brushite Cements was observed. Conclusion The results from the present study demonstrated that the media influenced how compressive strength changes and storage in SBF resulted in an increase in the compressive strength initially compared to a reduction in Tris buffer. The presence of strontium inhibited the formation of brushite probably because the Sr2+ cation cannot replace Ca2+ ions in the Brushite crystal lattice. It would therefore appear according to the results obtained that storing the combined CPC/Bioglass composition in Tris buffer solution and Simulated Body Fluid had an influence on both the compressive strength and the phase formed over the media used to store the cements influenced the phases formed and in particular the conversion. References 1. 2. 3. 4. 5. 6. 7. 8. 9. 10. Klokkevold, P. 2002. Surface enhancement to optimize the success of dental implants. Interview by Arun K. Garg. Dent Implantol Update, 13, 17-23 Laurencin, C., Khan, Y. & EL-AMIN, S. F. 2006. Bone graft substitutes. Expert Rev Med Devices, 3, 4957. Prasanna JS, Karunakar P, Rajashree D, Solomon RV. 2013 .Bone regeneration in a periodontally challenged hopeless tooth. J NTR Univ Health Sci;2:296-301 Legeros, R. Z. 1988. Calcium phosphate materials in restorative dentistry: a review. Adv Dent Res, 2, 16480. Hench, L. L. 1998. Bioceramics. Journal of the American Ceramic Society, 81, 1705-1728. T. V. Thamaraiselvi and S. Rajeswari.2004.Biological Evaluation of Bioceramic Materials - A Review.Trends Biomater.Artif. Organs, Vol 18 (1), pp 9-17 . Dahl, S. G., Allain, P., Marie, P. J., Mauras, Y., Boivin, G., Ammann, P., Tsouderos, Y., Delmas, P. D. &Christiansen, C. 2001. Incorporation and distribution of strontium in bone. Bone, 28, 446-53. Wang, X., Chen, L., Xiang, H. & YE, J. 2007. Influence of anti-washout agents on the rheological properties and injectability of a calcium phosphate cement. Journal of Biomedical Materials Research Part B: Applied Biomaterials, 81B, 410-418 Sadiasa, A., Sarkar, S. K., Franco, R. A., Min, Y. K. & LEE, B. T. 2014. Bioactive glass incorporation in calcium phosphate cement-based injectable bone substitute for improved in vitro biocompatibility and in vivo bone regeneration. J BiomaterAppl, 28, 739-56. YU, L., LI, Y., Zhao, K., Tang, Y., Cheng, Z., Chen, J., Zang, Y., WU, J., Kong, L., Liu, S., Lei, W. & WU, International Dental Journal of Student’s Research, April-June 2015;3(2):43-45 {48} Z. 2013. A novel injectable calcium phosphate cementbioactive glass composite for bone regeneration. PLoS One, 8, e62570 11. Karpukhina D.N., Kent &Hill (2013 submitted) highly bioactive glass reacts to form in vitro setting calcium phosphate bone cement. __________________________________________________ International Dental Journal of Student’s Research, April-June 2015;3(2):43-45 {46} SHORT COMMUNICATION Natural and synthetic hydrogels for periodontal tissue regeneration Dr. Marco Laurenti1 Dr. Mohamed-Nur Abdallah2 1 MSc, PhD, Postdoctoral Fellow, BDS, MSc, PhD, Faculty of Dentistry, McGill University, Montreal, Quebec, Canada 2 Corresponding Author Dr. Marco Laurenti Email: [email protected] Access this Article Online www.idjsr.com Use the QR Code scanner to access this article online in our database Article Code: IDJSR SE 0161 Quick Response Code Over the past few decades, there has been a great interest in periodontal regeneration therapy to restore the tissues destroyed by periodontal diseases, which are probably one of the most common bacterial infections in humans and the leading cause of tooth loss in adults. Periodontal diseases involve a set of inflammatory processes that are progressively destroying the tooth-supporting tissues (gingiva, periodontal ligament (PDL), alveolar bone and root cementum). Untreated periodontitis may cause irreversible destruction to these tissues leading to increased tooth mobility and subsequent tooth loss[1]. Currently, there is still no ideal therapeutic method to cure periodontitis or to optimally regenerate periodontal tissues in a predictable manner. Conventional mechanical or anti-infective periodontal therapies eliminate the inflammatory processes, and hinders or halts the diseases resulting generally in tissue repair without any notable signs of regeneration. Therefore, various regenerative approaches have been proposed and evaluated to restore the lost tooth-supporting tissues. These approaches included a wide range of surgical procedures and the use of various bone grafts, occlusal barrier membranes, purified protein mixtures, growth factors. Some of these approaches have achieved some success in regenerating the damaged periodontal tissues in certain ideal clinical cases; however, the outcomes are still very variable and unpredictable. Even with bone grafts, which are considered the current gold-standard material for bone regeneration, have reported failure rates up to 30% in maxillofacial and craniofacial surgeries, in addition to their drawbacks such as limited availability and donor-site morbidity. It is evident that a considerable research activity is required to improve the current periodontal therapies and to develop novel treatments to reach the ultimate goal of periodontal therapy, which is the predictable reconstruction of the lost periodontal tissues. Periodontal tissue possess the capacity to regenerate itself and substantial efforts in the tissue engineering field have been done to understand this ability in order to overcome the current limitations of therapeutic and regenerative procedures [3]. Tissue engineering is a multidisciplinary field that aims to guide body regeneration by specifically controlling the biological environment or developing biological substitutes to restore tissue functions. The damage to any tissue or organ results in the destruction and loss of extracellular matrix (ECM) with the absence of functional cells. For this reason, it is of paramount importance to restore the structure, properties, and functions of the native natural tissue. The general approach to restore the initial tissue condition is to use a three-dimensional (3D) scaffold which has the function to temporary supports the cell growth and new tissue development. The 3D scaffold may be designed as purely structural support providing with biological moieties incorporated into the scaffold to guide cell and tissue growth. Regeneration of dental/craniofacial tissues may be successfully achieved from the inimitable blend of human cells seeded biomaterial scaffolds with/without growth factors [4]. The combination of stem-cells, biomaterials, and physio-biochemical factors is the basis and major contribution of tissue engineering to regenerative medicine. In this approach, the biomaterial is critical to the regeneration of tissue since it serves as a three dimensional artificial ECM or scaffold to provide structural organization and support for the proliferation and differentiation of cells to create a neo-tissue. It is the interaction of the cells’ with the artificial ECM that is pivotal in recreating and maintaining the functional and 3D International Dental Journal of Student’s Research, April - June 2015;3(2):46-48 {47} structural integrity of the tissue [4a, 4c]. These scaffolds should be biocompatible facilitating cell attachment and proliferation, and biodegradable so that they do not require any surgical procedure for removal[5]. Hydrogels are highly hydrated polymeric biomaterials composed of hydrophilic polymeric network, either of synthetic or natural origin, and used as 3D scaffolds for periodontal tissue engineering applications[6]. Hydrogels are biodegradable, can be tailored to confer mechanical and structural properties similar to many ECM tissues, processed under mild conditions required to encapsulate biological moieties, and delivered in a minimally invasive manner[7]. Natural biomaterials have been extensively used in the development of matrix-based regenerative therapies that aim to accelerate clinical application due to their excellent biocompatibility, biodegradability, affinity for biomolecules and wound healing activity [8]. Materials such as collagen, hyaluronic acid, alginate and chitosan scaffolds have been used in periodontal regenerative research for more than two decades. The natural origin of these materials allows the design and engineering of biomaterial systems that function at the molecular level, often minimizing chronic inflammation. They can also be easily chemically and physically modified to form desired structures. The use of natural polymers in the form of hydrogels allows for the incorporation of biological agents by promoting cross-linking when the growth factor is dispersed in the polymer solution. Because natural polymers are often soluble in water, the creation of hydrogels may occur under mild fabrication conditions that are relatively harmless to the bioactivity of the growth factors. Normally, these hydrogels are degraded by enzymes and/or acid hydrolysis at a rate depending on the degree of crosslinking or the molecular weight [8]. Collagen is one of the most used biomaterials due to its excellent biocompatibility, weak antigenicity, biodegradability, and safety. Collagen hydrogel fits well with injectable cell delivery and highly porous cross-linked scaffolds provide good mechanical stability. For these reasons, collagen hydrogels have been used as support for in vitro growth of many types of tissues and to deliver different kinds of growth factors. Following the clinical use of collagen carriers delivering bone morphogenetic proteins for tibial shaft fractures [9], spine fusions and long-bone nonunions [10], collagen is currently being evaluated for widespread clinical periodontal regeneration. For example, there are commercially available collagen composite scaffolds such as Formagraft™ and OssiMend™ for periodontal regeneration currently used in animal studiesand clinical trials [11]. Chitosan is biodegradable natural polymer and it has been used as a cell vehicle material due to its ability to be molded into various geometries (e.g. porous structures). In addition, chitosan has minimal foreign body reaction and high affinity for in vivo macromolecules [12]. However, chitosan is not strongly supportive of tissue regeneration as demonstrated by its effect on the width of keratinized gingiva in dogs [13]. The addition of hydroxyapatite to chitosan hydrogels produced a 3D scaffold in which the pore sizes and interconnectivity were preserved, resulting in a suitable 3D environment to support cellular structure, proliferation and mineralization [14]. Synthetic polymers have been widely used for scaffolding applications because of their ability to provide controllable and reproducible structural properties, biocompatibility, and tailored biodegradation rates [15]. Scaffolds made of synthetic polymers can be produced by a variety of fabrication techniques, and they can be manufactured into preformed sizes and shapes according to clinical requirements. There are several synthetic polymers approved by the US Food and Drug Administration such as poly(glycolic acid) (PGA), poly(L-lactic acid) (PLA), their copolymers poly(lactic-co-glycolic acid) (PLGA), and poly(caprolactone) (PCL). Solid scaffolds are typically porous matrices fabricated by techniques such as solvent casting, gas foaming, particulate leaching, and electrospinning [5, 15]. Other degradable polymers have also been explored and tested for periodontal tissue regeneration including poly(ethylene glycol) (PEG) [16], polylactide and polyglycolide [17]. Amorphous poly(D,L-Lactic acid) (PDLLA) used in combination with bioactive glasses ensure the creation of a macroporous structure within the bioceramic materials showing promising properties for periodontal tissue regeneration [5]. Synthetic polymers can be also used in combination with natural biomaterials. For example, the rapid degradation of fibrin, a biopolymer critical to hemostasis and wound healing, can be decelerated by modification with (PEG) [18]. Recently, nanocomposites based on polymers and nanosilica nanoparticles have been applied in periodontal tissue engineering. Silica nanoparticles are obtained by the sol–gel method and they can be incorporated into a polymeric matrix resulting in a nanocomposites with unique properties such as high mechanical resistance, chemical stability, and heat resistance [19]. Tubular nanocomposite scaffolds of poly(ethyl methacrylate-co-hydroxyethyl acrylate) [P(EMA-co-HEA)] were synthesized with different concentrations of silica nanoparticles by fibertemplating method in order to mimic the structure and functions of natural dentin [20]. These tubular structures were found to induce the precipitation of {48} hydroxyapatite on their surface, and they can facilitate odontoblastic cell growth with the integration of host mineralized tissue. Hybrid P(EMA-co-HEA)/SiO2nanocomposite matrix incubated in simulated body fluid for 14 days showed the best cellular distribution and neo-dentin like pattern. These constructs also showed enhanced mechanical properties to withstand functional stresses [20]. All these results indicate that the nanohybrid matrix scaffolds could be promising potential sources for dentin repair and regeneration. Summary and Challenges Periodontal regeneration remains a highly challenging task, since the existing therapeutic and regenerative approaches have not achieved a complete or predictable regeneration of the lost periodontal tissues in humans. The purpose of research into periodontal regeneration is to establish a new approach that overcomes the limitations of the current therapeutic and regeneration procedures. The synergistic approach of nanomedicine and tissue engineering is a promising field and has lead, so far, to a remarkable progress in the field of periodontal tissue regeneration. Several approaches utilizing collagen, chitosan, or nano-composites as hydrogels or rigid scaffolds have been found to promote and guide periodontal tissue regeneration. However, these studies are still limited to in vitro and in vivo studies which highlights the need of future investigation in this promising field. Acknowledgment The authors would like to acknowledge the Network of Oral and Bone Health Research, and the Faculty of Dentistry of McGill University for their financial support. References 1. 2. 3. 4. 5. 6. Yuan, Y. Zhu, Y. Y. Zhang, X. L. Li, Y. Zhang, K. D. Yao, Eur. Polym. J. 2006, 42, 2013-2022. 7. J. L. Drury, D. J. Mooney, Biomaterials 2003, 24, 4337-4351. 8. J. F. Mano, G. A. Silva, H. S. Azevedo, P. B. Malafaya, R. A. Sousa, S. S. Silva, L. F. Boesel, J. M. Oliveira, T. C. Santos, A. P. Marques, N. M. Neves, R. L. Reis, J. R. Soc. Interface 2007, 4, 999-1030. 9. R. Cancedda, P. Giannoni, M. Mastrogiacomo, Biomaterials 2007, 28, 4240-4250. 10. A. P. White, A. R. Vaccaro, J. A. Hall, P. G. Whang, B. C. Friel, M. D. McKee, Int. Orthop. 2007, 31, 735741. 11. aF. Feng, K. Akiyama, Y. Liu, T. Yamaza, T. M. Wang, J. H. Chen, B. B. Wang, G. T. Huang, S. Wang, S. Shi, Oral. Dis. 2010, 16, 20-28; bT. G. Kim, U. M. Wikesjo, K. S. Cho, J. K. Chai, S. D. Pippig, M. Siedler, C. K. Kim, J. Clin. Periodontol. 2009, 36, 589597. 12. I. Y. Kim, S. J. Seo, H. S. Moon, M. K. Yoo, I. Y. Park, B. C. Kim, C. S. Cho, Biotechnol. Adv. 2008, 26, 1-21. 13. G. Lotfi, M. A. Shokrgozar, R. Mofid, F. M. Abbas, F. Ghanavati, A. A. Bagheban, R. P. Shariati, J. Periodontol. 2011, 82, 1367-1375. 14. aA. C. Akman, R. S. Tigli, M. Gumusderelioglu, R. M. Nohutcu, J. Biomed. Mater. Res. A 2010, 92, 953-962; bY. Zhang, B. Shi, C. Li, Y. Wang, Y. Chen, W. Zhang, T. Luo, X. Cheng, J. Control Release 2009, 136, 172-178; cF. Liao, Y. Chen, Z. Li, Y. Wang, B. Shi, Z. Gong, X. Cheng, J. Mater. Sci. Mater. Med. 2010, 21, 489-496. 15. P. Gunatillake, R. Mayadunne, R. Adhikari, Biotechnol. Annu. Rev. 2006, 12, 301-347. 16. P. Valderrama, R. E. Jung, D. S. Thoma, A. A. Jones, D. L. Cochran, J. Periodontol. 2010, 81, 737-747. 17. G. Serino, W. Rao, G. Iezzi, A. Piattelli, Clin. Oral. Implan. Res. 2008, 19, 26-31. 18. J. M. Galler, A. C. Cavender, U. Koeklue, L. J. Suggs, G. Schmalz, R. N. D'Souza, Regen. Med. 2011, 6, 191200. 19. aD. S. Achilias, D. N. Bikiaris, V. Karavelidis, G. P. Karayannidis, Eur. Polym. J. 2008, 44, 3096-3107; bM.Sadej-Bajerlain, H. Gojzewski, E. Andrzejewska, Polymer 2011, 52, 1495-1503; cS.Barus, M. Zanetti, M. Lazzari, L. Costa, Polymer 2009, 50, 2595-2600. 20. A. V. Lluch, A. C. Fernandez, G. G. Ferrer, M. M. Pradas, J. Biomed. Mater. Res. B 2009, 90b, 182-194. __________________________________________________ B. L. Pihlstrom, B. S. Michalowicz, N. W. Johnson, Lancet 2005, 366, 1809-1820. H. F. Rios, Z. Lin, B. Oh, C. H. Park, W. V. Giannobile, J. Periodontol. 2011, 82, 1223-1237. F. M. Chen, Y. Jin, Tissue Eng. Part. B-Re 2010, 16, 219-255. aV. Rosa, A. Della Bona, B. N. Cavalcanti, J. E. Nor, Dent. Mater. 2012, 28, 341-348; bS. F. Yang, K. F. Leong, Z. H. Du, C. K. Chua, Tissue Eng. 2001, 7, 679-689; cS. F. Yang, K. F. Leong, Z. H. Du, C. K. Chua, Tissue Eng. 2002, 8, 1-11. K. Rezwan, Q. Z. Chen, J. J. Blaker, A. R. Boccaccini, Biomaterials 2006, 27, 3413-3431. aC. R. Nuttelman, S. M. Henry, K. S. Anseth, Biomaterials 2002, 23, 3617-3626; bB. Duan, X. Y. International Dental Journal of Student’s Research, April - June 2015;3(2):46-48 {49} CLINICAL CASE REPORT Bone fenestration: A case report of management of a lower anterior buccal bone fenestration Dr. Murai Khalifa1 Yuliya Mulyar2 1 BDS, DDS, GPR, McGill University, Family Dental Clinic, Strathmore, Alberta, Canada 2 BA Psychology, DDS candidate Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada. Corresponding Author: Dr. Murai Khalifa Email:[email protected] Access this Article Online www.idjsr.com Use the QR Code scanner to access this article online in our database Article Code: IDJSR 0162 Quick Response Code Abstract The term fenestration refers to a circumscribed defect that creates a "window" through the bone over the prominent root. This lesion will be seen when the alveolar bone is exposed by a flap surgery, its associated with localized periodontal destruction.This case reports a 62year-oldmale patient who presented with persistent tooth pain at the lower right 2ed incisor, despite repeated root canal treatments by a general practitioner. When the patient visited our clinic, a CT examination was performed and apical fenestration was diagnosed. The tooth was not restorable due to a fracture in the apical third from a previous root canal therapy. Suggested treatment plan was to extract the tooth, perform bone grafting and a future implant. revealed by a flap operation or on a skull preparation the coronal portion of the root is often not covered by bone"dehiscence" or there is a fenestration. Isolated areas in which the root is denuded of bone and the root surface is covered only by periosteum and overlying gingiva are termed fenestrations. In these areas, the marginal bone is intact [1,2]. Such defects occur on approximately 20% of the teeth; they occur more often on the facial bone than on the lingual bone, they are more common on anterior teeth than on posterior teeth, and they are frequently bilateral. Microscopic evidence of lacunar resorption may be present at the margins. The causes of these defects are not clear. However, predisposing factors include prominent root contours, malposition and labial protrusion of the root in combination with a thin bony plate[3]. Incidence The incidence of apical fenestration is between 7.5% and 20%, and is higher in the maxillary than in the mandibular teeth. It has also been reported that the incidences higher in the anterior than in the posterior teeth. The most commonly observed regions are the canine root and the mesio-buccal root of the maxillary first molar. Although the cause of apical fenestration is still unclear, it has been suggested that anatomical factors such as age-related changes and the positional relationship between the tooth and the alveolar bone might be involved. Moreover, occlusal dysfunction should also be considered.[4] Management Fenestration and dehiscence defects have been managed with barrier membranes or simply with flap closure. Also, bone grafts has been used in this case. The only controlled comparison studies between membrane treatment and periosteal flap coverage of exposed implant surfaces in humans demonstrated Keywords:Bone fenestration; Tooth;Graft. that the membrane treatment was far superior with regard to bone fill.11Another controlled study in Introduction humans has shown better results in the membrane groups; Infour outof six sites (67%) treated with a In a healthy periodontium the facial margin of the membrane resulted in 95% to 100% elimination of alveolar crest lies approximately 2 mm apical to the the dehiscence and total coverage of the threads. In gingival margin, which courses near to the cement the control sites, only 2 of 6 sites (33%) showed enamel junction. The facial aspect of the alveolar moderate-to-complete bone fill.43All other clinical bone covering the root is usually very thin. As studies which are in the form of case reports, International Dental Journal of Student’s Research, April - June 2015;3(2):49-51 {50} demonstrates coverage of an implant dehiscence using a barrier membrane. Admittedly, without a biopsy, it cannot be determined whether the tissue covering the implant is bone or firm connective tissue.[5,6] A one-year multicenter study evaluating 55 Brånemark implants (i.e., machined-surface, external hex) with bone dehiscence in 45 patients, treated by ePTFE membrane alone, demonstrated an average bone fill of 82%.12. The average initial defect height was 4.7 mm. The 1-year follow-up of these implants demonstrated a favorable response to loading. Of the 55 implants, a total of 6 failed, corresponding to a cumulative survival rate of 84.7% in the maxilla and 95.0% in the mandible, which is similar to previously published results for this implant design. A clinical report on the use of TR membranes demonstrated the biologic potential to fill a large protected space in four patients. 24Bone dehiscence at implant sites ranged from 5 to 12 mm (mean:8.2 mm). They were covered with a TR membrane alone (no graft). Re-entry after 7 to 8 months of submerged healing found complete bone coverage over all the implants. Radiographic evaluation demonstrated that the implants were functioning with normal crestal bone support after 1 year. No clinical comparisons are available in the literature evaluating the placement of bone grafts with or without barrier membranes on implant dehiscence defects. Most evidence supports the use of graft materials in conjunction with membrane treatment, particularly the use of FDBA in conjunction with GBR. In a study with 40 patients, 110 implants were placed in conjunction with barrier membranes and FDBA; a success rate of 96.8% was achieved with complete bone fill (defined as >90% fill of dehiscence)[7].This study reported a membrane exposure rate of 29%, but noted little adverse effect on the bone regeneration. Figure 1. Preoperative diagnostic CT scan Figure 2. Post extraction Periapical radiograph It was decided and explained to the patient that the best approach was to extract tooth 4.2 followed by a bone graft and an implant 6 months later. The patient consented to the proposed treatment and subsequently full mouth impressions were taken to develop study casts. Upon extraction, the bone fenestration was noticed with loss of the buccal alveolar bone wall. Fig3 Case report A 62 years male patient presented to our clinic with persistent pain related to the lower 2ed right incisor tooth. Patient wanted the tooth extracted and implant placed. Patient medical history was normal. Upon oral examination, tooth 4.2 was positive to percussion with possible horizontal fracture. Mobility class II was noticed and upon radiographic examination, well circumscribed radiolucent lesion surrounding the apex of tooth 4.2 was notices, which turned to be a periapical granuloma after a biopsy.Fig 1,2 Figure 3. Intraoral view showing the alveolar bone loss and fenestration Curettage of the lesion was completed and was sent for biopsy. Next step was to graft the socket using 2 International Dental Journal of Student’s Research, April - June 2015;3(2):49-51 {51} types of bone. We used a mix of allograft material which is composed of 50% mineralized cortical and 50% mineralized cancellous size 0.25 mm with the 4bone bch composed of 60 % hydroxyapatite and 40% beta-Tricalcium Phosphate that was covered with a resorbable membrane. Fig4 Figure 4:Facial view showing bone graft covered by the membrane References 1. Color atlas of Dental Medicine Periodontology By Herbert F. Wolf, Edith M. Rateitschak-Pluss 2. Caranza's Clinical periodontology , By Michael G. Newman, Henry Takei, Perry R. Klokkevold, Fermin A. Carranza 3. Elliot JR, Bowers GM: Alveolar dehiscence and fenestration. Periodontics1:245, 1963. 4. Bulletin of Tokyo Dental College, 53(1): 23-26 5. Jensen OT, Greer RO, Jr, Johnson L, et al: Vertical guided bone-graft augmentation in a new canine mandibular model. Int J Oral MaxillofacImplants 10:335–344, 1995. 6. Klokkevold PR, Han TJ, Camargo PM: Aesthetic management of extractions for implant site development: delayed versus staged implant placement. Pract Periodontics Aesthet Dent 11:603– 610, quiz 612,1999. 7. Klokkevold PR, Han TJ, Camargo PM: Aesthetic management ofextractions for implant site development: delayed versus staged implant placement. Pract Periodontics Aesthet Dent 11:603– 610, quiz 612,1999. __________________________________________________ The flap then was closed using silk sutures size 3.0. An antibiotic course was started and analgesics were prescribed to patient and he was scheduled for follow-ups.Fig5 Figure5: Periapical radiograph6 weeks follow up. The patient reported that pain and discomfort disappeared. Site healing was optimal and patient will be ready for an implant in a four months period. Conclusion The etiology of bone fenestration is still unclear. In most cases, early diagnosis and treatment can lead to successful management and do not require extensive treatment. In this case, we were able to preserve the socket for a future implant and give patient more options to restore the missing tooth. International Dental Journal of Student’s Research, April - June 2015;3(2):49-51 {52} CLINICAL DIAGNOSIS & GUIDELINES A Histological and Clinical Evaluation of Shallow and Deep Probing Depths Siavash Hassanpour1, Shlomi Tamam2, Timur Shigapov3, 1 H.BSc., M.Sc. DDS, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Canada 2 H.BSc., M.Sc. DDS, East Village Dental Center, Oshawa, Canada. 3 H.BSc, DDS, De Man and Hoediono Dentistry, Kitchener, Canada Corresponding Author Siavash Hassanpour Email: [email protected] Access this Article Online www.idjsr.com Use the QR Code scanner to access this article online in our database Article Code: IDJSR SE 0163 Quick Response Code New and improved diagnostic aides are constantly being developed and deployed to improve our ability to correctly identify, diagnose and treat a variety of medical conditions. Despite several attempts at developing a new gold standard tool for the diagnosis of periodontal disease [1], dental professionals still largely, although not exclusively, rely on the periodontal probe and periodontal probing depth (PD) for the identification and classification of periodontal diseases. PD is commonly used to evaluate the presence or absence of periodontal disease, and PD values are relied on for the determination of periodontal disease severity[2,3]. To these authors’ knowledge, no clinical study has conclusively defined the healthy PD. Generally speaking, in the absence of gingival recession or enlargement, shallow PDs of 1-3mm are considered to be indicative of a healthy or normal periodontal sulcus. Sites with PD values of 4-6mm are considered moderately deep and indicative of early stages of periodontal disease. Ultimately, sites with PD values greater than 7mm are considered as deep sites, typically caused by advanced periodontal disease. As such, clinicians view sites with progressively larger PDs with increasing concern. The aim of this short commentary is to use evidence from histological and clinical studies to discuss the merits of considering a shallow probing depth as healthy and to highlight changes induced by periodontal disease that lead to deepening periodontal pockets. PD is defined as the distance from the gingival margin to the deepest part of the probable crevice. Historically, the terms PD and pocket depth were used interchangeably. In 1971, Listgarten noted that PD measurements only offer an estimation of the true pocket depth, as the probe tip routinely goes beyond the sulcus and into the attachment apparatus of the tooth [4]. He further suggested that the only way to measure the depth of the anatomical sulcus, or pocket, is through histological means. One of the first studies to examine the physiological attachment around healthy human teeth was carried out in 1961 by Gargiulo et al. In this study, Gargiulo defined the dentogingival complex as the physiological and functional supporting tissue of teeth[5]. Measurements were made along 325 surfaces of presumably healthy teeth in human cadaver jaws. They concluded that the dentogingival junction is composed of the junctional epithelium (formerly the epithelial attachment) and the connective tissue fibrous attachment. In the study, Gargulio measured the depth of the true gingival sulcus, the length of the junctional epithelium and the length of connective tissue attachment throughout various phases of passive eruption. The study found the average sulcus depth to be 0.69mm (range 0.61mm - 1.71mm). The average length of the junctional epithelium was 0.97mm (range 0.71mm – 1.35mm), while the average length of the connective tissue attachment was 1.07mm (range 1.03mm – 1.07mm) [5](Figure 1). As such, assuming a periodontal probe could precisely measure the depth of a healthy sulcus, we would expect to get measurements ranging from 0.61-1.71mm. However, the anatomic sulcus or pocket depth rarely corresponds to the clinical PD measurement. The periodontal probe routinely goes beyond the sulcus and penetrates the coronal part of the junctional epithelium [6]. Even in periodontal health and in absence of inflammation, the periodontal probe penetrates the junctional epithelium by 0.5mm, stopping 0.4mm coronal to the termination of the junctional epithelium [6]. This increases the expected PD in healthy tissues to 1.112.21mm (1.11mm = 0.61mm +0.5mm; 2.21mm = 1.71mm + 0.5mm). Also, seeing that probing depth International Dental Journal of Student’s Research, April - June 2015;3(2):52-55 {53} measurements are accurate to within 1mm 90% of the time [7], we can expect healthy probing depths to range between 0.11mm-3.21mm (0.11mm = 1.11mm - 1mm ; 3.21 mm = 2.21mm + 1mm) or simply up to 3mm(Figure 2A). To understand why PD values increase with periodontal disease progression, one has to examine the pathophysiology of gingivitis and periodontitis. Gingivitis is defined by the presence of gingival inflammation without the loss of periodontal attachment [8]. Histologically, gingivitis is characterized by an increase in blood flow, an influx of inflammatory cells and breakdown of perivascular connective tissue (Figure 2B)[9]. All together, these changes lead to clinical features consisting of edematous, erythematous and friable gingival tissues that typically extend coronal to the cement enamel junction (CEJ) and readily bleed upon probing. Even without periodontal attachment loss, clinical PD values are increased in gingivitis when compared to clinically healthy sites. The increased PD during gingivitis is explained partly by the gingival enlargement and partly by increased penetration of the periodontal probe in inflamed tissues. Studies have shown that the tip of a periodontal probe penetrates the full length of the junctional epithelium, stopping 0.1mm to the apical termination of the junctional epithelium in an inflamed site[6], compared to 0.4mm in healthy periodontium as discussed above. Together, these two factors explain why increased PD values are often associated with gingivitis in the absence of concurrent loss of periodontal attachment or loss of alveolar bone. Unlike gingivitis, periodontitis is characterized by the pathological loss of collagen fibers, apical migration of the junctional epithelium, loss of alveolar bone and periodontal attachment (Figure 2C). Histologic studies of the progression of periodontitis have shown that early periodontal lesions are initially localized to the gingival sulcus and later progress to the periodontium proper, the periodontal ligament, cementum and alveolar bone. Histological studies in more advanced periodontal disease have shown that during probing, the probe tip penetrates the full length of the junctional epithelium and extends deep into the connective tissue attachment[6,10], resulting in progressively increased probing depth proportional to the degree of attachment loss. For this reason, deep PDs are seen in tissues undergoing periodontal breakdown due to the concurrent apical migration of the junctional epithelium, the inflamed nature of the connective tissues and the loss of alveolar bone. Clinical studies routinely classify pockets into three groups: 1-3mm, 4-6mm and 7mm and greater [1113].The reason for this common classification system is twofold. Firstly, this classification system stratifies diseased sites and helps determine the aggressiveness of treatment including the need for surgical periodontal therapy [14]. Secondly, the shallow, moderate and deep stratification system allows clinicians to determine the expected outcome of treatment. Clinicians are often less concerned with shallow pockets, as shallow pockets are more amendable to routine oral hygiene at home and are easier to maintain under professional care. Shallow pockets are more likely to be adequately cleansed by manual tooth brushing, which penetrate 0.9mm below the gingival margin [15]. Likewise, selfadministered oral hygiene routines can remove interproximal plaque up to 2.5mm subgingivally [16]. Also, when receiving professional prophylaxis and scaling and root planning (SRP), Brayer et al. found that even inexperienced practitioners can adequately cleanse shallow pockets[17]. These shallow pockets are compatible with good health and function for many years [18]. While SRP is less efficient at removing the etiological agents that cause disease progression in deeper sites, it can effectively remove plaque and calculus up to 3.73mm subgingivally. Deeper pockets (> 5mm), however, are rarely free of plaque and calculus following SRP [19]. Longitudinal studies comparing surgical and nonsurgical therapy have validated the need for surgical therapy when faced with deep(>7mm) PDs [13,20]. For pockets greater than or equal to 7mm, surgical periodontal intervention resulted in significantly greater reduction in probing depth than SRP alone. This can be attributed to increased access, direct visualization and correction of unfavorable osseous architecture during periodontal surgery. In 1982, Lindhe described the concept as the critical probing depth (CPD) [21]. CPD is defined as the threshold PD where gain of clinical attachment can be expected following treatment. Lindhe found that the CPD for SRP was 2.9mm, while the CPD of surgical therapy (Modified Widmann Flap) was 4.2mm. Together, these results indicate that shallow pockets < 3mm can be adequately maintained without further loss of attachment by scaling alone, while promoting the use of surgical therapy in sites with deeper initial PDs. Periodontal probing is not a flawless diagnostic instrument and should not be used in isolation. A high PD value alone does not imply active disease nor disease progression[22]. Variations in PD can be introduced by probing force, probe angulation, probe thickness, probe type as well as tooth site and local anatomy [3]. Further, intra- and inter-clinician variability and reproducibility in PD have been reported [7,23]. In addition, the depth of probe penetration is directly proportional to the degree of inflammation [24-26]. In fact, rather than simply measuring PD, a better indicator of periodontal disease severity is clinical attachment level (CAL). CAL, which is calculated by summing PD and recession, more accurately estimates loss of periodontal attachment as a result of periodontal International Dental Journal of Student’s Research, April - June 2015;3(2):52-55 {54} disease [27]. Nonetheless, periodontal probing provides clinicians with a useful estimate of the location of the most coronal insertion of the intact connective tissue fibers and thereby verifying the presence or absence of periodontal disease. Gingival enlargement in combination with increased periodontal probe penetration result in increased PD in tissues suffering from gingivitis. Typically, in tissues affected by gingivitis, the tip of a periodontal probe penetrates the full length of the junctional epithelium and enters the connective tissue attachment, resulting in probing depth of ≥4mm. C) Periodontitis is characterized by the pathological loss of collagen fibers, apical migration of the junctional epithelium, loss of alveolar bone and periodontal attachment. In advanced periodontal disease, the probe tip penetrates the full length of the junctional epithelium and extends deep into the connective tissue attachment; the more severe the periodontal attachment loss, the greater the corresponding probing depths. Inspiration for image comes from the Color Atlas of Dental Medicine Periodontology, 3rd Edition, and was kindly illustrated by Ms. Gretchen Kramer. References 1. Figure 1. The Dentongingival Complex The attachment apparatus of a tooth is comprised of the dentogingival complex and the periodontal ligament. The dentogingival complex is composed of the junctional epithelium and the connective tissue fibrous attachment. The average sulcus depth is 0.69mm (range 0.61mm 1.71mm). The average length of the junctional epithelium is 0.97mm (range 0.71mm – 1.35mm) and the average length of the connective tissue attachment is 1.07mm (range 1.03mm – 1.07mm). Inspiration for image comes from the Color Atlas of Dental Medicine Periodontology, 3rd Edition, and was kindly illustrated by Ms. Gretchen Kramer. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. Figure 2. The Progression of Periodontal Disease A) In periodontal health, the periodontal probe penetrates the entire length of the anatomical sulcus and half way through the junctional epithelium, resulting in probing depths of 1-3mm. B) Histologically, gingivitis is characterized by an increase in blood flow, an influx of inflammatory cells and breakdown of perivascular connective tissue resulting in edematous, erythematous and friable gingival tissues. 13. 14. 15. Wolf DL, Lamster IB. Contemporary concepts in the diagnosis of periodontal disease. Dent Clin North Am 2011;55:47–61. Listgarten MA. A perspective on periodontal diagnosis. J Clin Periodontol 1986;13:175–81. Listgarten MA. Periodontal probing: What does it mean? J Clin Periodontol 1980;7:165–76. Listgarten MA. Normal development, structure, physiology and repair of gingival epithelium. Oral Sci Rev 1972;1:3–67. Gargiulo AW, Wentz FM. Dimensions and relations of the dentogingival junction in humans. Journal of Periodotology 1961;32:261–7. Armitage GC, Svanberg GK, Loe H. Microscopic evaluation of clinical measurements of connective tissue attachment levels. J Clin Periodontol 1977;4:173–90. Advances in measurements of periodontal bone and attachment loss. 2000;17:56–72. Theilade E, Wright WH, Jensen SB, Loe H. Experimental gingivitis in man. J Periodontal Res 1966;1:1–13. Page RC, Schroeder HE. Pathogenesis of inflammatory periodontal disease. A summary of current work. Lab Invest 1976;34:235–49. JF S, FG B. Probing of pockets related to the attachment level. J Periodontol 1976;47:281–6. Hill RW, Ramfjord SP, Morrison EC, Appleberry EA, Caffesse RG, Kerry GJ, et al. Four types of periodontal treatment compared over two years. J Periodontol 1981;52:655–62. Ramfjord SP, Morrison EC, Burgett FG, Nissle RR, Shick RA, Zann GJ, et al. Oral hygiene and maintenance of periodontal support. J Periodontol 1982;53:26–30. Kaldahl WB, Kalkwarf KL, Patil KD, Molvar MP, Dyer JK. Long-term evaluation of periodontal therapy: I. Response to 4 therapeutic modalities. J Periodontol 1996;67:93–102. Greenwell H, Stovsky DA, Bissada NF. Periodontics in general practice: perspectives on nonsurgical therapy. J Am Dent Assoc 1987;115:591–5. Waerhaug J. Effect of Toothbrushing on Subgingival Plaque Formation. J Periodontol 1981;52:30–4. International Dental Journal of Student’s Research, April - June 2015;3(2):52-55 {55} 16. 17. 18. 19. 20. 21. 22. 23. 24. 25. 26. 27. Waerhaug J. The interdental brush and its place in operative and crown and bridge dentistry. Journal of Oral Rehabilitation 1976;3:107–13. Brayer WK, Mellonig JT, Dunlap RM, Marinak KW, Carson RE. Scaling and root planing effectiveness: the effect of root surface access and operator experience. J Periodontol 1989;60:67–72. Merchant AT, Oranbandid S, Jethwani M. Position paper: epidemiology of periodontal diseases. J Periodontol 2005;76:1406–19. Stambaugh RV, Dragoo M, Smith DM, Carasali L. The limits of subgingival scaling. The International Journal of Periodontics & Restorative Dentistry 1981;1:30–41. Becker W, Berg L, Becker BE. The long term evaluation of periodontal treatment and maintenance in 95 patients. The International Journal of Periodontics & Restorative Dentistry 1984;4:54–71. Lindhe J, Socransky SS, Nyman S, Haffajee A, Westfelt E. “Critical probing depths” in periodontal therapy. J Clin Periodontol 1982;9:323–36. Claffey N, Nylund K, Kiger R, Garrett S, Egelberg J. Diagnostic predictability of scores of plaque, bleeding, suppuration and probing depth for probing attachment loss. 3 1/2 years of observation following initial periodontal therapy. J Clin Periodontol 1990;17:108– 14. Isidor F, Karring T, Attström R. Reproducibility of pocket depth and attachment level measurements when using a flexible splint. J Clin Periodontol 1984;11:662– 8. Schroeder HE, Listgarten MA. Fine structure of the developing epithelial attachment of human teeth. Monogr Dev Biol 1971;2:1–134. Spray JR, Garnick JJ, Doles LR, Klawitter JJ. Microscopic demonstration of the position of periodontal probes. J Periodontol 1978;49:148–52. Caton J, Greenstein G. Depth of Periodontal Probe Penetration Related to Clinical and Histologic Signs of Gingival Inflammation*. Journal of Periodotology 1981. Armitage GC. Position Paper: Diagnosis of Periodontal Diseases. J Periodontol 2003;74:1237–47. ___________________________________________ International Dental Journal of Student’s Research, April - June 2015;3(2):52-55 {56} CLINICAL DISCUSSION Prevention of mandibular nerve injury associated with dental implant placement: preoperative and intraoperative recommendations Thomas T. Nguyen DMD, MSc, Resident in Periodontology, University of Minnesota, School of Dentistry, Minnesota, USA. Corresponding author: Dr. Thomas T. Nguyen Email: [email protected] Access this Article Online www.idjsr.com Use the QR Code scanner to access this article online in our database Article Code: IDJSR SE 0164 Quick Response Code Clinical Question How can we minimize mandibular nerve injury during dental implant placement? One of the common complications after implant placement is nerve injury. This complication usually results in the altered sensation of the lower lip, chin, mucosa, alveolar gingiva, teeth and/or tongue. The mandibular nerve injury is the most frequent due to the reabsorption of the alveolar ridge after its atrophy due to lack of stimuli after extraction. Pre-operative recommendations Informed consent for implant placement must be given to the patient. The surgeon should forewarn the patient regarding the possibility of postoperative impaired sensation. A thorough clinical examination and treatment planning must be performed. Radiographic examination to assess the location of the inferior alveolar canal (IAC) of the mandibular nerve and the mental foramen: The panoramic radiograph, preferably digital, is useful as the primary image study to assess the vertical distance from the crest of the edentulous mandibular alveolar ridge to the superior aspect of the IAC. If the panoramic film shows inadequate distance from the edentulous alveolar crest to the IAC to place an implant, a computed tomography (CT) scan is required. CT scans have improved resolution and allow: Visualization of the IAC in 3 dimensions.1 Coronal sections which are the most useful to assess structural bucco-lingual relationships and assessment of the IAC’s shape (round, teardrop, dumbbell) and osseous cortication.2 Intra-operative recommendations Intra-operative radiographs during implant preparation and after implant placement are required in order to assess the position and angulation of the implant. Surgical equipment with predetermined depth stops as well as careful surgical technique is necessary. During implant placement, cancellous bone may be compressed against the contents of the IAC, possibly causing mandibular nerve damage. If the post-operative radiograph indicates encroachment on the IAC: The implant should be removed; Dexamethasone should be introduced into the osteotomy site as follows3: Dexamethasone liquid 4 mg./ml. applied topically for 1 minute and repeated once. Followed by a 7day day regimen of dexamethasone orally as follows: Days one and two: 8mg. per day Days three and four: 6 mg. per day Days five and six: 4 mg. per day Day seven: 2 mg. An appropriate antibiotic must be prescribed. Oral dexamethasone must be voided in patients with preexisting cardiac, hypertensive and/or renal issues since such high doses may lead to a hypertensive crisis. A shorter implant should be considered and no bone grafting materials should be placed in order to avoid migration into IAC.4 International Dental Journal of Student’s Research, April - June 2015;3(2):56-57 {57} Conclusion One of the serious complications of posterior mandibular implant placement is nerve injury. Proper understanding of the involved anatomy and the surgical procedures, along with proper treatment planning, will reduce the chances of such morbidity to the patient. If nerve injury occurs, early and proper management is the key to maximizing the chances of recovery. References 1. Khan I, Halli R, Gadre P, et al. Correlation of panoramic radiographs and spiral CT scan in the preoperative assessment of intimacy of the inferior alveolar canal to impacted mandibular third molars. J Craniofacial Surg. 2011;22(2):566-570. 2. Ueda M, Nakamori K, Shiratori K, et al. Clinical significance of computed tomographic assessment and anatomic features of the inferior alveolar canal as risk factors for injury of the inferior alveolar nerve at third molar surgery. J Oral Maxillofac Surg. 2012;70:514520. 3. Misch CE, Resnik Randolph. Mandibular nerve neurosensory impairment after dental implant surgery : management and protocol. Implant Dentistry. 2010;19(5). 4. Bagheri AC, Meyer RA. Management of mandibular nerve injuries from dental implants. Atlas Oral Maxillofac Surg Clin N Am. 2011;19:47-61. __________________________________________________ International Dental Journal of Student’s Research, April - June 2015;3(2):56-57 {58} CLINICAL CASE REPORT Periodontal Disease and Diabetes Mellitus: Case Report H Marie Keeling1 Tamara L. Wright2 and dental conditions along with her social habits was taken. 1 Medical History Dental Student at Dalhousie University, BA, MA, Faculty of Dentistry, Dalhousie University, Halifax, NS, Canada 2 BSc, MSc, DMD, Dip Perio, Department of Dental Clinical Sciences, Faculty of Dentistry, Dalhousie University, Halifax, NS, Canada Corresponding author: Heather Marie Keeling Email: [email protected] Access this Article Online www.idjsr.com Use the QR Code scanner to access this article online in our database Article Code: IDJSR SE 0165 Quick Response Code Introduction The mechanisms linking oral and systemic health are of utmost importance in patients with chronic inflammatory disease. In this particular case, the patient presented with chronic poorly diabetes mellitus, which is known to have a negative impact on periodontal health. Periodontal pathogens have been found to illicit biomolecular responses that result in poor glycemic control. Aside from diabetes the patient was also being treated for cardiovascular disease; hypertension and also for hyperlipidemia. Patients with diabetes have a greater chance of cardiovascular complications than those without diabetes(1). Periodontitis and cardiovascular disease are linked through several biomolecular pathways, including the inflammatory response to bacteremia. The patient also presented a 10 pack-year history of smoking. Patient History The present case report is about a 65-year-old female patient who was admitted to the Dalhousie University Dental Clinic for an initial screening and treatment planning. The patient had a chief complaint of, “My teeth are getting loose and my private dentist’s fees are too expensive, so I cannot afford my needed dental treatments”. Detailed history of her medical The patient’s medical history included type II diabetes mellitus (DM), hypertension, hyperlipidemia, and gout. The patient was diagnosed with DM 12 years prior, and her average blood glucose was relatively well controlled (5-7mmol/L). The prescribed medications for treatment of DM were Metformin, 500mg 1x/day, and Gliclazide, 30mg 2x/day. Dental implications to consider are taste disorder and morning scheduling of appointments to decrease risk of stress induced hypoglycemia. The patient was diagnosed with hypertension and hyperlipidemia one year prior to initial dental visit at the school. The patient’s blood pressure was well controlled (120/80 mmHg) with Sandoz-Condesartan 12.5mg, 1x/day. The patient’s hyperlipidemia was well controlled with Rosuvastatin, 40mg 1x/day. The patient also reported a history of Gout. The last episode was in February 2013 and she had two episodes within the prior year. The patient believed that specific foods and drinks brought on the episodes. Dental history The patient stated that in the last 4 years her teeth had become progressively looser. She also reported sensitivity to hot and cold of the remaining maxillary teeth. Prior to being treated at the dental school, the patient reported no flossing and brushing once per day with a manual toothbrush. The last time she had seen a dentist was 6 years prior. Social habits The patient does not currently smoke but reported that she quit smoking over 20 years ago. She confessed to smoking about a half pack of cigarettes a day for 20 years. The patient does not consume alcohol. Clinical and radiographic findings Extraoral findings were within normal limits. Intraoral clinical and radiographic findings indicated questionable prognosis for all of her teeth. A panoramic radiograph and full mouth series of intraoral radiographs (including vertical bitewings and periapical) were taken at the screening International Dental Journal of Student’s Research, April - June 2015;3(2):58-65 {59} appointment (September 2013) (see Appendix A).Gingival margins were red and edematous and generalized heavy calculus and plaque accumulation (PI=100%) were noted. The following teeth were missing: 3-8, 3-6, 4-5 and 4-6. Full mouth probing depths, recession, clinical attachment level, mobility, and furcation involvement were recorded. The deepest pockets were present at 4-3, 4-4, and 4-7. More than half of her teeth showed generalized advanced bone loss with CAL ranging from 8mm to 12mm plus mobility of grade 2 or 3.The 4 lower incisors were splinted with a composite periodontal splint. The maxillary teeth were especially sensitive to cold, air, and water. (See Appendix B for periodontal charting). There was sinus pneumatization due to loss of the posterior maxillary teeth (2-6) as well. Diagnosis Upon completion of the initial comprehensive examination a diagnosis of generalized advanced chronic periodontitis was made for her based on Armitage classification (2). Modifying factors included diabetes mellitus and a history of smoking. Prognosis Initial comprehensive examination determined the prognosis of all maxillary teeth, 4-1, 3-1, 4-3 and 4-7 to be hopeless, according to McGuire’s classification system(3), and teeth4-2 and 3-2 had questionable prognosis as well. Two major factors contributing to the patient’s periodontal status could be due to her poor oral hygiene and systemic medical condition besides being a former smoker. The patient mentioned that her diabetes was recently controlled, however, her fasting blood glucose level was still greater than the normal range (7.5mmol/L) on the morning of most of her appointments. Treatment Plan Initial periodontal therapy; included full arch scaling and root planing (SRP) with hand instruments and ultrasonics under local anesthetic in 2 separate appointments, following by a reevaluation after 6 weeks. At the time of the reevaluation (Appendix B), periodontal and prosthodontics consultations were done. Because of the hopeless prognosis of the maxillary teeth, clearance was planned for the maxillary arch followed by fabrication of a complete upper denture. In the mandibular arch, teeth with hopeless prognoses were extracted. The prognoses of teeth 48, 44, 33, 34, 35 and 37 had improved from questionable to poor, so it was decided to maintain these teeth (Fall 2013). Since the patient’s oral hygiene was still not acceptable (PI>25%) a second round of scaling and root planning was completed under local anesthesia with the hope of patient’s selfperformed oral hygiene (including brushing and flossing) would improve. After each appointment oral hygiene instructions were reviewed, following by emphasizing the important association between her periodontal status and her medical systemic condition (4). Oral hygiene instruments that were suggested to the patient were manual or electric tooth brushing, and the use of floss, super floss, Sulcabrush, and interproximal brush. Oral hygiene effectiveness was recorded at each appointment. Unfortunately, the BOP and plaque index measurements at all reevaluations and recall examinations were not ideal. After eight weeks a the second reevaluation was done (Winter 2014) (Appendix B) at which time better oral hygiene status was noted with decreasing probing pocket depths so we were able to proceed with the prosthodontic treatment including fabrication of the lower removable partial denture that was planned for her at the initial reevaluation. The splint was also removed from the lower incisors at the end of the second round of SRP, so they could be extracted. Oral hygiene was again stressed with the patient as she was planned for a L-RPD; which itself may cause plaque accumulation. The patient’s diagnosis after the extraction of all hopeless teeth was generalized moderate chronic periodontitis. Due to inadequate width of keratinized gingiva on the facial aspect of teeth 33, 34, 35 and44 (<2mm)(5), after the periodontal consultation by one of the periodontics instructors two free gingival grafts (FGG) were planned for her prior to the L-RPD treatment. In the meantime, we proceed with the CUD. Upon completion of the treatment in the graduate periodontics clinic by a periodontics resident (SD) (Appendix C for clinical photographs), the L-RPD treatment started. During the fabrication of the CUD, due to the curve of Spee of the lower teeth and the mobility associated with the 3-2 and 4-2, it was determined that these two teeth should also be extracted prior to RPD fabrication. Extractions were completed on April 16, 2014. In Fall 2014 the patient was seen for a recall examination including complete periodontal charting. BOP and plaque index had again improved since the last appointment. It was decided that the patient should continue to be seen on 3-monthperiodontal recalls and fabrication of the L-RPD could now begin. Three-month recall was suggested because of the patient’s periodontal status (moderate chronic periodontitis) and the presence of modifying factors (diabetes). Recall intervals that are 3months apart will allow for removal of subgingival plaque containing periodontal pathogens, close monitoring of periodontal condition to prevent further attachment loss, and to reinforce optimal oral hygiene(6). Also, since the patient will be wearing a partial denture in International Dental Journal of Student’s Research, April - June 2015;3(2):58-65 {60} the near future it is especially important to ensure that the patient is keeping up with oral hygiene, as partial dentures are known to act as a plaque trap. Discussion Modifying factors Diabetes mellitus Poorly controlled diabetes is linked to a variety of oral health complications. Patients with poorly controlled diabetes have an increased risk of oral infections, decreased salivary flow and impaired wound healing (7). They also respond differently to bacterial plaque due to increased levels of cytokines in the gingival tissues. Also increased glucose concentration in the crevicular fluid may change the bacterial composition of the oral microbiota(8). Periodontal disease can also have systemic effects. Systemic response to periodontal disease results in an increase in inflammatory mediators such as tumor necrosis factor-alpha and interleukins (6). These inflammatory mediators may result in increased insulin resistance and therefore make it more difficult for the patient to maintain glycemic control (1, 7, 9). Some studies have indicated that periodontal therapy may decrease HbA1c levels by approximately 0.4% (7, 10). Cardiovascular Disease: Hyperlipidemia &Hypertension Periodontitis and cardiovascular disease are linked through their inflammatory effects. Both diseases lead to chronic states of inflammation and have effects on the vasculature if appropriate treatment is not initiated. Stimulation of inflammatory mechanisms by periodontal pathogens has been shown to have negative effects on atherosclerotic pathogenesis (1, 9). Former smoker The impacts of smoking on oral health are numerous. Smoking results in an increased risk for periodontitis due to impaired microcirculation, inhibition of neutrophil function, and increased calculus formation. Patients who smoke present with greater bone loss than those that don’t and have decreased response to periodontal therapy (11). The cessation of smoking can decrease the progression of periodontitis, however attachment loss will not be regained (11). In order to see the most optimal results following initial therapy coronal and root surfaces need to be completely debrided and free of calculus and plaque deposits (12). When initial probing depths are greater than 6mm, then surgical debridement is favoured and greater clinical attachment gain can be achieved along with a greater reduction in probing depths (12). When pockets are greater than 6mm, instrumentation of deeper root surfaces may not be achievable and, therefore, flap surgery provides better access (12). Unfortunately, in this patient’s case due topoor oral hygiene and constant high PI (>25%) we did not proceed with surgical debridement due to the need for a low plaque score for optimal healing to occur (12). Limitations of patient treatment in the undergraduate clinic: Unfortunately, due to the academic schedule it is difficult for treatment in undergraduate clinics in dental faculties to permit ideal timing in terms of periodontal treatment. At Dalhousie, in the summer only the third year students provide treatments in the clinic. This may contribute to longer wait times for patient treatment. In this case, more than half the patient’s teeth had hopeless prognoses and were ultimately be extracted. The patient’s oral health condition was extremely poor and she also had a large draining periodontal abscess distal to the 4-3. Ideally, the patient should have been seen immediately in the clinic after treatment planning instead of having waited nearly 6 months for removal of the hopeless teeth. International Dental Journal of Student’s Research, April - June 2015;3(2):58-65 {61} Appendix A March 27, 2013 International Dental Journal of Student’s Research, April - June 2015;3(2):58-65 {62} Appendix B September 19, 2013 November 26, 2013 March 4, 2014 September 10, 2014 International Dental Journal of Student’s Research, April - June 2015;3(2):58-65 {63} Appendix C May 29, 2014 44 graft site Pre-treatment donor site (palate) 33, 34, 35 graft site Pre-treatment graft site (44) 44 graft in place Pre-treatment graft site (33, 34, 35) 33, 34, 35 graft in place International Dental Journal of Student’s Research, April - June 2015;3(2):58-65 {64} June 5, 2014 44 graft site 1-week post op 33, 34, 35 graft site 1-week post op 33, 34, 35 graft site 6 weeks post op Donor site (palate) 6 weeks post op March 2015 Donor site (palate) 1-week post op Delivery of L-RPD Lower Left Side View July 17, 2014 44 graft site 6 weeks post op Delivery of L-RPD Lower Right Side View International Dental Journal of Student’s Research, April - June 2015;3(2):58-65 {65} References 1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. Masayuki Shimoe TY, Yoshhiro Iwamoto, Nobuyuki Shiomi, Hiroshi Maeda, Fusanori Nishimura and Shogo Takashiba. Chronic Periodontitis with Multiple Risk Factor Syndrome: A Case Report. Journal of the International Academy of Periodontology. 2011;13(2):40-7. Armitage GC. Development of a classification system for periodontal diseases and conditions. Ann Periodontol. 1999 Dec;4(1):1-6. Review. MK M. Prognosis versus actual outcome: a long-term survey of 100 treated periodontal patients under maintenance care. Journal of periodontology. 1991;62(1):51-8. Weinstein P, Milgrom P, Melnick S, Beach B, Spadafora A. How effective is oral hygiene instruction? Results after 6 and 24 weeks. Journal of public health dentistry. 1989 Winter;49(1):32-8. PubMed PMID: 2911075. Kim DM NR. Periodontal soft tissue non-root coverage procedures: a systematic review from the AAP Regeneration Workshop. Journal of periodontology. 2015;86(2):56-72. Darcey J, M. A. (2011). "The rationale for the three monthly periodontal recall interval: a risk based approach." Br Dent J 211(8): 279-385. Casanova L, Hughes FJ, Preshaw PM. Diabetes and periodontal disease: a two-way relationship. British dental journal. 2014 Oct;217(8):433-7. Wang TF, Jen IA, Chou C, Lei YP. Effects of periodontal therapy on metabolic control in patients with type 2 diabetes mellitus and periodontal disease: a meta-analysis. Medicine. 2014 Dec;93(28):e292. Li C, Lv Z, Shi Z, Zhu Y, Wu Y, Li L, et al. Periodontal therapy for the management of cardiovascular disease in patients with chronic periodontitis. The Cochrane database of systematic reviews. 2014;8:CD009197. Simpson TC, Needleman I, Wild SH, Moles DR, Mills EJ. Treatment of periodontal disease for glycaemic control in people with diabetes. The Cochrane database of systematic reviews. 2010 (5):CD004714. Sham A, Cheung L.K., Jin L.J., Corbet E.F. The Effects of Tobacco Use on Oral Health. Hong Kong Med J. 2003 August;9(4):271-277 Heitz-Mayfield LJ, Lang NP. Surgical and nonsurgical periodontal therapy. Learned and unlearned concepts. Periodontology 2000. 2013 Jun;62(1):218-31. __________________________________________________ International Dental Journal of Student’s Research, April - June 2015;3(2):58-65 {66} SYSTEMATIC REVIEW Sinus lift grafting materials and immediate implant placement: A systematic review Dr. Kashif Hafeez1 Dr. Aiyesha Wahaj2 Dr. Muhammad Sohail Zafar3 Dr. Sana Shahab4 1 BDS, MFDS(RCSI), FFDS(RCSI), FFDS(RCSEd) Postgraduate Dental Foundation Trainer, Oxford Deanery; Broadshires Dental Practice, Carterton, Oxon, OX18 1JA, UK 2 BDS, FCPS. Postgraduate Resident, Department of Orthodontics, Dr. Ishrat-ul-Ebad KhanInstitute of Oral Health Sciences, Dow University, Karachi, Pakistan 3 BDS, MSc, PhD, FADI, FICD, Assistant Professor, College of Dentistry, Taibah University, Madinah Al Munawwarah, Saudi Arabia. 4 BDS, MSc. Department of Dental Materials Science, Sir Syed College of Medical Sciences for Girls, Karachi, Pakistan Corresponding Author Dr. Muhammad Sohail Zafar E-mail: [email protected] Access this Article Online www.idjsr.com Use the QR Code scanner to access this article online in our database Article Code: IDJSR SE 0166 Quick Response Code Abstract Sinus lift is one of the intricate methods of increasing bone height in the posterior maxilla. Graft materials are used to provide the height and hence increase the implant support and success rate. Successful osseointegration of dental implants required a stable and sufficient amount of bone. There are different types of bone grafting materials including autogenous bone grafts, allografts and xeno grafts. Other newly used materials such as platelet rich plasma is also found to have optimal results. The current study was aimed to assess the implications of dental implants after immediate sinus augmentation and type of graft materials which are suitable to support the sinus lift procedures. A data search was performed based on available electronic data bases (Cochrane data base, Medline/PubMed) for articles published from19902013.Sinus lift is combined with various graft materials to increase the bone height. The long term survival of implant depends on surgical techniques, bone volume, graft materials and Implant surface features. Keywords: Endosseous implant, Bone grafting materials, Hydroxyapatite (HA). Introduction Surgical placement of dental implant is a demanding technique particularly if alveolar bone height is compromised in the posterior maxillary region. Multiple surgical methods have continuously been adopted to encounter these clinical problems including reduced alveolar ridge height and density13 .The most common surgical procedure for obtaining clinically adequate bone height before the placement of endosseous implants in the maxilla is grafting of the maxillary sinus floor. The sinus augmentation technique was discovered about forty years ago. This was achieved using the autogenous cancellous bone material from the lateral iliac crest and repaired though Caldwell-Luc yechnique. Later on, various methods were discovered in the precision of the sinus grafting techniques1-6. This was performed to make the procedure more comprehensive yet clinically an effective way to increase bone height. A wide range of materials including allografts, xenografts and alloplastic grafts have been used for bone substitution to make implantation more predictable and successful clinically5,7-9. Implant success is found to have dictated by primary stability factors such as implant diameter, shape, thread forms and pitch values. Secondary stability factors included the host environment where bone density plays a vital role in their placement and successful osseointegration. For example, osseointegration can be enhanced using osteogenic surface coated dental implants10. Radiographic techniques including cone beam tomography is frequently used for anatomic assessment of orodental tissues11-13. For example, computed tomography is used to assess the core basal value and density of alveolar bone in order to make sinus augmentation valuable in long term. This International Dental Journal of Student’s Research, April - June 2015;3(2):66-71 {67} review discusses the significance of sinus lift procedures with immediate dental implant placement in combinations with different graft materials. Based on previous clinical studies, clinical survival predictability of graft materials and implant success rate has been discussed. Material and Methods A data search was performed based on available electronic data base(Cochrane, Medline and PubMed) for studies published during 1990-2013. The search strategy was based on search terms such as; endosseous implant, bone grafting, sinus lift, and implant survival. Inclusion criteria included the sinus lift procedure with significant results using proper implant techniques. Table 1: Lekholm classification scheme for evaluating bone and dental implant14 Type Criteria I Dense bone that delivers great cortical anchorage; limited vascularity II Delivers better cortical anchorage for primary stability and better vascularity III Soft bone texture IV Least successful soft bone texture Exclusion criteria included syndromic patients, interrupted treatment timings, bone graft failures with no conclusive results, immunocompromised, postoperative infections, autoimmune diseases, history of trauma or re-implant procedures, tumor and systematic metabolic diseases. In order to evaluate the quality of bone and dental implant placement, Lekholm classification scheme14 was used (Table 1). Results Initial search recovered 3510 peer reviewer papers (figure 1) and reduced to 1724 after filtering out duplicate papers. After going through the titles, abstracts and full texts of 279 papers we excluded 151 papers because of high risk of bias. Considering the inclusion criteria carefully, only 40 papers were included in the review. Figure 1: Article screening criteria used in this study. PubMed/MEDLINE and Cochrane electronic databases were searched for articles published from 2000 to 2013. International Dental Journal of Student’s Research, April - June 2015;3(2):66-71 {68} The key outcome of inclusive research studies including the type of graft augmentation has been summarized (table2). Table 2: Reviewed studies with basic research outcome Researcher Graft type for augmentation Cochrane1 Not specified Lazzara et al2 Not mentioned Khang et al3 Not specified Wallace et al4 Del et al5 Stach et al6 Autogenous allograft/ direct Autogenous composite/ non autogenous Not specified Peleg et al15 Not specified Winter et al16 Not specified Peleg et al17 Lozada et al18 Hallman et al19 Engelke20 Not specified Autogenous Bovine HA and autogenous bone Particulate alloplastic bone (autogenous) and blood Autogenous Autologous calvarial bone, human recombinant tissue factor, platelet plasma & tetracycline. Deproteinated bovine bone + platelet rich plasma Autogenous Iliac corticocancellous bone Autogenous & ePTFE membrane Alloplastic Autogenous Autologous Autogenous Organic bovine bone with/without autogenous bone. Autogenous Autogenous/allograft/Gore-Tex membrane Autogenous cancellous bone/HA Autogenous Autogenous bone/xenograft mixture 2:1 Resorbable membrane, collagen and inorganic bone mineral McCarthy et al21 Philippart et al22 Rodriguez et al23 Stricker et al24 Bloomqvist et al25 Hurzeler et al26 Zinner et al27 Block et al28 Daelemans et al29 Block et al30 Wallace et al31 Karabuda et al32 Fugazzotto33 Kaptein et al34 Van et al35 Hatano et al36 Schwarz et al37 Valentine et al38 Emmerich et al39 Leonardis et al40 Khoury et al41 Lekholm et al14 Porous bone minerals Various Calcium sulphate Autogenous Inlay/onlay graft Peleg et al42 Lovenzoni et al43 Autogenous Autogenous Main Outcome Sand blasted/acid etched titanium implants promote osseous contact than plasma sprayed. Cumulative implant survival rate 99.8% at 10.5 months loading in non-complicated implants. Clinically investigation suggested that functional loading is possible at 2 months. Cumulative success rate for post loading three year 96.8% (acid etched) and 84.8% (machined surface). Survival rate of implant in augmented sinus ~92.6%. Bone substitutes are successful for sinus augmentation. Cumulative success rate (4 years) for machined implants 92.7% (dense bone) & 88.2% (poor bone) Immediate implant insertion can be a likely choice for patients with 1-2mm of vertical residual bone height. In atrophic posterior maxilla, primary stability was achieved with tapered implants. Simultaneous implant placement favorable results. Less dense bone required large diameter implants. Acceptable short term results and less resorption. Adequate bone height achieved. Sufficient bone volume achieved. High bone regeneration capacity. Favorable results obtained. Grafted bone showed good prognosis. Total implant survival rates report favorable. 98.8% survival rate. Good alternative Good on functional stability. Favorable results. Significant volume of bone for augmentation Vital bone formation in sinus graft when a membrane is placed. Implant survival similar in both types Overall survival rate 95.9%. Favorable response 97.5%. Cumulative success rate 82%. Favorable response. Favorable response. The survival rate of implant placed under repaired membrane correlates inversely with size of perforation; less than 5mm showed good results. Good osteoconductive properties. Elevation with osteotome; short term clinical success. Suitable material for sinus augmentation. Best bone regeneration. Implant placement (23%failure). Inlay/onlay technique; 60% less favorable results. Favorable results. Success rate of 92.7% for implants. International Dental Journal of Student’s Research, April - June 2015;3(2):66-71 {69} Pal et al44 Butz et al45 Guers et al46 Kahnberg et al47 Autogenous Alloplastic Autogenous Autogenous A number of researchers4,5,18-21,24-26 reported a high success rate for using either autogenous bone grafts or composite materials containing autogenous bone (Table 2). Use of alloplastic grafts also produced favorable results27,45. Zinner et al27 described alloplastic grafts as a good alternative to autogenous bone grafts. Regarding the applications of inorganic biomaterials, bioactive materials based on calcium and phosphates have been used either alone or in combination with natural organic materials. Porous bone minerals showed great osteoconductive properties38. Leonardis et al40 has reported calcium sulphate as a suitable material for sinus lift applications. Discussion There are various techniques for sinus augmentation such as lateral window, crestal approach, summers osteotomy, bone aided augmentation. The most popular technique for sinus lift is found to be lateral window with autogenous corticocancellous grafts. Autogenous bone grafts have always been considered the most effective standardized grafting material due toosteoinductiveand osteoconductive potential 1-6,15,16. Various alternative materials havealso been used in this context, however compromising the osteoinductive potential. The property of biomaterials in providing graft maturation and effective provision to the endosseous implants is the most significant element believed for the success of sinus graft augmentation procedures17-24. Implants placed in grafts composed of a combination of autogenous bone and synthetic materials found to have better survival rates than implants placed using the autogenous graft only17-24. Such response is probably due to its high resorption values. The reviewed studies explained that a majority of implants had textured surface followed by machined surface. Textured surface implants have shown significant results (p<0.05) contrast to machined (p>0.05). No association was observed in context to bone graft materials. This might refer to the adequate results with rough surfaced implants in immunological risk patients or those who have insufficient bone this seems regardless in bone with adequate height and density1-5.Direct implant placement is usually a recommended protocol in such cases25-31. Primary implant stability and graft is related to adequate bone height. Delayed implant placement is not recommended for badly destructed alveolar ridge with no proper implant base. Significant gain in bone height; mean 8.5mm, Favorable success rate. Good response. Favorable results. Implant surgical procedures are found to have a profound effect on implant placement32-39.This included significant results (p<0.05) using lateral swing door technique, osteotome sinus elevation. Clinically, these techniques provided a significant amount of bone height for implant placement. A recent study by Pal et al44 explained that the increase in bone height found to be significantly greater with lateral antrostomy than in indirect method by crestal approach. This might be beneficial when more than 6mm bone height present and increase required up to 4mm. In case of advanced bone loss, a direct method using lateral antrostomy is beneficial. Implant survival comparison showed no significant differences14,40-47. Reviewed studies (Table 2) showed different types of graft materials amongst which autogenous iliac crest corticocancellous were the commonest one. A combination of autogenous and xenograft have been used because of better success predictability in relation to less bone resorption postoperatively36.Advanced graft materials such as platelet rich plasma, xenograft mixture with autogenous and deproteinated bovine have showed promising results when used in conjuncture with autogenous graft. These graft types provided stabilized bone base for immediate implant placement following sinus augmentation. These graft materials also assessed for cross antibody reaction and resorption, and later found to be insignificant in this regard (p>0.05). A significant failures with inlay/onlay graft (p<0.05) upon three year interval has been reported14.Membranes (absorbable and nonabsorbable) found no significant effects (p<0.05) in relation to osseointegration37. Although the mean values showed good results with absorbable however no statistically significant effects. Short term data explicitly suggested that implant placement after sinus augmentation is found to be a stable procedure however; a large number of longitudinal data is required. Considering the complications of such procedures, sinus perforation was found to be the most frequent however not affecting the osseointegrationin case of perforation 5mm or less. The repaired sinus with graft and ePTFE membranes26 is also found to have significant good prognosis in this regard, however long term prognosis is required to be assessed. Sinus infection can affect the osseointegration potentially. None of the included studies reported the sinus infection postoperatively following the sinus augmentation. Natural silk based materials have been reviewed recently for bone grafting and International Dental Journal of Student’s Research, April - June 2015;3(2):66-71 {70} regeneration48. From biomaterials prospective, there is an intense need of new materials for these applications. The limitations of clinical studies included inadequate sample size, lack of integrated systemized similar approaches and variability in data collection. All accounts towards the specific need of more rational case control and randomized clinical trials. This approach can further encompass the various human physiologically mediated conditions required to be discussed. There is also found to have constant need of long term follow up related to implant stability. Conclusion 8. 9. 10. 11. 12. Predictability of sinus augmentation is substantially based upon factors that need further understanding. This comprehensively explained statistically significant results using rough surfaced implant compared to the smooth surface. The most commonly used graft material is autogenous corticocancellous iliac crest. New graft materials (such as xenografts, deprotonated bovine, platelet rich plasma) are being used in combination with autogenous graft sand providing promising basal support forimplant insertion. Success rate improves remarkably with immediate implant placement in a good quality basal bone support. However, immediate implant placement is not recommended if site is lacking a good quality bone support. 13. 14. 15. 16. 17. References 1. 2. 3. 4. 5. 6. 7. Cochran D, Schenk R, Lussi A, Higginbottom F, Buser D. Bone response to unloaded and loaded titanium implants with a sandblasted and acid-etched surface: A histometric study in the canine mandible. J Biomed Mater Res. 1998; 40: 1-11. Lazzara RJ, Porter SS, Testori T, Galante J, Zetterqvist L. A prospective multicenter study evaluating loading of osseotite implants two months after placement: One‐Year results. J Esthetic Restorative Dent. 1998; 10: 280-9. Khang W, Feldman S, Hawley C, Gunsolley J. A multi-center study comparing dual acid-etched and machined-surfaced implants in various bone qualities. J Periodontol. 2001; 72: 1384-90. Wallace SS, Froum SJ. Effect of maxillary sinus augmentation on the survival of endosseous dental implants. A systematic review. Ann periodontol. 2003; 8: 328-43. Del Fabbro M, Testori T, Francetti L, Weinstein R. Systematic review of survival rates for implants placed in the grafted maxillary sinus. J Prosthet Dent. 2005; 94: 266. Stach RM, Kohles SS. A meta-analysis examining the clinical survivability of machined-surfaced and osseotite implants in poor-quality bone. Implant Dent. 2003; 12: 87-96. Blus C, Szmukler-Moncler S, Salama M, Salama H, Garber D. Sinus bone grafting procedures using 18. 19. 20. 21. 22. 23. ultrasonic bone surgery: 5-year experience. Int J Periodontics Restorative Dent. 2008; 28: 221-9. Wahaj A, Hafeez K, Zafar MS. Role of bone graft materials for cleft lip and palate patients: A systematic review. Saudi J Dent Res 2015;Epub Ahead of print; doi:10.1016/j.sjdr.2015.02.001. Semb G. Alveolar bone grafting. Front Oral Biol. 2012; 16: 124-36. Javed F, Vohra F, Zafar S, Almas K. Significance of osteogenic surface coatings on implants to enhance osseointegration under osteoporotic-like conditions. Implant Dent. 2014; 23: 679-86. Zafar MS, Javed E. Extraoral radiography: An alternative to intraoral radiography for endodontic (root canal system) length determination. Eur Sci J. 2013; 9: 51-61. Alrahabi M, Zafar MS. Evaluation of root canal morphology of maxillary molars using cone beam computed tomography. Pak J Med Sci. 2015; 31: 42630. Zafar MS AM. Cone beam computed tomography for exploring morphology of mandibular first molar. British J Med Medical Res. 2015; 6: 514-21. Lekholm U, Wannfors K, Isaksson S, Adielsson B. Oral implants in combination with bone grafts. Int J Oral & Maxillofac Surg. 1999; 28: 181-7. Peleg M, Mazor Z, Chaushu G, Garg AK. Sinus floor augmentation with simultaneous implant placement in the severely atrophic maxilla. J Periodontol. 1998; 69: 1397-403. Winter AA, Pollack AS, Odrich RB. Placement of implants in the severely atrophic posterior maxilla using localized management of the sinus floor: A preliminary study. Int J Oral Maxillofac Implants. 2002; 17: 687-95. Peleg M, Garg AK, Mazor Z. Predictability of simultaneous implant placement in the severely atrophic posterior maxilla: A 9-year longitudinal experience study of 2132 implants placed into 731 human sinus grafts. Int J Oral Maxillofac Implants. 2006; 21: 94-102. Lozada JL, Emanuelli S, James RA, Boskovic M, Lindsted K. Root-form implants placed in subantral grafted sites. J Calif Dent Assoc. 1993; 21: 31-5. Hallman M, Hedin M, Sennerby L, Lundgren S. A prospective 1-year clinical and radiographic study of implants placed after maxillary sinus floor augmentation with bovine hydroxyapatite and autogenous bone. J oral and maxillofac surg. 2002; 60: 277-84. Engelke W, Schwarzwaller W, Behnsen A, Jacobs HG. Subantroscopic laterobasal sinus floor augmentation (SALSA): An up-to-5-year clinical study. Int J Oral Maxillofac Implants. 2003; 18: 135-43. McCarthy C, Patel RR, Wragg PF, Brook IM. Sinus augmentation bone grafts for the provision of dental implants: Report of clinical outcome. Int J Oral Maxillofac Implants. 2003; 18: 377-82. Philippart P, Brasseur M, Hoyaux D, Pochet R. Human recombinant tissue factor, platelet-rich plasma, and tetracycilne induce a high-quality human bone graft: A 5-year survey. Int J Oral Maxillofac Implants. 2003; 18: 411-6. Rodriguez A, Anastassov GE, Lee H, Buchbinder D, Wettan H. Maxillary sinus augmentation with deproteinated bovine bone and platelet rich plasma with simultaneous insertion of endosseous implants. J Oral and Maxillofac Surg. 2003; 61: 157-63. International Dental Journal of Student’s Research, April - June 2015;3(2):66-71 {71} 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. 37. 38. Stricker A, Voss PJ, Gutwald R, Schramm A, Schmelzeisen R. Maxillary sinus floor augmention with autogenous bone grafts to enable placement of SLA‐surfaced implants: Preliminary results after 15– 40 months. Clin Oral Implants Res 2003; 14: 207-12. Blomqvist JE, Alberius P, Isaksson S. Retrospective analysis of one-stage maxillary sinus augmentation with endosseous implants. Int J Oral Maxillofac Implants 1996; 11: 512-21. Hurzeler MB, Kirsch A, Ackermann KL, Quinones CR. Reconstruction of the severely resorbed maxilla with dental implants in the augmented maxillary sinus: A 5-year clinical investigation. Int J Oral Maxillofac Implants 1996; 11: 466-75. Zinner ID, Small SA. Sinus-lift graft: Using the maxillary sinuses to support implants. J Am Dent Assoc. 1996; 127: 51-7. Block MS, Kent JN. Sinus augmentation for dental implants: The use of autogenous bone. J Oral Maxillofac Surg 1997; 55: 1281-6. Daelemans P, Hermans M, Godet F, Malevez C. Autologous bone graft to augment the maxillary sinus in conjunction with immediate endosseous implants: A retrospective study up to 5 years. Int J Periodontics Restorative Dent. 1997; 17: 27-39. Block MS, Kent JN, Kallukaran FU, Thunthy K, Weinberg R. Bone maintenance 5 to 10 years after sinus grafting. J Oral Maxillofac Surg. 1998; 56: 70614. Wallace SS, Froum SJ, Cho SC, Elian N, Monteiro D, Kim BS, Tarnow DP. Sinus augmentation utilizing anorganic bovine bone (bio-oss) with absorbable and nonabsorbable membranes placed over the lateral window: Histomorphometric and clinical analyses. Int J Periodontics Restorative Dent. 2005; 25: 551-9. Karabuda C, Arisan V, Hakan Ö. Effects of sinus membrane perforations on the success of dental implants placed in the augmented sinus. J Periodontol. 2006; 77: 1991-7. Fugazzotto PA, Vlassis J. Long-term success of sinus augmentation using various surgical approaches and grafting materials. Int J Oral Maxillofac Implants. 1998; 13: 52-8. Kaptein ML, de Putter C, de Lange GL, Blijdorp PA. Survival of cylindrical implants in composite grafted maxillary sinuses. Joral maxillofac surg. 1998; 56: 1376-80. van den Bergh J, ten Bruggenkate CM, Krekeler G, Tuinzing DB. Sinus floor elevation and grafting with autogenous iliac crest bone. Clin Oral Implants Res. 1998; 9: 429-35. Hatano N, Shimizu Y, Ooya K. A clinical long‐term radiographic evaluation of graft height changes after maxillary sinus floor augmentation with a 2: 1 autogenous bone/xenograft mixture and simultaneous placement of dental implants. Clin Oral Implants Res. 2004; 15: 339-45. Schwartz-Arad D, Herzberg R, Dolev E. The prevalence of surgical complications of the sinus graft procedure and their impact on implant survival. J Periodontol. 2004; 75: 511-6. Valentini P, Abensur D, Wenz B, Peetz M, Schenk R. Sinus grafting with porous bone mineral (bio-oss) for implant placement: A 5-year study on 15 patients. Int J Periodontics Restorative Dent. 2000; 20: 245-53. 39. Emmerich D, Att W, Stappert C. Sinus floor elevation using osteotomes: A systematic review and metaanalysis. J Periodontol. 2005; 76: 1237-51. 40. De Leonardis D, Pecora GE. Augmentation of the maxillary sinus with calcium sulfate: One-year clinical report from a prospective longitudinal study. Int J Oral Maxillofac Implants. 1999; 14: 869-78. 41. Khoury F. Augmentation of the sinus floor with mandibular bone block and simultaneous implantation: A 6-year clinical investigation. Int J Oral Maxillofac Implants. 1999; 14: 557-64. 42. Peleg M, Mazor Z, Garg AK. Augmentation grafting of the maxillary sinus and simultaneous implant placement in patients with 3 to 5 mm of residual alveolar bone height. Int J Oral Maxillofac Implants. 1999; 14: 549-56. 43. Lorenzoni M, Pertl C, Wegscheider W, Keil C, Penkner K, Polansky R, Bratschko RO. Retrospective analysis of frialit-2 implants in the augmented sinus. Int J Periodontics Restorative Dent. 2000; 20: 255-67. 44. Pal U, Sharma NK, Singh R, Mahammad S, Mehrotra D, Singh N, Mandhyan D. Direct vs. indirect sinus lift procedure: A comparison. Nat j maxillofac surg. 2012; 3: 31-7. 45. Butz SJ, Huys LW. Long-term success of sinus augmentation using a synthetic alloplast: A 20 patients, 7 years clinical report. Implant Dent. 2005; 14: 36-42. 46. Geurs NC, Wang IC, Shulman LB, Jeffcoat MK. Retrospective radiographic analysis of sinus graft and implant placement procedures from the academy of osseointegration consensus conference on sinus grafts. Int J Periodontics Restorative Dent. 2001; 21: 517-23. 47. Kahnberg K, Nilsson P, Hirsch J, Ekestubbe A, Gröndahl K. Sinus lifting procedure. Clin Oral Implants Res. 2001; 12: 479-87. 48. Zafar MS, Al-Samadani KH. Potential use of natural silk for bio-dental applications. J Taibah Uni Med Sci. 2014; 9: 171-7. __________________________________________________ International Dental Journal of Student’s Research, April - June 2015;3(2):66-71 {72} SYSTEMATIC REVIEW Laboratorial and clinical impacts of tobacco on periodontal health: A systematic review Fahad Sikander Khan1, Aisha Aziz2, Dr. Sana Shahab3, Dr. Muhammad Sohail Zafar4 1 B.D.S, MSc, Dip (Public Health), Health officer, Comfort Care, St. Albans, United Kingdom 2 B.D.S, Dip (Microbiology), General Dental Practioner, Private Clinic, Karachi, Pakistan 3 BDS, MSc. Department of Dental Materials Science, Sir Syed College of Medical Sciences for Girls, Karachi, Pakistan 4 BDS, MSc, PhD, FADI, FICD. Assistant Professor, College of Dentistry, Taibah University, Madinah Al Munawwarah, Saudi Arabia. Corresponding Author Dr. Sana Shahab E-mail: [email protected] Access this Article Online www.idjsr.com Use the QR Code scanner to access this article online in our database Article Code: IDJSR SE 0167 Quick Response Code Abstract Use of tobacco has been documented a significant risk factor for the progression of periodontal conditions. The consumption of tobacco is related to intensified depth of periodontal pockets and ligament detachment, alveolar bone loss and susceptible to tooth loss. The main aggravating part in development of periodontal disease is contributed by nicotine that is pharmaceutically active component and a primary constituent of tobacco. It have been documented that negative impact on periodontal tissues is due to hindrance in connective tissue proliferation as a result of affected gingival blood circulation and neutrophil, cytokine production caused by nicotine. Thus, abeyance of tobacco is most important part of periodontal treatments as this renders positive influence on the patients' oral and systemic wellbeing. Tobacco cessation has been included as a part of periodontal therapy and encouraged by dental practitioners. Tobacco cessation counselling is required to educate patient and for the best interest of their oral health. The aim of this review is to report the clinical and laboratory findings on periodontal health. In additions risks involved in tobacco use and possible professional intervention has been discussed. Keywords: Tobacco, Periodontal Diseases, Gingivitis. Introduction It has been known since late 1040's that tobacco exhibits detrimental effects on periodontal tissues health when association of ANUG (Acute Necrotising Ulcerative Gingivitis) was noticed with smoking1. A diverse range of periodontal diseases have been investigated observing their relationship with smoking. A substantial amount of scientific literature is available on the basis of which this review has been formulated. The basic aim and objective of this review is to conduct effective review for having clear idea of tobacco’s effect on periodontal health from the perspective adopted in clinical trial and error and to analyse the effect of tobacco consumption on periodontitis and its types (Chromic periodontitis, Aggressive periodontitis). Cross-sectional studies prove that smokers exhibit prevalence and severity of periodontal disease more than twice as compared to individuals who do not smoke. This is evidence that smoking exhibits a major connection with periodontal diseases. More cases of tooth loss are also reported in smokers with periodontal disease as it aggravates the pathological condition. According to research conducted in 19972, observe that negligence of oral hygiene among youth is ironical because they tend to be more health conscious, as the number of people visiting gyms and health clubs has increased with the onset of the new millennium. While awareness on diseases like obesity has received widespread awareness, the issues in relation to maintenance of oral health have not come into limelight. Life threatening diseases like cancer have been the chief concern of medics in relation to tobacco addiction. The knowledge on maintaining proper oral hygiene among general people is very limited. From the onset of childhood pupils are taught about brushing twice International Dental Journal of Student’s Research, April - June 2015;3(2):72-78 {73} and dental flossing to keep the menace of tooth decay at bay. However, importance of food habit and lifestyle in relation to oral health remains ignored. Observing the adverse influence of smoking on periodontal health, the aim of this review is to highlight the unfavorable impact of smoking in relation to periodontal conditions and focus on cessation of smoking, highlighting its significance on periodontal health and outcomes of diseased periodontal tissues' treatment. Literature review Epidemiological literature provides various evidences related to connection of smoking with detrimental periodontal diseases when studies of about more than two decades are reviewed3-5. The conclusions provided by Asmaand Tomar6 based on statistics from the NHANES III study, may be considered a sturdy body of evidence of nicotine as a peril factor for periodontal diseases. Authors proposed that in USA almost fifty percent of patients suffering from periodontal pathologies were smokers. It was also noted that susceptibility of periodontal disorder was directly related to rate of smoking intensity of an individual and prevalent smokers were more than 3 times susceptible to periodontal diseases in comparison to non-smokers. Literature based in clinical studies clearly reveals that smoking has deleterious effects on periodontal tissue; it causes quick and greater loss of periodontal attachment, gingival recession, pocket formation and bone loss in diseased condition7. Clinical study conducted by Mullally et al. in 2004 reviled that odds ratio between periodontal disease was as high as 14.1 for patients who smoked, exhibiting that smoking acts as the strong predator of progressive periodontitis, causing early loss of periodontal attachments and progression of other signs and symptoms.8 Connection between smoking and periodontal disease is known very clearly and proved by epidemiological studies but the true mechanism by which smoking causes deleterious effects on progression of periodontal pathology are still unknown. Methodology To obtain the desire article following database will be use like Pub Med, Medline, Lancet, Google scholar, Cochrane library and from different organization who are actively working on tobacco like TTAC (tobacco technical assistance consortium America). The following keywords will be used during searching; effects, role, tobacco, smoking, oral, mouth and periodontium or gingivitis. Articles published between 2000 and 2012 and in English language were included for review of literature. English language was selected for articles so that reviewer did not face any difficulty in understanding of materials and thus kept himself away from any potential language bias in translating materials from one language to other. Moreover with the aim that only studies relevant to the topic were included in the systematic review, this step was taken. This step of selection involved assessments of the study topic, their abstracts and list of references identified through search strategy. Full length reports of all potentially relevant articles were then accessed for eligibility assessment, which was based on review inclusion criteria 9. Reference lists of eligible studies were then again analysed for obtaining further studies. Critical appraisal stage that is the last stage in selecting studies was done and the studies whose topic did not resemble with review, those published before inclusion review years and those which were not in English were excluded 10. Even those studies in which participants’ age was not clearly stated were excluded from review (as exclusion criteria of study). All studies were searched, analysed and selected by the researcher. The study has been concluded based on three main forms of studies, epidemiological, laboratorial and studies based on perceptions of the common public on dental health care. For the epidemiological studies, the range of age varies from 18 and above. This is because; the rate of tobacco consumption is high among this particular age group. Since the consumption of tobacco is more common among males than the females, a gender based study has also been included in the research. An effort has also been made to find out cultural influences on tobacco consumption and its impact on gum diseases like periodontitis 11. For the study, the primary research outcome is formed based on the impact of tobacco intake on dental diseases like periodontitis. On the other hand secondary outcomes are measured on laboratorial studies on the relationship of tobacco consumption and periodontitis. The already existing knowledge of dental healthcare professionals and their recommended ways to cessation of smoking are also measured. Awareness of general public on dental health and impact of tobacco are also taken into account to determine the research outcomes. While conducting a research on any topic, it is very important to evaluate the quality of the studies that are selected to establish and attain the proposed research outcomes12. This is done with the objective to avoid any form of bias that may affect the research findings and outcomes. In case of the concerned research study, the articles that are included are independently assessed by utilizing a critical appraisal framework. By following the framework, the researcher has been successfully International Dental Journal of Student’s Research, April - June 2015;3(2):72-78 {74} able to critically evaluate the qualitative and quantitative methods. As per CASP or Critical Appraisal Skills Programme, the framework consists of several questions that are keenly analysed to meet up factors like relevance of the study, the results, validity, reliability and similar such 13. Through the aid of the questions for study assessment, the following segments of the entire study for instance, study design, objective of the research, target population sample, concerns related to research ethics, data analysis and findings and many such. Data Extraction and Analysis For this research study, narrative synthesis of the research outcomes has been done as the data that has been extracted from each of the articles do not bring about similar quantitative findings and analyses. The data extracted from each of the study include, the process of identification of each study, determining the characteristics of the study and the ultimate results of the study. While identifying the study, the name of the researcher, title, publication year, origin of the study and such are found. The characteristics of the study on the other hand make sure that the aim of the study, study design along with inclusion and exclusion criteria. The methods of data collection will be based on the nature and design of the study and also the source and kind of the collected data. Since, the data analysis of the study has been conducted by using the narrative approach; the study involves a process of detailed documentation. The document reflects relationship established between the understandings from all the studies and the evidence of overall validity of the studies made. As the study is not made in accordance to the meta-analysis format, an effective approach towards in detailed documentation and analysis of the research outcomes from various studies related to the topic has been adopted. The process of data analysis has been purposely made transparent by observing research ethics and values. Therefore, the entire study has been presented in a narrative format providing all possible details. This process has made way to effectively understand the relationship between tobacco consumption and development of gum diseases or periodontitis. Results After analyzing literature, we concluded different tables (1, 2 and 3) with author’s details, year of reporting, title of study, study design, which population targeted, country of study, their aims with result outcomes. Table 1: Epidemiological Studies Population/ Country Aim/Aims of Age of Study Study Author Year of Publication D.F. Kinane and I.G. Chestnutt14 Title of Study Study Design Smoking and Periodontal Disease Cross Sectional and Longitudinal Study on Patients 1361 Case (n= 873) Target Age Group 25-74 Erie County, New York State Strength of Association between Tobacco and Periodontal Brian H. Mullally15 The Influence of Tobacco Smoking on the Onset of Periodontitis in Young Persons Tobacco Use and Its Effects on the Periodontium and Periodontal Therapy Hospital Case Study 17, 22 Case, Target Age Group 17-34 years. Africa Prevalence of early onset or aggressive periodontitis in young adults. Cross sectional 12,329 Case, Target Group 18 years and above USA Reviewing potential biological mechanisms related to effects of tobacco on periodontal disease. Vandana K. Laxman and Sridhar Annaji16 Results The risk of severe tooth and bone loss is more in case of smokers. Ratios vary from 3.25 to 7.28, from light smokers to heavy smokers. Identification of significant genetic components in relation to aggressive periodontitis. Tobacco smoking significantly contributes to the development of periodontal disease. International Dental Journal of Student’s Research, April - June 2015;3(2):72-78 {75} NurcanBud uneli17 Jose LopezLopez et al 18 Effects of Tobacco Smoking on Chronic Periodontitis and Periodontal Treatment Tobacco Smoking and Radiographic Periapical Status Cross Sectional Study 550 Case Target Group 18 to 45 Turkey Impact of Tobacco Smoking on Chronic Periodontitis A positive association between smoking and various biochemical, clinical signs of periodontitis and chances of periodontitis among smokers. Case Control Study 79 Case, Target Group – 18 and above UK Radiographic investigating the relationship between tobacco smoking and periapical status. After taking into account the age, gender, total number of teeth, endodontic status, root filling quality and diabetic level, tobacco is strongly considered to be associated to periapical lesions. Table 2: Laboratorial Studies Country Aims and Objective of Study of study Author Name and Year of Publication Cristina Cunha Villar and Antonio Fernando Martorelli de Lima 19 Title of Study Smoking Influences on the Thickness of Marginal Gingival Epithelium Brazil Evaluation of the thickness of marginal gingival oral epithelium and non0smokers who have clinically healthy gingivae. FlonaM.Coll ins20 Tobacco Cessation and the Impact of Tobacco Use on Oral Health United States Types of tobacco and its prevalence among US population. Impact of tobacco use on oral health. Biochemical and generic factors associated to oral health. Recommended methods for tobacco cessation. GirishParma r, et al 21 Effect of Chewing a Mixture of Areca Nut and Tobacco on Periodontal Tissues and Oral Hygiene Status Prevalence of and Risk Indicators for Chronic Periodontitis in Males from Campeche. India Clarifying the effects of chewing areca nut and tobacco on periodontal tissue and status of oral hygiene. Determining prevalence of extension/severity of chronic periodontitis and determining the various risk factors among the policemen of Campeche, using electron probe. MirnaMinay a-Sanchez et al22 Brazil Results Among all significant negative consequences of tobacco on the periodontium, influence of tobacco on the various signs and symptoms of gingival inflammation that are induced by plaque accumulation must be necessarily considered. Smoking must be taken as a high risk factor for the development of chronic periodontal disease. Interventions made by an able dental team can help in quitting tobacco intake. The dental experts can effectively educate and motivate patients who suffer from the danger of tobacco on oral health. Interventions recommended on a routine patient care. Tobacco cessation can best be carried out under medical supervision and restrictions. Chewing areca nut and tobacco plays a causative role in the development of oral lesions, degradation of periodontal status and oral hygiene. Many of the participants were found to suffer from poor periodontal disease. Over use of tobacco along with aging causes presence of gingivitis. International Dental Journal of Student’s Research, April - June 2015;3(2):72-78 {76} Author Names and Year of Publication ZHS Lung, MGD Kelleher, R W J Porter, J Gonzalez and R F H Lung. 23 Table 3: Studies based on Knowledge and Service Delivery Design of Target Country Aims and Study Population of Study Objectives Title of the Study Poor Patient Awareness of the Relationship between Smoking and Periodontal Diseases. Cross Sectional Survey based on wellstructured interview Smoker patients attending dental clinic London, UK Investigating patient’s knowledge of the effects of smoking on periodontal disease. Yuval Vered and Harold D Sgan-Cohen.24 Self-perceived and clinically diagnosed dental and periodontal health status among young adult and their implications for epidemiological surveys. Cross Sectional Survey based on wellstructured interview 21 year old Israeli Defense Forces after being released from military services. Israel Investigating parity between selfperceived and clinically diagnosed dental and periodontal health status. ShaileeFotedar et al, et et al 25 Knowledge of Attitude Towards and Prevalence of Tobacco Use Among Dental Students in Himachal Pradesh. Cross Sectional Survey based on wellstructured interview Third year Students of Bachelor of Dental Surgery. India Assessing prevalence of tobacco and its use along with knowledge of cessation counseling among dental students in the state of Himachal Pradesh, India. Results The patients were found to be less aware of the relationship between smoking and periodontal diseases as only 6% of the total were aware of the fact. The selfassessment of individuals was found to have been low. The low level awareness will directly impact care-taking behavior and need for public health action. The prevalence of tobacco use among the students is found to be low. Though, use of skills to support quitting of smoking is necessary. having a variety of general consequences which can change the reaction of the effected person, cigarette smoking would also emerge to have substantial Regarding the “influence of tobacco smoking on the limited effects which may report for the early onset of periodontitis in young person’s” written by establishment of the disease development in, at risk Brian H.Mullally8, in which he has tried to explain young adults. Legal restrictions in the developed his views on the extent of cigarette smoking can countries like U.S or Europe has barred the tobacco actually put forth on periodontal tissues were not companies for their advertisements and sales and thus completely agreed by other theorists like. According these companies have targeted the developing 2 to Vandana K. Laxman and Sridhar Annaji , its countries as their replacement. Thus, the developing consequences are linked to the period and number of nations are susceptible to face an increase in the cigarettes consumed. The smoking habits of the prevalence of harsh periodontal situation including family members may also be significant both in view acute periodontitis. To be able to follow the strategies to behavioral influences and the probable results of of avoidance, early recognition of the disease and passive smoking 26 is of the opinion that the second quick intrusion by the dental professionals should be spot is in need of additional research. Moreover able to aim and inform the young patients about the International Dental Journal of Student’s Research, April - June 2015;3(2):72-78 Discussion {77} consequences of smoking on periodontal health. By this mode dental department will also be playing a major role in the general health and well-being of the youth. By the epidemiological study about the treatment of the individuals addicted to cigarettes, the theorists have suggested that non-surgical treatment will generally be productive in requisites of penetrating depth reduction and gingivitis. Thus the consequences of smoking on the result of periodontitis treatment may be concluded as Short term effects in terms of less gingivitis resolution, less probing depth reduction, less attachment gain among the smokers. It has been noticed and stated by López-López, J.,18 80 to 90% of the cases in long term effects are treatment failures among the smokers. He has also stated that 70-80% of the cases are not successful in the implantation among the smokers. Thus smoking does have a negative effect on the treatment procedure of the periodontitis diseases. Keeping this in mind, the patients suffering from periodontitis diseases need to be advised and counselled about the consequences of smoking. Study on effects of tobacco use on periodontal health and treatment has made evident that over the past 50 years, knowledge and concern about obnoxious effects of tobacco smoking on periodontal health has progressed significantly. Hence, in today’s world there is little doubt on the fact that tobacco smokers are at a high risk of getting affected by periodontitis. They are also found to be slow responders to periodontitis treatment. Tobacco smoking in fact, is found to have wide spread effects on oral health. The mechanisms of tobacco smoking for increased level of susceptibility to periodontitis and poorer response to remedial treatments are some of the ill-effects on over all oral health and recovery process. Another, significant outcome of the tobacco smoking is that it acts as a strong environmental factor. Smoking of tobacco interacts with the body of host and bacterial growth associated with the disease periodontitis 14. The genetic built of the host and interaction with the environmental factors like cigarette smoking makes way to further exploration of the relationship between tobacco smoking and related genetic factor of the host. The effects of tobacco smoking on formation of chronic periodontitis have also been found to impair the microcirculatory system and changes on the vascular formation system. This in turn leads to negatively influence the immune system and inflammatory reactions on healthy periodontal tissues. It is found that smokers have less number of vessels and existence of highly inflamed gingival tissues as compared to non-smokers. According to researchers it has been found that smoking of tobacco on long term basis makes a negative impact on the process of vasculature of the periodontal tissues. Also, high level exposure to long term smoking leads to gingival hyperaemia. The gingival hyperaemia is mainly caused due to increase in blood pressure levels in comparison to small yet significant sympathetic vasoconstriction on healthy gingival tissues22. Due to continued process of repeated attacks of vasoconstriction and presence of impaired revascularization caused due to cigarette smoking may lead to low rate of immune response, delayed healing process and ultimately increased risk of periodontal disease 21. It is quite evident from the study on tobacco smoking and radiographic periapical status that that there is a statistically significant association between radiographically diagnosed lesions and tobacco smoking. In this particular study the oral health maintained by the subject was quite satisfactory but due to tobacco smoking, there had been consistent bone loss. Also, among smokers it was found that functions of polymorph nuclear leukocytes, T-cell lymphocytes, antibodies, immunoglobulin’s A, G and M and macrophages are highly suppressed18. The swelling results due to the dental plaque gathering can be modified by tobacco by-products, such as cotinine, a by-product of nicotine that has a marginal vasoconstriction action that decreases gingival clinical symptoms of bleeding, redness, and oedema. The morphometric investigation showed an augment in the MET in clinically healthy gingival samples in comparison to the swelled samples in the case of both smoking and non-smoking patients. This difference did not attain statistical implication. The gingival swelling decreases the epithelial thickness and can lead to clinical ulceration19.The findings thus propose that among all the adverse effects of tobacco on the periodontium, the effect of tobacco related to the symptoms of gingival swelling caused by plaque gathering should be taken into consideration. Even though the accurate means of its power is still not very clear, yet smoking should be considered to be the main risk factor for chronic periodontal disease. Conclusions Smoking is a well-known causing and aggravating factor for periodontal problems. Smokers exhibited more than twice chances of having periodontitis. Therefore, cessation of smoking plays a significant role in the cure of periodontal diseases. Patients should be educated and motivated by their dentists to quit smoking. Dentists can have very effective and influential position, as patients tend to visit them more regularly in comparison to their physicians. However, success in cessation of smoking may be International Dental Journal of Student’s Research, April - June 2015;3(2):72-78 {78} certainly influence by mutual coordination among dentists and physicians. References 1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. Pindborg, Jens Jorgen. Tobacco and gingivitis. Journal of dental research 1947: 26(3): 261-264. Laxman, Vandana K., and Sridhar Annaji. "Tobacco use and its effects on the periodontium and periodontal therapy." J Contemp Dent Pract 2008:9(7): 97-107. Stoltenberg, J. L., Osborn, J. B., Pihlstrom, B. L., Herzberg, M. C., Aeppli, D. M., Wolff, L. F., & Fischer, G. E. Association between cigarette smoking, bacterial pathogens, and periodontal status. Journal of periodontology, 1993: 64(12), 1225-1230. Tonetti M. Cigarette smoking and periodontal diseases: Etiology and management of disease. Ann Periodontol 2001; 3:88-101. Krall EA, Dawson-Hughes B, Garvey Al, Garcia RI (1997).Smoking, smoking cessation, and tooth loss. Journal Dent Res 76:1653-1659. Tomar, Scott L., and Samira Asma. "Smokingattributable periodontitis in the United States: findings from NHANES III." Journal of periodontology 2000: 743-751. Baljoon M, Natto S, Bergstorm J JClinPeriodontol 2005:32(7):789-97. Mullally, B. H. The influence of tobacco smoking on the onset of periodontitis in young persons. TobInduc Dis 2004: 2.1, 6. Kervin, J. Methods for Business Research 1992: 3rd ed. Torrington, D. management Face to Face. 1991 4th ed. London: Prentice Hall Kothari, C. R. Research methodology: methods and techniques. New Age International. Denscombe, M (2002) Ground Rules for Good Research, Maidenhead, Open University Press 2004. Silverman, D. Interpreting Qualitative Data, Methods for Analysing Talk, Text and Interaction 1993. 4th ed. Kinane, D. F., & Chestnutt, I. G. Smoking and periodontal disease. Critical Reviews in Oral Biology & Medicine, 2000:11:356-365. Mullally, B. H. The influence of tobacco smoking on the onset of periodontitis in young persons. TobInduc Dis, 2004: 2: 6. Laxman, V. K., & Annaji, S. Tobacco use and its effects on the periodontium and periodontal therapy. J Contemp Dent Pract 2008: 9(7): 97-107. Buduneli, N. Effects of Tobacco Smoking on Chronic Periodontitis and Periodontal Treatment. López-López, J., Jané-Salas, E., Martín-González, J., Castellanos-Cosano, L., Llamas-Carreras, J. M., VelascoOrtega, E., & Segura-Egea, J. J. Tobacco smoking and radiographic periapical status: a retrospective case-control study. Journal of endodontics 2012: 38(5): 584-588. Villar, C. C., & Lima, A. F. M. D. Smoking influences on the thickness of marginal gingival epithelium. Pesquisa Odontológica Brasileira, 2003: 17(1): 41-45. Collins, F. M. Tobacco Cessation and the Impact of Tobacco Use on Oral Health. RDH. Oklahoma: Penwell Dental Group 2010. Parmar, G., Sangwan, P., Vashi, P., Kulkarni, P., & Kumar, S. Effect of chewing a mixture of areca nut and tobacco on periodontal tissues and oral hygiene status. Journal of oral science 2008: 50(1): 57-62. Minaya-Sánchez, M., Medina-Solís, C. E., Maupomé, G., Vallejos-Sánchez, A. A., Casanova-Rosado, J. F., & Marquez-Corona, M. D. L. Prevalence of and risk indicators for chronic periodontitis in males from Campeche, Mexico. Revista de SaludPública 2007: 9(3): 388-398. 23. Lung, Z. H. S., Kelleher, M. G. D., Porter, R. W. J., Gonzalez, J., & Lung, R. F. H. Poor patient awareness of the relationship between smoking and periodontal diseases. British dental journal 2005: 199(11): 731-737. 24. Vered, Y., & Sgan-Cohen, H. D. Self–perceived and clinically diagnosed dental and periodontal health status among young adults and their implications for epidemiological surveys. BMC Oral Health 2003: 3(1): 3. 25. Fotedar, S., Sogi, G. M., Fotedar, V., Bhushan, B., Singh, B., Dahiya, P., & Thakur, A. S. Knowledge of, Attitude Towards, and Prevalence of Tobacco Use Among Dental Students in Himachal Pradesh State, India. Oral health and dental management 2013: 12(2): 73-79. 26. Preshaw PM, HeasmanL, Stacey F, Steen N, McCracken GI,Heasman PA J ClinPeriodontaol, 2005;32(8):869-79. __________________________________________________ International Dental Journal of Student’s Research, April - June 2015;3(2):72-78 {79} SHORT COMMUNICATION A review of the use of laser in periodontal therapy Dr. Amir Manzoor Shah1, Dr. Khurram Khan2, Dr. Fahd Ahmed3, Dr. Nida Amir4 1, BDS, MSc, M-Dent Perio Resident, 2BDS, MSc, BDS, MSc, Faculty of Dentistry, Division of Oral Health and Society, McGill University, Montreal Quebec, Canada. 1 Division of Periodontics, Dental Diagnostics and Surgical Sciences, Faculty of Dentistry University of Manitoba, Winnipeg Manitoba, Canada 4 BDS, MSc, MSc-Peads-Dent Resident, Department of Pediatric Dentistry, Nobel Biocare Oral Health Centre, University of British Columbia, Vancouver BC, Canada. 3 Corresponding Author: Dr. Amir Manzoor Shah E-Mail: [email protected] Access this Article Online www.idjsr.com Use the QR Code scanner to access this article online in our database Article Code: IDJSR SE 0168 Quick Response Code Abstract The use of lasers has significantly developed in modern dentistry, however the clinical value and awareness on the benefits of their use is limited. Lasers have made their way in dental treatment since 1994. They have been granted Food and Drug Administration (FDA) approval, however the American Dental Association (ADA) is still researching on the outcomes of laser therapy1. This article reviews the use of laser therapy in the initial non-surgical phase of periodontal therapy. field of dentistry with applications in various aspects of clinical practice. The field of restorative dentistry has utilized lasers, for the removal of tooth decay, and this is where laser therapy evolved from in dentistry. Lasers have been used for the removal of the carious lesion and preparation of the tooth for a restoration. Many restorative materials incorporate laser in their setting/curing system. Lasers help in controlling the spread of pathological and non-pathological lesions as well as acquiring tissue for biopsies. Lastly, tooth whitening has become a quick and easy procedure for patients and practitioners with the use of laser technology. More recently, mechanical debridement of subgingival root surfaces and periodontal pockets are being treated with lasers as an adjunct or substitute4. Laser therapy has greatly advanced in recent years, especially in restorative dentistry, however general dentists are not aware of the many benefits they can receive with the help of this developing therapeutic tool in periodontology. How Lasers Work There are various types of lasers in the market and each device emits energy at a specific wavelength. For example, diode (gallium: arsenide) lasers emit wavelengths ranging from 635-950 nm. Carbon dioxide (CO2) lasers emit wavelengths at 10 600 nm4. Secondly, radiation is delivered in different forms. The radiations can be continuous, pulsed or running pulse waveforms. The photon emitted through the collimator for a unidirectional, monochromatic light being emitted in coherence. Thirdly, the laser beam is infrared and invisible; therefore light is incorporated into the device to act as an aiming beam. Upon exposure, the targeted tissue will absorb, reflect or scatter the laser beam5. Two variables control this affect; these are the wavelength and the properties of the target tissue. Biologic tissues primarily absorb the beam, while scattering only occurs in deep tissue penetration5, 6. Applications of lasers in dental procedures: Introduction The term “LASER” stands for “light amplification by stimulating emission of radiation”2. Lasers were introduced in the field of medicine approximately 50 years ago3. This technology trickled its way to the Restorative Dentistry: Erbium-doped yttrium aluminum garnet (Er: YAG) Lasers are useful in the detection/removal of caries with maximal preservation of healthy mineralized enamel and the cavity preparation for placement of filling materials. Lasers have also been beneficial in the curing of International Dental Journal of Student’s Research, April - June 2015;3(2):79-82 {80} filling materials such composite resins7,8. asphotopolymerizationin Oral and Maxillofacial Surgery (OMFS): Lasers in OMFS are generally used for excisional/ incisional, biopsy, ablation and hemostasis. Lasers are used for biopsy in removing tissue for examination/inspection of potential neoplastic tissue. Ablation is a process in which superficial tissue is removed and unnecessary tissue removal could be avoided9.Hemostasis is the control of bleeding at the site if surgery. Endodontics: Laser fibers and endodontic tips carry out the following procedures: diagnosis, pulpotomy, cleaning and obturation of the root canal system, retreatment and apical surgeries. The laser Doppler flowmetry is an advancement in endodontics as it analyzes the blood flow in the canal system. This evaluation was considered as one of the most accurate methods in testing pulp vitality10. Periodontal Disease: Periodontitis is a polymicrobial infection caused by multiple types of bacteria, harmfully interacting with the body’s immune system. There are various types of bacteria that accumulate in plaque (biofilm), which is found lining the gums of the oral cavity. Inadequate oral hygiene causes migration of these bacteria along the root surface to form a hardened calcified substance known as calculus (tartar). In response to this invasion of bacteria, gum tissues become inflamed and ulcerated. Further neglection may cause periodontal tissue damage with regression of bone around the tooth, ultimately leading to tooth loss11. Lasers in periodontal therapy: Arresting the disease process is the primary goal of periodontal treatment12. Initial therapy is the first step to reestablish a healthy oral cavity. This is the process of cleaning and disinfecting the affected root surfaces. Once infection and inflammation in the oral cavity are under control, secondary goals are outlined to help in regeneration of healthy periodontal attachment to the tooth surface, which may include surgical treatment12, 13. Treatment techniques for initial therapy: Phase 1 of periodontal therapy includes mechanical debridement of the biofilm, which requires removal of bacteria and calculus from the root surfaces of affected teeth. This process is known as “scaling and root planing” or “root debridement”14. Hand instruments and/or ultrasonic (high frequency) instruments are traditionally used to carry out the procedure and require a high level of skill and tactile sensitivity. An alternative option is non-surgical therapy but this technique is sensitive and also time consuming. An invasive procedure carried out in the first phase of periodontal therapy known as curettage, where the lining of an inflamed pocket was removed from the tooth or tissue has recently seen advancements in research. Current literature suggests if bacteria are affectively removed, tissue repair can occur without the need for surgical intervention12, 14. Treatment with lasers: An adjunct for non-surgical debridement of bacteria from root surfaces can now be conducted with the use of lasers. Neodymium: Yttrium-Aluminium-Garnet (Nd: YAG) laser is one example used in the treatment of periodontitis providing the ability to carry out sub gingival curettage, removal of sub gingival plaque and calculus from infected root surfaces15. Periodontal tissues have varying water and mineral content, pigment and tissue density, which allows them to absorb beams from Nd: YAGand diode lasers16. On the other hand, CO2 lasers are better suited for soft tissue procedures because its energy beam is absorbed mostly by water. Hydroxyapatite is better suited for other types of lasers17. Other factors that affect absorption of energy beam into the target include power, pulse duration, duration of exposure, angle of energy delivery and waveform (pulsed or continuous). When choosing the type of laser, a specific goal should be made in order to achieve the desired results. This is because, energy absorption will cause the target to warm up, coagulate, vaporize or melt and recrystallize as seen in hard tissues. Periodontal Disease and Lasers techniques: The American Academy of Periodontology (AAP) has suggested that using lasers during scaling and root planing (SRP) may provide improvements in procedures by decreasing bleeding, swelling and discomfort during surgery. Laser Assisted New Attachment Procedure (LANAP) is a relatively new treatment option that helps remove plaque and calculus, while limiting bacterial infection to help fight periodontitis by regenerating rather than resecting tissues. LANAP helps to remove infectioncausing bacteria in a safe and painless procedure that promotes epithelial and periodontal fiber attachments in the affected area18. Other uses of this technique include removal of caries and preparation of teeth for restorations or crowns. Caution must be taken when using laser therapy because of varying power levels and wavelengths. Incorrect wavelength and/or power levels can result in damage during periodontal treatment causing more harm than good. Benefits of Laser Treatment International Dental Journal of Student’s Research, April - June 2015;3(2):79-82 {81} Modern techniques using lasers can control the spread of harmful bacteria and limit tooth loss compared to standard periodontal treatment options. Some benefits of laser treatment for gum disease include: elimination of cutting and bleeding, soreness and discomfort of the gums. Isolation of deep periodontal pockets. Reduction in tooth loss. Regeneration of bone and ligament tissues. Lastly, increased chances of success with a solution in case of setbacks that may occur19. As compared to a dental hand piece, lasers are advantageous in certain conditions causing less pain, anxiety and discomfort for the patient. In addition, soft tissue damage is minimized and the need for anesthesia may be avoided in less invasive procedures. benefit of a less invasive treatment option coupled with shorter treatment duration and discomfort for the patient are attractive features of laser therapy but studies have yet to prove its effectiveness. 10-15% of the population that suffer from periodontal disease is treated with long-term daily oral hygiene instructions. Professional monitoring and evaluation with regular periodontal maintenance every 3 months is the desired level of care with no short cuts in the foreseeable future. References 1. 2. Conclusion With all the benefits of laser therapy outlined in this review, we point out the lack of studies supporting its use alone. There is no evidence in the literature to suggest it may control adult chronic periodontitis without conventional SRP and surgical treatment. The advantage of laser therapy in conjunction with traditional therapy are of benefit, yet current challenges include increased operating costs for the dentists and patients coupled with technique sensitivity for operators. The main advantages of laser therapy over conventional methods are reduced tissue inflammation and bleeding. Sterilization of the affected area leading to a reduction in post-treatment discomfort with higher patient satisfaction. With this in mind, are lasers considered more advantageous than traditional therapy? Current literature is inconclusive. In order to come up with a final conclusion, evidence-based science provides strict research protocols and parameters to make fair comparisons between various treatments. Studies should have an adequate sample size, be randomized with controls and have specific treatment goals and criteria. To show effectiveness and long-term results, an appropriate time-line should be set. In a systematic review of the literature on the use of lasers in periodontal therapy, only 8 of 300 studies met the criteria above. Researchers in only 5 out of 8 studies assessed the tissue attachment after treatment, a gold standard in assessing periodontal treatment outcome. The results of the review could not point to any advantages of Nd:YAG lasers over conventional periodontal therapy in the treatment of initial periodontitis. Initial non-surgical therapy of periodontitis remains the treatment of choice with growing interest in lasers as an adjunct treatment option for gum disease. The 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. 14. 15. 16. 17. ADA.org, ‘Statement on Lasers in Dentistry’ 2009. http://www.ada.org/en/about-the-ada/ada-positionspolicies-and-statements/statement-on-lasers-in-dentistry N.p.2015 . Gould, R. Gordon (1959). "The LASER, Light Amplification by Stimulated Emission of Radiation". In Franken, P.A. and Sands, R.H. (Eds.). The Ann Arbor Conference on Optical Pumping, the University of Michigan, 15 June through 18 June 1959. p. 128. Maiman TH. Stimulated optical radiation in ruby. Nature. 1960;187:493-4. Matthews, D. C. (2009). Seeing the Light--the truth about soft tissue lasers and nonsurgical periodontal therapy. Journal (Canadian Dental Association), 76, a30-a30. Cobb CM. Lasers in periodontics: a review of the literature. J Periodontol. 2006;77(4):545-64. Dederich DN, Bushick RD; ADA Council on Scientific Affairs and Division of Science; Journal of the American Dental Association. Lasers in dentistry: separating science from hype. J Am Dent Assoc. 2004;135(2):20412. Erratum: J Am Dent Assoc. 2004;135(6):726-7. PM Freitas, RS Navarro, JA Barros, C de Paula Eduardo The use of Er:YAG laser for cavity preparation: an SEM evaluation Microsc Res Tech, 70 (9) (2007), pp. 803–808. U Keller, R Hibst Effects of Er:YAG laser in caries treatment: a clinical pilot study Lasers Surg Med, 20 (1997), p. 32. Bornstein, M. M., Winzap-Kälin, C., Cochran, D. L., &Buser, D. (2005). The CO2 laser for excisional biopsies of oral lesions: a case series study. The International journal of periodontics & restorative dentistry, 25(3), 221-229. Y Kimura, P Wilder-Smith, K Matsumoto Lasers in endodontics: a review IntEndod J, 33 (2000), pp. 173– 185. Pihlstrom, B. L., Michalowicz, B. S., & Johnson, N. W. (2005). Periodontal diseases. The Lancet, 366(9499), 1809-1820. PERIODONTICS, S. O. (2001). Guidelines for periodontal therapy. J Periodontol. Nyman, S., Lindhe, J., Karring, T., &Rylander, H. (1982). New attachment following surgical treatment of human periodontal disease. Journal of clinical periodontology, 9(4), 290-296. Van Dyke, T. E. (2008). The management of inflammation in periodontal disease. Journal of periodontology, 79(8S), 1601-1608. Cobb, C. M. (2006). Lasers in periodontics: a review of the literature. Journal of periodontology, 77(4), 545-564. Folwaczny, M; Aggstaller, H; Mehl, A; Hickel, R: Removal of bacterial endotoxin from root surface with Er: YAG laser. Am J Dent 16(1):3–5, 2003. Crespi, R., Barone, A., Covani, U., Ciaglia, R. N., &Romanos, G. E. (2002). Effects of CO2 laser treatment International Dental Journal of Student’s Research, April - June 2015;3(2):79-82 {82} on fibroblast attachment to root surfaces. A scanning electron microscopy analysis. Journal of periodontology, 73(11), 1308-1312. 18. Yukna, R. A., Carr, R. L., & Evans, G. H. (2007). Histologic evaluation of an Nd: YAG laser-assisted new attachment procedure in humans. The International journal of periodontics & restorative dentistry, 27(6), 577. 19. White, J. M., Goodis, H. E., & Rose, C. L. (1991). Use of the pulsed Nd: YAG laser for intraoral soft tissue surgery. Lasers in surgery and medicine, 11(5), 455-461. __________________________________________________ International Dental Journal of Student’s Research, April - June 2015;3(2):79-82 {83} ORIGINAL RESEARCH Prevalence of bleeding gums while tooth brushing among betel nut chewers vs non betel nut chewers in school going children Dr. Syed Misbahdduin1, Dr. MansoorUl Aziz2, Dr. Asma Fazal3, Dr. Tayyaba Khairuddin4, Dr. Safia Khairuddin5 University College Cork, Cork, Ireland Associate Professor at Altamash Institute of Dental Sciences 3 Our Lady’s Children Hospital Crumlin, Dublin, Ireland 4 Registrar Psychiatrist at St. Joseph Intelectual Disability, St. Itas Mental Health Services, Donabate, Portrane, Dublin, Ireland 5 Department of Proteomics and HIV Research Lab, Post-doc University of Southern California, USA 1 2 Corresponding Author: Dr. Syed Misbahuddin Email: [email protected] Access this Article Online www.idjsr.com Use the QR Code scanner to access this article online in our database Article Code: IDJSR SE 0169 Quick Response Code Abstract Betel nut (BN) also referred to as Chalia/Supari has been used for thousands of years. BN chewing is an important and popular cultural habitin India, Bangladesh and Pakistan (the subcontinent).It is being used regularly on individual and family basis. The use of BN is prevailing in the rural and urban areas of Pakistan. In several studies, an association between BN chewing and oral health problems like bleeding gums, sore gums have been identified. These lesions are reported in children and adolescents. This is of great concern not only because of the high cost involved in their management but the morbidity and mortality associated with it. Low cost, easy availability, advertising, role modelling, social acceptance and perception of BN as harmless, contribute to its use. The aim of this cross-sectional study in Central District of Karachi (CDK) was to assess the prevalence of oral soft tissue lesions and to investigate the associations which may exist between oral conditions and BN chewing among the young school going children. Three hundred and sixty students from 17 different schools participated in the study. The mean age was 13.86 ±1.2 years with the age range of 12 to 16 years. Out of these 360 students, 175 were females and 185 were males. The results showed a high prevalence of bleeding gums while tooth brushing among BN chewers (BNC) compared to non-chewers (NBNC) (19% and 3% respectively). The high prevalence of BN chewing (59% of the low socio-economic young population studied) should be addressed at local and government level through support for effective preventive programs and health promotion campaigns. Promotion of oral health and eradication of BN chewing are important goals for the prevention of oral cancer among this population. Introduction The chewing of betel nut (BN) is an old practice in South-East Asia, especially in the Indian subcontinent1. This tradition is inherited by generation after generation and has become a popular cultural activity among people of Pakistan, India, Sri Lanka, and Bangladesh2. BN is a fruit of areca tree that widely grows in tropical Pacific, Asia and east Africa3. It is a small feathery plant that grows to the height of 1.5 m. The most common method of using BN is to chop it into very small pieces with the help of an especial instrument known in local language Urdu as “sarota”. Slurry of slaked lime and catechu boiled in water is applied on a betel leaf and the chopped pieces of BN are rolled in it to be kept in mouth4,5. BN contains the alkaloid arecoline in addition to nitrosamines, which is carcinogenic.Various studies have been conducted to determine the relation of BN and other alternative chewing material to oral and other associated cancers6,7. It has been proved that BN, Gutka and Paan cause oral cancers8,9and alone in India, out of 700,000 cancers diagnosed each year International Dental Journal of Student’s Research, April - June 2015;3(2):83-87 {84} 300,000 cases are related to tobacco smoking and BN chewing10,11. Although, the use of BN and Gutka is associated with certain oral conditions; the prevalence and effect on oral health of school going children of 12-16 years of age is not clearly known in local context. This study was conducted to identify the prevalence of oral lesions and to investigate association which may exist between the oral conditions and BN/Gutka chewing among the school going children. Objectives of the study To identify oral lesions present in school going children and to compare the prevalence of oral lesions among those who chew betel nut versus nonchewers. Methodology A cross sectional study was conducted in City District, Karachi. A research questionnaire was prepared, and sent along with the parental consent and student assent forms to the Clinical Research Ethics Committee of the Cork Teaching Hospital, University College Cork for ethical approval, which approved it. Ethical approval was also granted from Baqai Dental College, Karachi for this study. The questionnaire was scrutinized by the subject specialists and was coded for statistical purposes. The research questionnaire consisted of chewing habits, clinical interview and findings of the clinical examination. The sample comprised of 360 students from 17 different schools in CD, Karachi. The age range of sample population was 12 - 16 years. The authors was trained and calibrated for the examination of oral health and identification of oral lesions at Cork University School and Dental Hospital. In Karachi, the author hired qualified dental assistants and trained and calibrated them. The subjects had their oral examination done on the specified date by the author and the trained dental staff. Individuals were examined in the natural day light on a chair in a separate room to maintain privacy. A hand torch was also used in some cases, where natural daylight was insufficient. A sterile CPITN probe was used to observe bleeding on gentle probing. When required the teeth were dried using cotton wool rolls. Universal precautions were followed. Personal protective clothing and equipment was worn by all the examiners and recorders in attendance. Latex free examination gloves were used for the examination of each child and were changed before examining the next child. A facemask was worn and changed at frequent intervals. A disposable paper sheet was used under each set of instruments and disposed after each examination. The CPITN probes and mirrors were placed in a container used solely for the transport of “contaminated” instruments. All re-usable instruments were washed and autoclaved at the end of each session. All contaminated waste, which included gloves, facemasks, cotton wool rolls, tissues and wipes, were disposed into ‘hazardous waste’ yellow bags in accordance with infection control best practice. Two plastic boxes for instruments were used, of which; one box was for transporting sterile instruments only while the other box was for contaminated instruments. All children were given protective eye covers to wear. These were cleaned with disinfectant wipes between examinations. The torches were wiped with a disinfectant between examinations. Each child was asked to sit on the chair. Face masks and tinted protective eyewear were used for all the children. First, a visual examination was done to check for dental caries. Teeth were examined wet, and a CPITN probe was used to remove food debris as well as to confirm dental cavitations. Soft tissues and oral cavity were then examined for any abnormalities. Statistical Analyses SPSS Version 18® was used for statistical analysis. The following information was obtained and analysed: The number of children examined by gender and age. The mean age in years of children that participated in the study. Habit of BN chewing and its association with gender. Frequency of tooth brushing habit. Distribution of subjects by gender and tooth brushing habit Association of gender with the frequency of bleeding gums while brushing teeth Results The distribution of males and females by the status of betel nut chewing: As illustrated in the table below, 61.61% of males and 38.39% of females chewed betel nut (Table 1.1). Table 1.1 BN chewing and gender distribution Betel nut Males% (n) Females% (n) chewing Yes 61.61 (130) 38.39 (81) No 36.91 (55) 63.08 (94) The value of chi square is 21.32, df =1 and p= 0.000. There was a significant difference in the habit of betel nut chewing among males and females. Almost International Dental Journal of Student’s Research, April - June 2015;3(2):83-87 {85} twice the numbers of males (61.61%) chewed betel nut as compared to females (38.39%). Never Twice a day More 60 50 40 30 20 10 0 53.71% 38.37% 37.83% 29.16% 12 years 13 years 14 years 15 years 16 years Male 37 39 49 34 26 Female 32 44 40 34 25 Figure 1.1 Gender and age of the subject Tooth brushing habit: Frequency of tooth brushing was ascertained from question 1 of the oral hygiene and chewing habits questionnaire (Appendix VI). It was noted that 15.85% subjects never brushed their teeth whereas45.83% brushed once a day, 33.61% twice a day and 4.71% more than that (Graph 1.2). 16.75% Males BN chewing % 53.69% 42.65% 40.28% 15.85% 4.71% Females The association of tooth brushing with betel nut chewing: When the tooth brushing habit of both the groups was compared, it was noted that among BNC, 11.86% never brushed their teeth, 40.28% brushed once a day, 42.65% twice a day and 5.21% brushed more than twice a day. In comparison, among NonBNC, 21.47% never brushed their teeth, 53.69% brushed once a day, 20.80% twice a day and 4.04% brushed more than that (Graph 1.4). 33.61% Twice a day 2.28% Figure 1.3: The distribution of subjects by gender and tooth brushing habit 45.83% Once a day 14.85% 7.05% Brushing frequency Never Once a day 21.47% 20.80% 11.86% 5.21% 4.04% More Figure 1.2: Frequency of tooth brushing Tooth brushing habit among males and females: The results showed that 16.75% males never brushed their teeth, 38.37% brushed once a day, 37.83% twice a day and 7.05% more than twice a day. Among females, 14.85% never brushed their teeth, 53.71% brushed once a day, 29.16% twice a day and 2.28% more than that (Graph 1.3). Never Once a day Twice a day More Figure 1.4: Association of tooth brushing with BNC Chi square value is 21.29, df= 1 and p= 0.001. There was a significant difference in the frequency of tooth brushing among BNC and NonBNC. Eighty eight point one four percent of BNC were brushing teeth as compared to 78.53% of NonBNC. International Dental Journal of Student’s Research, April - June 2015;3(2):83-87 {86} The association of gender with the frequency of bleeding gums while brushing teeth: It was noted that 67.02% of males and 81.14% of females never had bleeding gums while brushing their teeth. Eighteen point three nine percent of males and 14.86% of females occasionally had bleeding, while 14.59% of males and 4% of females frequently had bleeding from their gums while brushing teeth (Graph 1.5) Frequency of bleeding gums Male 67.02% Female 81.14% 14.59% 18.39% 14.86% Never Occasional 4% Frequently Figure 1.5: Association of gender with the frequency of bleeding gums while brushing teeth The value of chi square is 13.78, df= 2 and p=0.001. There was a significant difference in complaints of bleeding gums while brushing teeth between the genders with females having gums that bleed less often. The association of betel nut chewing with the frequency of bleeding gums while brushing teeth: Among BNC, 18.95% occasionally had bleeding gums and 10.44% frequently had bleeding from gums while brushing their teeth. However, among NonBNC; 13.43% occasionally had and 8.05% frequently had bleeding from gums while brushing their teeth (Table 1.2). Table 1.2: The association of bleeding gums while brushing teeth and betel nut chewing Frequency of BNC Non BNC bleeding gums while % (n) % (n) brushing teeth Never 70.61 78.52 (149) (117) Occasionally 18.95 (40) 13.43 (20) Frequently 10.44 (22) 8.05 (12) Total 100 (211) 100 (149) The value of chi square is 2.865, df= 2 and p= 0.239. There was no significant difference in gum bleeding while brushing teeth between BNC and NonBNC. The association of bleeding gums without brushing teeth with betel nut chewing: It was noted that 5.69% BNChad bleeding gums without brushing teeth. On the other hand, 0.68% NonBNC had bleeding gums without brushing theirteeth (Table 1.3). Table 1.3 The association of bleeding gums without brushing teeth and betel nut chewing Bleeding gums BNC NonBNC without brushing % (n) % (n) teeth Yes 5.69 (12) 0.68 (1) No 94.31 (199) 99.32 (148) Total 100 (211) 100 (149) The value of chi square is 6.313, df= 1 and p= 0.012. There was a significant difference among both the groups with more BNC having bleeding gums without brushing teeth as compared to NonBNC. Discussion In our study, we found out that twice a number of males were indulged in Betel nut chewing as compared to females. Females had the highest percentage of brushing teeth but males predominated in brushing teeth twice a day. It was also noticed that tooth brushing was more common among BNC as compared to Non BNC. In contrast to males, bleeding gums was less common in females. It is seen that mostly periodontal diseases progress un-noticed. People commonly recognize it at an advance stage. It is therefore important to make dental health education mandatory for control and maintenance of periodontal health. Early recognition of periodontal conditions is not so common because people do not understand the connection between gum bleeding and gum disease. According to Brady, 73% of patients with periodontal disease did not know that they had it12. Almas et al reported 42% of Saudis with bleeding gums13. Khawamura and Eva Motto found that 3 quarters of Japanese employees had bleeding gums14. Gingivitis with bleeding gums is the first symptom of periodontal disease. This symptom of disease is selfdetected and is the most reliable indicator of the condition15. In order to prevent progression of periodontal disease, the public needs dental health education to connect gingival bleeding with gum disease16. The aspect of dental health education therefore has a key role in the awareness of periodontal disease among different groups of society. International Dental Journal of Student’s Research, April - June 2015;3(2):83-87 {87} Self-Reporting Research conducted in Dundee dental hospital and school revealed astonishing results about the knowledge and understanding of periodontal disease among the people who took part in the study. People were unable to recognize that they have periodontal disease while having gingival bleeding17. Although people were aware of bleeding gums at times, there were other times when it went unnoticed A.D. Gilbert & N.M. Muttall. Radiographs were not used in order to validate the self-reporting instrument and also to avoid unnecessary radiation. The CPITN method was normally used to determine periodontal treatment needs and was considered more appropriate to make patient realize that they have gum disease. Conclusion 11. 12. 13. 14. 15. 16. The results of this study indicate dental and oral care education must be included in a national program that promotes preventive oral care in schools as well in other oral health educational programs aimed at the general public. This recommendation is based on the finding that children were aware of the importance of dental care but need proper education and guidance. 17. Summers RM, Williams SA, Curzon ME. The use of tobacco and betel quid ('pan') among Bangladeshi women in West Yorkshire. Community Dent Health. 1994;11(1):12-6. Brady WF. Periodontal disease awareness. The Journal of the American Dental Association. 1984;109(5):70610. Taani DQ. Periodontal awareness and knowledge, and pattern of dental attendance among adults in Jordan. International dental journal. 2002;52(2):94-8. Okada M, Kawamura M, Kaihara Y, Matsuzaki Y, Kuwahara S, Ishidori H, et al. Influence of parents’ oral health behaviour on oral health status of their school children: an exploratory study employing a causal modelling technique. International Journal of Paediatric Dentistry. 2002;12(2):101-8. Walsh MM. Effects of school‐based dental health education on knowledge, attitudes and behavior of adolescents in San Francisco. Community dentistry and oral epidemiology. 1985;13(3):143-7. Murtomaa H, Meurman J, Rytömaa I, Turtola L. Periodontal status in university students. Journal of clinical periodontology. 1987;14(8):462-5 Gilbert A, Nuttall N. periodontology: Self-reporting of periodontal health status. British dental journal. 1999;186(5):241-4. ___________________________________________ References 1. 2. 3. 4. 5. 6. 7. 8. 9. 10. Rooney DF. Betel nut chewing in south East Asia.http://rooneyarchive.net/lectures/lec_betel_chewi ng_in_south-east_asia.htm Gupta PC, Ray CS. Epidemiology of betel quid usage. Ann Acad Med Singapore, 2004;33(suupl):31s-36s en.wikipedia.org/wiki/Areca Patel K. Helping members of the South Asian community quit smokeless tobacco. http://www.nice.org.uk/newsroom/features/HelpingMe mbersOfTheSouthAsianCommunityQuitSmokelessTob acco.jsp Nair U, Bartsch H, Nair J. Alert for an epidemic of oral cancer due to use of the betel quid substitutes gutkha and pan masala: a review of agents and causative mechanisms. Mutagenesis. 2004;19(4):251-62. Yang YH, Lien YC, Ho PS, Chen CH, Chang JS, Cheng TC, Shieh TY. The effects of chewing areca/betel quid with and without cigarette smoking on oral submucous fibrosis and oral mucosal lesions. Oral Dis. 2005;11 (2): 88-94. Trivedy CR, Craig G, Warnakulasuriya S. The oral health consequences of chewing areca nut. Addict Biol. 2002;7(1):115-25. Thomas and MacLennan (1992). "Slaked lime and betel nut cancer in Papua New Guinea". The Lancet Oncology 340 (8819): 577–578. Shwu-Fei LC, Chiu-Lan C, Liang-Yi H, Pi-Chen H, Jung-H C, Chang-Ming S, Chin-Wen C, and TsungYun L. Role of oxidative DNA damage in hydroxychavicol-induced genotoxicity. Mutagenesis vol.11 no.5 pp.519-523, 1996 SangeetaDarvekar Charitable Trust [email protected] International Dental Journal of Student’s Research, April - June 2015;3(2):83-87 {88} SHORT COMMUNICATION Short communication: Stem Cells for Periodontal Tissue Regeneration Dr. Mohamed-Nur Abdallah1, Dr. Mai S. Ali2 1 Faculty of Dentistry, McGill University, Montreal, Quebec, Canada. 2 Department of Restorative Dentistry and Endodontics King Hussein Medical Center, Amman, Jordan. Corresponding Author: Dr. Mohamed-Nur Abdallah Email: [email protected] Access this Article Online www.idjsr.com Use the QR Code scanner to access this article online in our database Article Code: IDJSR SE 0170 Quick Response Code Introduction Periodontal disease is an inflammatory condition that causes pathological alterations in the periodontium, potentially leading to tooth loss [1]. In the world, 35% of adults in the population suffer from moderate periodontal disease, while up to 15 % were affected by a more severe form at some stage of their life [2, 3].The periodontium has always proved to be one of the structures with inherent regenerative capacities. It gives rise to osteoblasts, periodontal ligament (PDL) fibroblasts, and cementoblasts[4]. However, the periodontium – which includes the periodontal ligament, root cementum, alveolar bone and gingiva has a limited ability to regenerate once damaged [4]. For decades, periodontists have sought to repair the damage from periodontitis and to achieve regeneration through a variety of non-surgical procedures and surgical procedures that include root surface conditioning, bone graft placement, guided tissue regeneration and the application of growth factors [5-7]. However, current procedures allow the periodontal tissue to be repaired rather than regenerated with some approaches showing some limited unpredictable regenerative outcome [8-12]. Recent advances in tissue engineering and stem cell biology have paved the way to develop novel approaches in the regenerative periodontal therapy or to supplement existing treatment modalities for periodontal disease. Cell-based Tissue Engineering In order to achieve a successful periodontal regeneration, healing events must occur in an ordered and programmed order, starting with the migration of appropriate progenitor cells into the wound site and attach to the denuded root surface [13]. These progenitor cells have the ability to proliferate and mature into the different component of the functional periodontium. The success of these progenitor cells is also dependant on the availability of the appropriate growth factors and contact with the extracellular matrix [14, 15]. This allows the formation of a functional epithelial seal with an insertion of a new connective tissue fibers into the root in conjunction with a new acellular cementum covering the root surface and the reestablishment of a proper alveolar bone height that can ensure a healthy regenerated periodontium [16, 17]. Current tissue engineering trends have utilized the principles of periodontal healing and cell biology in developing cellular-based techniques for periodontal regeneration. In the context of periodontal therapy, cell-based tissue engineering involved incorporation of progenitor cells in a prefabricated three-dimensional biomaterial that is implanted into the defect site. This approach overcomes some of the limitations of conventional procedures and allows direct application of progenitor cells and growth factors in the defect area [18]. The success of the cell-based approach requires the following essential factors [19]: (1) appropriate progenitors cells that proliferate and mature to tissue-forming phenotypes, including fibroblasts, osteoblasts, and cement oblasts; (2) appropriate signals to control cellular differentiation and tissue formation; (3) a three dimensional scaffold to support the cells and facilitate the previous processes; and (4) good blood supply and promotion of new vascular networks. Consequently, the most critical factors in tissue engineering is the choice of optimal stem cell population that give rise to the progenitor cells and the selection of an appropriate biomaterial that can serve as a scaffold that both support these cells and possess adequate mechanical properties. International Dental Journal of Student’s Research, April - June 2015;3(2):88-92 {89} Stem cells By definition, a stem cell is a clonogenic, relatively undifferentiated cell that is capable of self-renewal and multi-lineage differentiation depending on intrinsic signals modulated by extrinsic factors in the stem cell niche[20, 21]. Stem cells (SCs) are classified according to their origin and their differentiation potential[16, 22]. They can be broadly classified into three categories: (1) embryonic stem cells (ESCs); (2) induced pluripotent stem cells (iPSCs); (3) adult or postnatal SCs. ESCs are totipotent or pluripotent cell, which means that they possess the capacity to proliferate extensively and differentiate into almost all possible cell types. However, using embryos to obtain human embryonic stem cells has raised many ethical concerns and limited their usage. These concerns pushed researchers to investigate the possibility of genetically reprogramming somatic cells back to the pluripotent phase, which lead to the generation of iPSCs[23, 24]. They are comparable to the ESCs in their function, morphology, gene expression and wide differentiation capacity[23, 24]. Efforts were also made to obtain iPSCs from gingival and periodontal fibroblasts[25-27]. In general, the behaviour of iPSCs can be unpredictable because the genetic manipulations may alter their development and growth characteristics, thus, limiting their in tissue engineering[16]. Moreover, both the ESCs and the iPSCs convey tumorigenic properties that raise serious safety concerns that further limit their use in regenerative therapies. Somatic adult or postnatal stem cells can be derived from the majority of fetal and adult tissues that continually replenish themselves (e.g. dermis, peripheral blood) [20, 28, 29]. They are multipotent and can differentiate into limited number of cell lineages such as endothelial, perivascular, neural, bone or muscle cells [30]. It is thought that they function in long-term tissue maintenance and replacing cells that are either lost or injured [31]. Despite their limited life span, SCs have extensive self-renewal capacity [32]. Even though adult SCs exhibit more restricted capabilities compares to ESCs, they are immunocompatible and are not associated with any ethical concerns (Han). Their most common sources are the bone marrow (hematopoietic stem cells) and bone marrow stromal cells (mesenchymal stromal stem cells) [33-35]. Hematopoietic stem cells were the first cells to be used in regenerative therapies, mainly in the treatment of blood malignancies and immunodeficiency syndromes, but they lack the ability to give rise to supporting connective tissues [36]. On the contrary, mesenchymal stem cells (MSCs) have been used to treat a range of musculoskeletal abnormalities and are capable of differentiate to connective tissue cells [36]. Consequently, MSCs serve as potential candidates for periodontal regeneration. Human mesenchymal stem cells in periodontal regeneration Human mesenchymal stems cells (hMSCs) are the most commonly used stem used in periodontal regeneration due to their extensive expansion rate, immunocompatibility (some may exhibit immunosuppressive properties), and extensive availability without any ethical restraints[37]. hMSCs used for periodontal regeneration can be obtained from both extraoral and intraoral tissues. Extraoral sources include bone marrow and adipose tissues. Past in vivo studies showed that implanted bone marrow-derived MSCs can induce the regeneration of cementum and alveolar bone up to 20% in experimental Class III defects [38], and can differentiate into cementoblasts, alveolar bone osteoblasts and PDL fibroblasts [38-41]. Small human clinical trials demonstrated that mixing bone marrow-derived MSCs with platelet-rich plasma (PRP) result in reduction in intrabony defect depth and resolution of bleeding, while mixing these cells with atelocollagen reduced the probing depth by 4mm [41]. Another potential extraoral source is adipose tissue. In vivo studies in rats demonstrated that adipose-derived stromal cells mixed with PRP promoted periodontal regeneration [42]. Future clinical studies are required to assess the candidacy of adipose-derived MSCs for periodontal regeneration, since they are more abundant, more easily accessible to lipoaspirates with lower morbidity compared to the more invasive procedures to obtain the bone marrowderived MSCs. Dental-tissue derived MSCs can be obtained intraorally from dental tissues (e.g. dentin or PDL or pulp) either from permanent or deciduous teeth [4345]. They can be simply obtained using a chair side procedure or immediately after tooth extraction [43]. However, these cells have a more restricted cellular potency in comparison to bone marrow-derived MSCs, as bone tissues are known to undergo more remodelling than dental tissues [16]. There has a been a particular interest in assessing the regenerative capacity of mesenchymal stems derived from PDL (PDLSCs), since being first isolated in 2004[46].They showed an increased proliferative potential of cell populations that can differentiate into either cementoblasts, osteoblasts, fibroblasts and even adipocytes[47]. PDLSCs can exert different benefits on multiple levels including tissue regeneration, neovascularization and immunomodulation, by which PDLSCs can inhibit International Dental Journal of Student’s Research, April - June 2015;3(2):88-92 {90} the immune response without an induction of an inflammatory response, thus enabling the use of allogenic PDL sources for periodontal regeneration[48, 49]. Animal studies showed that implanted PDLSCs in periodontal defects resulted in the new formation of cementum and alveolar bone, and new attachment apparatus[50-55]. Furthermore, a study conducted on mini-pegs demonstrated the formation of PDL- like tissue mimicking the orientation of the fibre bundles similar to Sharpey’s fibers within four weeks[51]. The results from human clinical studies using PDLSCs are encouraging yet still very limited and variable [56-58].Recent studies even showed that MSCs can be derived from normal and inflamed gingival tissues which might be a more abundant potential source compared to PDL[59, 60]. More future studies are required to assess the benefits and safety of using MSCs from dental origin and important challenges need to be addressed before applying such treatment protocols to standard clinical practice[56]. 3. Final remarks 10. The future applications of stem cells and tissue engineering in periodontal therapy are one of the most promising techniques capable of meeting a variety of patients’ needs. However, many challenges need to be tackled before reaching a clear consensus regarding stem cells utilization. Biologically, it is essential to fully understand the key cellular events that occur during periodontal development, knowledge of the specific cell types, the inductive factors, the cellular and molecular processes involved in formation of the periodontium, and the mechanism in which stem cells promote differentiation. Technical challenges such as the appropriate delivery system and the most suitable scaffold matrix should be addressed by advancing the techniques used in handling stem cells. Concerning the clinical challenges, it is very important to understand the immune response after the delivery of the stem cells and the safety considerations related to the use stem cells. In addition, the cost effectiveness and the patient benefits should be considered. More high quality clinical research is essential to ensure that such novel approaches supported by robust data and are effective in meeting the patients’ needs. 4. 5. 6. 7. 8. 9. 11. 12. 13. 14. 15. 16. 17. 18. References 19. 1. 2. Pihlstrom, B.L., B.S. Michalowicz, and N.W. Johnson, Periodontal diseases. Lancet, 2005. 366(9499): p. 1809-1820. Albandar, J.M., Epidemiology and risk factors of periodontal diseases. Dental clinics of North America, 2005. 49(3): p. 517-32, v-vi. 20. Burt, B., et al., Position paper: epidemiology of periodontal diseases. Journal of periodontology, 2005. 76(8): p. 1406-19. Polimeni, G., A.V. Xiropaidis, and U.M. Wikesjo, Biology and principles of periodontal wound healing/regeneration. Periodontol 2000, 2006. 41: p. 30-47. Keestra, J.A., et al., Non-surgical periodontal therapy with systemic antibiotics in patients with untreated chronic periodontitis: a systematic review and metaanalysis. J Periodontal Res, 2015. 50(3): p. 294-314. Darby, I.B. and K.H. Morris, A systematic review of the use of growth factors in human periodontal regeneration. J Periodontol, 2013. 84(4): p. 465-76. Chen, F.M. and Y. Jin, Periodontal tissue engineering and regeneration: current approaches and expanding opportunities. Tissue Eng Part B Rev, 2010. 16(2): p. 219-55. Xiao, Y., et al., Expression of extracellular matrix macromolecules around demineralized freeze-dried bone allografts. Journal of Periodontology, 1996. 67(11): p. 1233-1244. Venezia, E., et al., The use of enamel matrix derivative in the treatment of periodontal defects: A literature review and meta-analysis. Critical Reviews in Oral Biology & Medicine, 2004. 15(6): p. 382-402. Stavropoulos, A., et al., Deproteinized bovine bone (Bio-Oss((R))) and bioactive glass (Biogran((R))) arrest bone formation when used as an adjunct to guided tissue regeneration (GTR) - An experimental study in the rat.Journal of Clinical Periodontology, 2003. 30(7): p. 636-643. Ripamonti, U. and A.H. Reddi, Tissue engineering, morphogenesis, and regeneration of the periodontal tissues by bone morphogenetic proteins. Critical Reviews in Oral Biology & Medicine, 1997. 8(2): p. 154-163. Sander, L. and T. Karring, HEALING OF PERIODONTAL LESIONS IN MONKEYS FOLLOWING THE GUIDED TISSUE REGENERATION PROCEDURE A HISTOLOGICAL STUDY. Journal of Clinical Periodontology, 1995. 22(4): p. 332-337. Bartold, P.M., et al., Tissue engineering: a new paradigm for periodontal regeneration based on molecular and cell biology. Periodontology 2000, 2000. 24: p. 253-269. Aukhil, I., Biology of wound healing. Periodontology 2000, 2000. 22: p. 44-50. Ivanovski, S., et al., Stem cells in the periodontal ligament. Oral Diseases, 2006. 12(4): p. 358-363. Han, J., et al., Stem cells, tissue engineering and periodontal regeneration. Australian Dental Journal, 2014. 59: p. 117-130. Pejcic, A., et al., STEM CELLS FOR PERIODONTAL REGENERATION. Balkan Journal of Medical Genetics, 2013. 16(1): p. 7-11. Michalowicz, B.S., et al., Evidence of a substantial genetic basis for risk of adult periodontitis. Journal of Periodontology, 2000. 71(11): p. 1699-1707. Hughes, F.J., M. Ghuman, and A. Talal, Periodontal regeneration: a challenge for the tissue engineer? Proceedings of the Institution of Mechanical Engineers Part H-Journal of Engineering in Medicine, 2010. 224(H12): p. 1345-1358. Smith, A., A glossary for stem-cell biology. Nature, 2006. 441(7097): p. 1060-1060. International Dental Journal of Student’s Research, April - June 2015;3(2):88-92 {91} 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. 37. 38. 39. Morrison, S.J., N.M. Shah, and D.J. Anderson, Regulatory mechanisms in stem cell biology. Cell, 1997. 88(3): p. 287-98. Lin, N.H., S. Gronthos, and P.M. Bartold, Stem cells and periodontal regeneration. Australian Dental Journal, 2008. 53(2): p. 108-121. Takahashi, K., et al., Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell, 2007. 131(5): p. 861-872. Takahashi, K. and S. Yamanaka, Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell, 2006. 126(4): p. 663-76. Egusa, H., et al., Gingival fibroblasts as a promising source of induced pluripotent stem cells. PLoS One, 2010. 5(9): p. e12743. Wada, N., et al., Induced pluripotent stem cell lines derived from human gingival fibroblasts and periodontal ligament fibroblasts. J Periodontal Res, 2011. 46(4): p. 438-47. Nomura, Y., et al., Human periodontal ligament fibroblasts are the optimal cell source for induced pluripotent stem cells. Histochem Cell Biol, 2012. 137(6): p. 719-32. Young, H.E., et al., Human reserve pluripotent mesenchymal stem cells are present in the connective tissues of skeletal muscle and dermis derived from fetal, adult, and geriatric donors. Anatomical Record, 2001. 264(1): p. 51-62. Smith, C. and B. Storms, Hematopoietic stem cells. Clinical Orthopaedics and Related Research, 2000(379): p. S91-S97. Pittenger, M.F., et al., Multilineage potential of adult human mesenchymal stem cells. Science, 1999. 284(5411): p. 143-7. Blau, H.M., T.R. Brazelton, and J.M. Weimann, The evolving concept of a stem cell: Entity or function? Cell, 2001. 105(7): p. 829-841. Lin, N.H., S. Gronthos, and P.M. Bartold, Stem cells and future periodontal regeneration. Periodontol 2000, 2009. 51: p. 239-51. Till, J.E. and E.A. McCulloch, DIRECT MEASUREMENT OF RADIATION SENSITIVITY OF NORMAL MOUSE BONE MARROW CELLS. Radiation Research, 1961. 14(2): p. 213-&. Dominici, M., et al., Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy, 2006. 8(4): p. 315-317. Horwitz, E.M., et al., Clarification of the nomenclature for MSC: The international society for cellular therapy position statement. Cytotherapy, 2005. 7(5): p. 393395. Korbling, M. and Z. Estrov, Adult stem cells for tissue repair - A new therapeutic concept? New England Journal of Medicine, 2003. 349(6): p. 570-582. Gronthos, S., et al., Telomerase accelerates osteogenesis of bone marrow stromal stem cells by upregulation of CBFA1, osterix, and osteocalcin. Journal of Bone and Mineral Research, 2003. 18(4): p. 716-722. Kawaguchi, H., et al., Enhancement of periodontal tissue regeneration by transplantation of bone marrow mesenchymal stem cells. J Periodontol, 2004. 75(9): p. 1281-7. Hasegawa, N., et al., Behavior of transplanted bone marrow-derived mesenchymal stem cells in periodontal 40. 41. 42. 43. 44. 45. 46. 47. 48. 49. 50. 51. 52. 53. 54. 55. 56. 57. defects. Journal of Periodontology, 2006. 77(6): p. 1003-1007. Li, H., et al., Application of autologous cryopreserved bone marrow mesenchymal stem cells for periodontal regeneration in dogs. Cells Tissues Organs, 2009. 190(2): p. 94-101. Yamada, Y., et al., A novel approach to periodontal tissue regeneration with mesenchymal stem cells and platelet-rich plasma using tissue engineering technology: A clinical case report. Int J Periodontics Restorative Dent, 2006. 26(4): p. 363-9. Tobita, M., et al., Periodontal tissue regeneration with adipose-derived stem cells. Tissue Eng Part A, 2008. 14(6): p. 945-53. La Noce, M., et al., Dental pulp stem cells: State of the art and suggestions for a true translation of research into therapy. Journal of Dentistry, 2014. 42(7): p. 761768. Gronthos, S., et al., Postnatal human dental pulp stem cells (DPSCs) in vitro and in vivo. Proceedings of the National Academy of Sciences of the United States of America, 2000. 97(25): p. 13625-13630. Miura, M., et al., SHED: Stem cells from human exfoliated deciduous teeth. Proceedings of the National Academy of Sciences of the United States of America, 2003. 100(10): p. 5807-5812. Seo, B.M., et al., Investigation of multipotent postnatal stem cells from human periodontal ligament. Lancet, 2004. 364(9429): p. 149-55. Shi, S., et al., The efficacy of mesenchymal stem cells to regenerate and repair dental structures. Orthod Craniofac Res, 2005. 8(3): p. 191-9. Ding, G., et al., Allogeneic periodontal ligament stem cell therapy for periodontitis in swine. Stem Cells, 2010. 28(10): p. 1829-38. Koch, P.L., N. Tuross, and M.L. Fogel, The effects of sample treatment and diagenesis on the isotopic integrity of carbonate in biogenic hydroxylapatite. Journal of Archaeological Science, 1997. 24(5): p. 417-429. Isaka, J., et al., Participation of periodontal ligament cells with regeneration of alveolar bone. J Periodontol, 2001. 72(3): p. 314-23. Lang, H., et al., Formation of differentiated tissues in vivo by periodontal cell populations cultured in vitro. J Dent Res, 1995. 74(5): p. 1219-25. Lang, H., N. Schuler, and R. Nolden, Attachment formation following replantation of cultured cells into periodontal defects - a study in minipigs. Journal of Dental Research, 1998. 77(2): p. 393-405. Liu, Y., et al., Periodontal ligament stem cell-mediated treatment for periodontitis in miniature swine. Stem Cells, 2008. 26(4): p. 1065-73. Kim, S.H., et al., Alveolar bone regeneration by transplantation of periodontal ligament stem cells and bone marrow stem cells in a canine peri-implant defect model: a pilot study. J Periodontol, 2009. 80(11): p. 1815-23. Park, J.Y., S.H. Jeon, and P.H. Choung, Efficacy of periodontal stem cell transplantation in the treatment of advanced periodontitis. Cell Transplant, 2011. 20(2): p. 271-85. Trofin, E.A., P. Monsarrat, and P. Kemoun, Cell therapy of periodontium: from animal to human? Frontiers in Physiology, 2013. 4: p. 11. Feng, F., et al., Utility of PDL progenitors for in vivo tissue regeneration: a report of 3 cases. Oral Dis, 2010. 16(1): p. 20-8. International Dental Journal of Student’s Research, April - June 2015;3(2):88-92 {92} 58. Gault, P., et al., Tissue-engineered ligament: implant constructs for tooth replacement. J Clin Periodontol, 2010. 37(8): p. 750-8. 59. Ge, S., et al., Isolation and characterization of mesenchymal stem cell-like cells from healthy and inflamed gingival tissue: potential use for clinical therapy. Regen Med, 2012. 7(6): p. 819-32. 60. Yang, H., et al., Comparison of mesenchymal stem cells derived from gingival tissue and periodontal ligament in different incubation conditions. Biomaterials, 2013. 34(29): p. 7033-47. __________________________________________________ International Dental Journal of Student’s Research, April - June 2015;3(2):88-92 {93} CLINICAL DIAGNOSIS & GUIDELINES Periodontal Disease in Immunodeficient Patients: Clinical Guidelines for Diagnosis and Management Introduction Morvarid Oveisi1, Oriyah Barzilay2, Ahmed A. Hanafi3 1,2,3 Group of Matrix Dynamics, Faculty of Dentistry, University of Toronto. 3 Periodontology Department, Faculty of Oral and Dental Medicine, Cairo University Corresponding Author: Morvarid Oveisi Email: [email protected] Access this Article Online www.idjsr.com Use the QR Code scanner to access this article online in our database Article Code: IDJSR SE 0171 Quick Response Code Abstract Primary immunodeficiency diseases are rare hereditary conditions that usually occur at a young age; however, secondary immunodeficiency is acquired due to disease, drug treatment and is increasing in frequency among the population. Although periodontal diseases related to these conditions are secondary to other life threatening manifestations, they are very common and easily detectable by the patient, patient guardians and periodontists. Periodontists have a major role in both helping to detect undiagnosed diseases, as well as improving the oral care of diagnosed patients, thus a thorough knowledge of these conditions, causes, local and systemic involvement, diagnostic tools and proper management is very important. This article summarizes selected primary and secondary immunodeficiency conditions such as neutropenia, leukocyte adhesion deficiency (LAD) and ChediakHegashi syndrome, and places schematic, diagnostic, and management steps that may help periodontists manage unexplained severe periodontal diseases related to immunodeficiency. Immunodeficiency diseases are defects in the immune system in which the host defense mechanism cannot function properly. Primary immunodeficiency is inherited and therefore is caused by a gene defect. However, secondary immunodeficiency conditions are acquired and usually happen due a defect of lymphocyte function as a result of the usage of drugs, irradiation or invasion of pathogens such as HIV virus and measles.[1] There are many types of diseases caused by primary immunodeficiency , which is a result of a defect in Tcell or B-cell function, antibody deficiency, or loss of phagocyte function and/or number[2] ,which for the latter these can be included as any deficiency in the adhesion process of neutrophil [3], NADPH oxidase or chemotaxis.[4] The epidemiology of the diseases differ based on race, gender, ethnic factors and geographic region[4]. It has been estimated that one in 1200 people are affected by primary Immunodeficiency. Due to cancer therapies, usage of immunosuppressant’s and other biological therapies, the occurrence of secondary immunodeficiency is growing. Organ damage is preventable if there is minimal delay in the diagnosis of immunodeficiency[5]. Periodontal disease is an inflammatory state of the gingiva which affects the supporting structure of the teeth.[6, 7]. Periodontal disease is caused by plaque accumulation , and poor dental hygiene[8] leading to inflammation of surrounding tissue[4].It has been observed that individuals suffering from any types of immunodeficiency diseases may manifest some oral, dental and facial problems[4]which include periodontal diseases, oral lesions and developmental abnormalities. These can be a sign of immunity defect. Therefore, it is crucial for physicians and dentist to be able to recognize these systematic disorders by the oral manifestation, carry out an accurate diagnosis and perform the corresponding treatment[9]. Neutropenia Neutropenia is defined by a low absolute neutrophil count (ANC) in the blood lasting more than 6 months, which can cause recurrent infections to a patient [10] with varying severity from stomatitis International Dental Journal of Student’s Research, April - June 2015;3(2):93-104 {94} and gingivitis, to more severe pneumonia and sepsis. [11] Different forms of neutropenia such as cyclic neutropenia, chronic benign neutropenia and severe congenital neutropenia (Kostmann syndrome) can all cause periodontal disease.[10, 12] Chronic benign neutropenia Chronic neutropenia is defined by a non-cyclic low count of neutrophils in the blood without a known underlying systemic disease lasting less than 6 months. It is the most common form of neutropenia in infants and children and is usually not inherited. 80-98% of patients tested positive for the antineutrophil antibody [13]. Its manifestation is less severe than Kostmann syndrome, includes high incidence of otitis media, upper respiratory infections, lymphadenopathy and pneumonia but may develop into life threatening infections and sepsis.[10] Oral Manifestations includes severe gingival inflammation, edematous and hyperplastic papilla,[14] may progress into periodontitis [15]leading to severe horizontal bone loss and teeth mobility [16] Diagnosis is achieved by a persistent ANC 0.5x109/l with a normal total white blood cell count due to elevated numbers of lymphocytes and monocytes [17] and confirmed by anti-neutrophil antibody. Kostmann Syndrome Severe congenital neutropenia (Kostmann Syndrome) is a rare hereditary syndrome characterized by a very low ANC (less than 0.2x109/l) [18] due to maturation arrest during myelopoiesis process [4]and increased apoptosis of myeloid progenitor cells in bone marrow.[19] Initial symptoms can be summarized as recurrent bacterial infections of the skin, mucosa leading to cellulitis, perirectal abscess, stomatitis, meningitis, pneumonia, and sepsis [20] . Long term symptoms are periodontitis, splenomegaly and hepatomegaly, osteoporosis and myelodysplastic syndrome/acute myeloid leukaemia (MDS/AML) [19]. Oral findings are usually more severe than other forms of neutropenia, with recurrent painful ulceration, [21] diffuse gingival inflammation, alveolar bone loss, teeth mobility and loss of both dentition [22] Persistent ANC less than 0.5x109/l is a significant laboratory finding and diagnosis is confirmed with bone marrow aspiration showing an arrest of neutrophil hematopoiesis at the promyelocyte/myelocyte stage.[10] Cyclic neutropenia Cyclic Neutropenia is characterized by the repetitive occurrence of neutropenia at average of 21 day period and last for approximately 3-6 days[23].The mutation is passed along in an autosomal dominant manner. It has been observed that this disease is associated with the mutation in ELA2 gene mapped to chromosome 19p13.3 which encodes neutrophil elastase. Mutations in this gene lead to a shortened neutrophil life[24] . It is characterized by fever, mouth ulcers, lymphadenopathy, multiple abscess formation, exhaustion and susceptibility of infection which can be lethal [25-29]. Reduction in the number of polymorph nuclear leukocytes (PMNs) can be associated with rapid and destructive periodontal disease including aphthous-like lesions[10]. The initial oral characterization of patient includes repetitive ulceration showing little evidence of an inflammatory halo[30], severe gingival inflammation and recession[14, 25],which extended from the gingiva into the alveolar mucosa[25].Recurrent gingival bleeding along with fever was noted as a sign of this disease [25, 31],pocket depths exceeded the 6- to 8-mm range[25] with various levels of tooth mobility [31]. Diagnosis requires serial measurements of the ANC (<1,500) daily or at least three times per week for four to six weeks[32]. It has been demonstrated that Granulocyte ColonyStimulating Factor(GCSF) can be an efficient treatment for neutropenia[33], as it can lead to a 10 fold increase in ANC and result in a higher life expectancy[34]. Dental management is necessary for these patients to control infections. Regular dental appointments to check for the accumulation of bacterial plaque. Use of chlorhexidine gluconate mouth wash and a light polishing and scaling in some part of the teeth [25, 31]. Prophylactic antimicrobials. Invasive dentistry should be avoided in neutropenic episodes. Oral surgeries to be performed only under specific antibacterial (after microbiological testing) and corticosteroid coverage [21] LAD LAD is a rare, autosomal recessive, primary immunodeficiency syndrome; characterized by impaired phagocytic functions[35]. LAD is classified International Dental Journal of Student’s Research, April - June 2015;3(2):93-104 {95} according to causative gene mutation into 3 types: LAD I, LAD II and LAD III [36-38]. LAD I is caused by mutation in gene ITGB2 which encodes for CD11/CD18 [39, 40] and ultimately decreases the expression of three integrins on leukocyte surfaces CD11a,CD11b and CD11c and preventing the adhesion of neutrophils to endothelial cells)[41]. Characterized delayed separation of umbilical cord, major bacterial infections with no pus formation [35] and impaired wound healing [40], the severity of clinical features are directly related to degree of CD18 deficiency and can be divided into severe (less than 1% CD18 expression) and moderate ( 2.5% to 10% CD18 expression) [42, 43]. Morbidity rate of severe LAD I is high before the age of 5 [44]. In LAD II, different gene mutations cause defects in the specific Golgi GDP-fucose transporter [45, 46] which reduce CD15s (Sialyl-Lewis X) on the leukocyte surface, thus affecting the rolling phase of neutrophil adhesion [35]. This is characterized by mental retardation and less severe infections in adolescence [41] . Less common LAD III, was only reported in 4 patients suffering from bacterial infections [38] and severe bleeding tendency[47], is believed to be caused by general defect in integrin activation [38]. Periodontal involvement starts as gingivitis at a young age, just after primary teeth eruption. Deep pocket formation and extensive bone loss [48, 49] progress until partial or total premature loss of both primary and permanent teeth [35]. Several case reports showed oral ulceration and delayed wound healing in more than 80% of patients.[35] Primarily, blood test of patients with LAD shows leukocytosis (20,000 to 80,000 cells/ml)[40, 50].Rebuck skin window or Boyden Chamber shows decreased neutrophil migration and is confirmed using flow cytometry which shows CD18 deficiency in LAD I and sialy-Lewis x ligand deficiency in LAD II [51]. Additional histological analysis of gingival biopsy showed abundance of leukocytes in blood vessels and no leukocyte in tissue [52]. International Dental Journal of Student’s Research, April - June 2015;3(2):93-104 {96} Figure 1: Schematic laboratory tests for diagnosis of listed diseases. LAD, Leukocyte adhesion Deficiency. DHR, Dihydrorhodamine. HIES, Hyper immunoglobulin E syndrome. CVID, Common Variable Immunodeficiency. Bone marrow transplant is the treatment of choice for young LAD patients [53, 54].However if not possible, several maneuvers to adjust host response can be achieved such as white blood cell transfusion, antibiotics, interferon and allogenic stem cell transplant [55]. Periodontal treatment usually ends with tooth loss [10] however maintaining the teeth is advocated with the goal of improving patients’ physiologic and psychological health by Periodic oral prophylaxis [56] Prophylactic Antimicrobial Fluoride application and diet counseling Extraction should be avoided (due to delayed wound healing) Chediak-Hegashi Syndrome Chediakhigashi is a rare condition which is inherited in an autosomal manner. This disease is usually fatal and appears with the irregularly enormous lysosomal granules in the leukocytes [57, 58]. This disorder is characterized by numerous repetitive bacterial infections, oculocutaneous albinism, susceptibility to bruising, and mucosal bleeding as well as peripheral neuropathy. In addition, patients may show neurologic dysfunction and movement disorders[3]. Furthermore, the accelerated phase of CHS named Hemophagocyticlymphohistiocytosis (HLH) can be recognized by cytopenia, fever, bleeding, lymphadenopathy and hepatosplenomegaly [59-61]. This disorder is connected to the fusion of cytoplasmic granules which can take place in the myelopoieses and can lead to the death of myeloid precursors in the marrow and cause neutropenia. Also neutrophils can have a problem in phagocytosis, chemotaxis and killing bacteria[62]. Intraoral examination showed a full mouth plaque score of 85% [63], gingival bleeding and teeth mobility [3, 58] , high frequency of periodontal pockets and bone loss at an early age [10, 64, 65], probing showed more than 30% of the sites 5-8mm deep with concomitant recession defects [66] Blood testing and examination of giant granules within neutrophils, lymphocytes and natural killer cells using nitrobluetetrazolium dye [10] are essential for diagnosis. Bone marrow aspiration and examination of giant eosinophilic or azurophilic cytoplasmic inclusion bodies within the myeloid lineage cells show a positive reaction to peroxidase staining People with Chediak-Higashi disorder can be recognized at a young age, and bone marrow transplantation can be a positive treatment which can lower the risk of periodontal disease.[3] It has been noted that continuous periodontal treatments with regular follow up can help patients avoid further infection and reduce gingival inflammation [67]. In addition,Kornmanet et al described the importance of periodontal therapy in preventing progressive periodontitis [68]. Long-term antibiotic treatment such as amoxicillin[69] is administered to patients with progressive periodontal disease along with metronidazole [70]to help reduce the periodontal probing depth and promote attachment in patients with the milder form of the disease. [67].However, full mouth extraction is inevitable in patients with severe progressive periodontal disease refractory to treatment .[71] Chronic granulomatous Disease Chronic granulomatous disease(CGD) is a very rare immune deficiency syndrome perpetuated genetically. CGD can be characterized by the mutation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase component, leading to the failure of neutrophils and macrophages in killing invading pathogens by impairing the respiratory burst. CYBB is a gene responsible for encoding the gp91phox subunit and is reported as the most common site of mutation. Liver abscesses, skin infections, pneumonia, osteomyelitis, as well as cervical or inguinal lymphadenitis can be seen in young patients[72, 73]. Furthermore, infection and sterile hyper inflammation is also present in CGD patient[74]. Management of CDG is based on control of infections. Broad spectrum antibacterial (as cotrimoxazole)[75], antifungal prophylaxis (asitraconazole)[76]. Interferon-ƴ has been shown to improve oxidase activity in neutrophils and monocytes of some patients[77], and to reduce infection rates. Bone marrow transplantation is effective and a more predictable treatment [78]. Some case reports show oral difficulties including oral ulcers[79-84] such as multiple buccal mucosal ulcers in direct contact with dental plaque [81], severe gingivitis, periodontitis [79-81], enamel hypoplasia [84], oral candidiasis [85] , granulomatous mucositis in the upper lip[86] and the soft palate [87], geographic tongue[84] and generalized prepubertal periodontitis,[88] loss of attachment and recession of gingival was noted[88] International Dental Journal of Student’s Research, April - June 2015;3(2):93-104 {97} Variations of oral findings in CGD patients [72, 89, 90], and Neutrophil dysfunction[10, 91] are probably due to immunosuppressive therapy specially steroids [92]. Patients with CGD can be diagnosed through flowcytometery, dihydrorhodamine 123 (DHR) assay[93] and the nitrobluetetrazolium Test [94] Treatment: Regular dental care and frequent follow-up. Antibacterial mouth washes. Antibiotics such as clavulanic acid and amoxicillin is needed for any dental work and surgery related to bacteremia[95] Antimycotic prophylaxis[95] Hyper Immunoglobulin E Syndrome HEIS or Jobs syndrome is a rare multisystemic disease causing host immune defects as well as nonimmunological manifestations [96]. It presents itself in two forms, the more common autosomal dominant (AD-HEIS) and the less common autosomal recessive (AR-HEIS) [97], both characterized by high level of immunoglobulin E levels in blood, chronic eczema, recurrent skin and lung infections [98] and decreased bone density [99]. Although the etiology of disease is not clear, several studies concluded that immune defects may be due to defective neutrophil chemotaxis, humoral and cellular immune response impairment including T-cell cytokine signal disruption. Recent gene analysis showed that most AD-HIES have a mutation in gene STAT3 which causes poor maturation and activation of T17 helper cells, however AR-HIES (different clinical picture with no dental abnormalities) may be attributed to different gene mutation, TYK2 gene (Tyrosine Kinase 2), or the DOCK8 gene (Dedicator of Cytokinesis 8)[100]. Oral findings of AD-HIES is very characteristic to retention of primary teeth or ― Double-row‖ dentition which is found in most reported cases (due to persistence of Her twig epithelial root sheath on root surface, thus preventing root resorption causing delayed shedding) , severe candidiasis and oral infections, poor oral hygiene and high plaque index, gingivitis but rare progression to periodontitis, except for 2 patients [79, 101] ( . This data contradicts most immunodeficiency syndromes, which are more likely to cause severe periodontal destruction.) There is no one laboratory test that confirms HIES, rather a scoring system (fig. 1)-based on all clinical and laboratory tests is used to confirm HIES, therefore diagnosis of HIES is usually difficult. Recently some reported laboratory tests were added to NIH-HIES score, that helps better detect HIES, as very low Th17 CD4 cell count, and the genetic analysis.[102]The presence of retained primary teeth and unerupted permanent teeth resembles other syndromes like Cleidocranial syndrome, Gardner’s Syndrome, and Down syndrome. A differential diagnosis with these syndromes is noted and confirmed by clinical and laboratory tests. [103] Treatment is based on prophylactic antimicrobials, intravenous immunoglobulin, and bone marrow transplant shows some success but not a full recovery [104], dental management is focused on Periodic follow up and oral hygiene assessment. Extraction of primary teeth at correct time of shedding to prevent permanent teeth impaction and enhance oral hygiene. Antifungal and antibacterial for oral infections Orthodontic correction if needed. Prognosis of gingival healing after extraction is fairly good with no complications and in most patients periodontal health of permanent teeth can be maintained. International Dental Journal of Student’s Research, April - June 2015;3(2):93-104 {98} Table 1: Grimbacher et al[105] Scoring System for HIES 0 1 Highest serum immunoglobulin E <200 200-500 Skin abscesses None Pneumonia (episodes) None Parenchymal lung anomalie Absent Retained primary teeth None Clinical and Laboratory Findings 1 2 3 8 10 501-1000 1001-2000 >2000 1-2 3-4 >4 1 2 2 4 5 6 3 >3 Bronchiectasis Pneumatocele 3 o 15-20 7 >3 o Scoliosis <10 Fractures with minor trauma None Highest eosinophil count (cells/_L) <700 Characteristic face Absent Midline anomaly Absent Eczema Absent Mild Moderate Sever Upper respiratory infections/year 1-2 3 4-6 >6 Candidiasis None Oral Finger Nails Systemic Fatal infection Absent Present Hyperextensibility Absent Present Lymphoma Absent Present High palate Absent Present >20o 1-2 >2 700-800 >800 Mild Present Present Present <15 points: patient not affected; 15 to 39 points: possible diagnosis; 40 to 59 points: probable diagnosis; >60 points definitive diagnosis International Dental Journal of Student’s Research, April - June 2015;3(2):93-104 {99} Common Variable Immunodeficiency Common variable immunodeficiency (CVID) is a common heterogeneous primary immune deficiency. Patients with CVID have a deficiency in humoral immunity leading to a defective antibody response, causing repetitive infections of the gastrointestinal and upper respiratory tracts, and susceptibility to some cancers such as lymphoma and autoimmune diseases. Hypogammaglobulinemia has also seen in these patients. Although normal B-cell numbers have been identified in lymphoid tissue and peripheral blood of patients, it has been noted that B-cells of these individuals have difficulty in differentiating into immunoglobulin-secreting plasma cells. Furthermore, it should be mentioned that the deficiency in monocyte and macrophage function has also been recognized. In addition, in some CVID patients, Tcell malfunction has been identified with decreased CD4 lymphocytes and T-cell receptors, loss of antigen-specific and quick death of T-cells.[4] Individual suffering from CVID have been diagnosed by having a low level of IgG and IgA[106]. Mutations in a group of genes asTNFRSF13B gene[107] involved in B-cells result in having a defected immunity in CVID[108] Clinical examinations show dental problems such as gingivitis and lichenoid lesions with Wickham striae,[109] necrotizing ulcerative periodontitis (NUP) [109, 110] , severe periodontitis and gingival pain along with bleeding and tooth mobility was demonstrated in a case report.[110] In order to treat CVID, the primary method used is to replace the antibody by an intravenous or subcutaneous means. This occurs in doses of 400600mg of antibody per kilogram of the patient’s weight per month.[111] Dental management as reported in some case reports include Regular oral prophylaxis with crown polishing [109] Chlorhexidinedigluconate rinse is recommended twice a day. Antibiotic therapy[109, 110] such as Amoxycillin and clavulanic acid[109] Acquired immunodeficiency syndrome HIV advancement can be detect by monitoring the HIV viral load and T helper cells(CD4+) count,however,it should be noted that there are some common oral manifestations associated with HIV positive patients. Therefore, it can be a useful indicator for screening the immune condition of potential HIV positive individuals and can be easily recognized and detected by clinicians.[113-116] It has been estimated that 70-90% of individual suffering from HIV manifest oral lesions during their phase of the disease [117-119]. Individuals with oral manifestations have less CD4+ than ones without, and it has been observed that there is a correlation between oral candidiasis and a decrease in the CD4+ count(less than 200 cells/mm3).[120] Some of the important oral manifestation and Lesions present in HIV positive patients are Oral candidiasis (most common oral lesion) [113, 121-123] which are divided in to three groups;pseudomembranous candidiasis, erythematous candidiasis and angular cheilitis[114] Oral hairy leukoplakia [114]. ulcerative disease such as herpes simplex virus, Aphthous ulcerations[114, 124], Neutropenic ulceration [124] linear gingival erythema[120, 124] oral warts-human papilloma virus[124] Necrotizing Ulcerative gingivitis and Periodontitis(NUG/NUP)[124, 125] The follow up appointments are needed for dental care such as scaling and root planning. A 10% povidone-iodine lavage or 0 .12% chlorhexidine gluconate can be used for the elimination of dental plaque and necrotic soft tissue.Utilizing antibiotics such as clindamycin, metronidazole 500g and amoxicillin can be helpful for the treatment. It is crucial to establish proper nutrition in order to reduce potential issues in the oral cavity that can be produced by poor nutrition, as well as manage the patient’s pain.[124] Leukemia Leukemia is a type of a cancer caused by an uncontrolled differentiation and proliferation of blood cell precursors resulting in the production of immature cells.Clinically, leukemia is classified into two types: chronic and acute, with the acute phase possibly being fatal. In addition, according to histogenicity, leukemia is divided in to lymphocytic or myelocytic depending on the origin of the cells[126-128]. Acute myeloid leukemia(AML) is more common in adults and acute lymphoid leukemia (ALL) is mostly seen in children[128-130]. Acute myeloblastic leukemia (AML) is characterized by symptoms of Acquired immunodeficiency syndrome(AIDS) is a disease caused by a human immunodeficiency virus. In this condition HIV targets and attacks T helper cells(CD4) resulting in immune response suppression , the disability of the body’s response to the invading pathogen, predisposes the patient to neurological problems, opportunistic infections ,malignancies and oral manifestation [112]. International Dental Journal of Student’s Research, April - June 2015;3(2):93-104 {100} pancytopenia including fatigue, weaknesses, infection, gingival bleeding, ecchymoses, menorrhagia, and epistaxis [131, 132]. The direct penetration of leukemic cells in lymph nodes, spleen, central nervous system and gingival has been reported [126, 129, 133-135]. Oral complication can be observed in all types of leukemia[136]. Individuals having leukemia are suffering from extreme enlargement of the gingiva along with bleeding[127, 135-139], bulbous enlargement in the interdental papillae [126, 127] a pale blue gum with glazed texture ,and loss of stippling is one the symptoms of leukemia[126, 127], generalized horizontal bone loss was reported[127] however in some cases bone loss is not recognized[126] Ulceration and petechiae was noted as a frequent sign[135]. In patient with acute monocytic leukemia and acute myelomonocytic leukemia, gingival infiltration of leukemic cells are commonly seen[140]. Diagnosis by complete blood count peripheral blood smear, shows the presence of blast cells and reveals the type and quantity of white blood cells[126], and flow cytometry of peripheral blood are used for leukemia diagnosis[126, 127], biopsy such as bone marrow aspiration also can be used to confirm diagnosis and type of leukemia[126, 127, 135] Regular oral prophylaxis is needed. Antibacterials can be used in conjugation with scaling and sub gingival debridement to lower the risk of dental infection during the chemotherapy. Tooth extraction of hopeless teeth can eliminate the infection.[141] Managing periodontal disease of immunodeficient patients is essential for improvement of their physical and psychological health, thus knowledge of these conditions, diagnostic methods and management options is crucial for every dentist. Diagnosis of these diseases is challenging, however some clues may guide the clinician towards a definitive diagnosis, so it is important that precise steps of history taking (medical, familial and dental), clinical examination (extra and intra oral) and laboratory investigations are followed to achieve a successful diagnosis. References 2. 3. 5. 6. 7. 8. 9. 10. 11. 12. 13. 14. 15. Conclusion 1. 4. 16. 17. 18. 19. 20. Janeway, C.A., et al., Immunobiology: the immune system in health and disease. Vol. 2. 2001: Churchill Livingstone. Notarangelo, L., et al., Primary immunodeficiency diseases: an update. Journal of Allergy and Clinical Immunology, 2004. 114(3): p. 677-687. Sollecito, T.P., et al., Systemic conditions associated with periodontitis in childhood and adolescence. A 21. 22. review of diagnostic possibilities. Medicina oral, patologia oral y cirugia bucal, 2004. 10(2): p. 142-150. Szczawinska-Poplonyk, A., et al., Oral manifestations of primary immune deficiencies in children. Oral Surgery, Oral Medicine, Oral Pathology, Oral Radiology, and Endodontology, 2009. 108(3): p. e9e20. Bright, P.D., et al., Laboratory clues to immunodeficiency; missed chances for early diagnosis? Journal of clinical pathology. 68(1): p. 1-5. Ellison, S.A., Oral bacteria and periodontal disease. Journal of dental research, 1970. 49(2): p. 197-202. Genco, R.J., R.T. Evans, and S.A. Ellison, Dental research in microbiology with emphasis on periodontal disease. The Journal of the American Dental Association, 1969. 78(5): p. 1016-1036. Ram, V.S., et al., Bonebiomarkers in Periodontal Disease: A Review Article. Garcia, R.I., M.M. Henshaw, and E.A. Krall, Relationship between periodontal disease and systemic health. Periodontology 2000, 2001. 25(1): p. 21-36. Deas, D.E., S.A. Mackey, and H.T. McDonnell, Systemic disease and periodontitis: manifestations of neutrophil dysfunction. Periodontology 2000, 2003. 32(1): p. 82-104. Bernini, J.C., Diagnosis and management of chronic neutropenia during childhood. Pediatric Clinics of North America, 1996. 43(3): p. 773-792. Watanabeb, K., Prepubertal periodontitis: a review of diagnostic criteria, pathogenesis, and differential diagnosis. Journal of periodontal research, 1990. 25(1): p. 31-48. Bux J1, M.-E.C., Autoimmune neutropenia. Semin Hematal., 1992 Jan. 29(1): p. 45-53. Deasy, M.J., et al., Familial benign chronic neutropenia associated with periodontal disease: a case report. Journal of periodontology, 1980. 51(4): p. 206210. Zaromb, A., et al., Periodontitis as a manifestation of chronic benign neutropenia. Journal of periodontology, 2006. 77(11): p. 1921-1926. Reichart, P.A. and H. Dornow, Gingivo-periodontal manifestations in chronic benign neutropenia. Journal of clinical periodontology, 1978. 5(1): p. 74-80. Cutting, H.O. and J.E. Lang, Familial benign chronic neutropenia. Annals of internal medicine, 1964. 61(5_Part_1): p. 876-887. Aprikyan, A.A.G., et al., Cellular and molecular abnormalities in severe congenital neutropenia predisposing to leukemia. Experimental hematology, 2003. 31(5): p. 372-381. Carlsson, G.r., et al., Kostmann syndrome or infantile genetic agranulocytosis, part one: celebrating 50 years of clinical and basic research on severe congenital neutropenia. Acta Paediatrica, 2006. 95(12): p. 15261532. Hakki, S.S., et al., Periodontal status in two siblings with severe congenital neutropenia: diagnosis and mutational analysis of the cases. Journal of periodontology, 2005. 76(5): p. 837-844. Tirali, R.E. and S.B.Ç. Zeynep Yalçınkaya-Erdemci, Oral findings and clinical implications of patients with congenital neutropenia: a literature review. The Turkish journal of pediatrics. 55: p. 241-245. Defraia, E. and A. Marinelli, Oral manifestations of congenital neutropenia or Kostmann syndrome. Journal of Clinical Pediatric Dentistry, 2002. 26(1): p. 99-102. International Dental Journal of Student’s Research, April - June 2015;3(2):93-104 {101} 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. Dale, D.C. and W. Hammond, Cyclic neutropenia: a clinical review. Blood reviews, 1988. 2(3): p. 178-185. Dale, D.C., et al., Mutations in the gene encoding neutrophil elastase in congenital and cyclic neutropenia. Blood, 2000. 96(7): p. 2317-2322. Prichard, J.F., et al., Prepubertal periodontitis affecting the deciduous and permanent dentition in a patient with cyclic neutropenia: a case report and discussion. Journal of periodontology, 1984. 55(2): p. 114-122. Spencer, P. and J.E. Fleming, Cyclic neutropenia: a literature review and report of case. ASDC journal of dentistry for children, 1985. 52(2): p. 108. Andrews, R.G., et al., Chronic benign neutropenia of childhood with associated oral manifestations. Oral Surgery, Oral Medicine, Oral Pathology, 1965. 20(6): p. 719-725. Barrett, A.P., Neutropenic ulceration: a distinctive clinical entity. Journal of periodontology, 1987. 58(1): p. 51-55. Gates, G.F., Chronic neutropenia presenting with oral lesions. Oral Surgery, Oral Medicine, Oral Pathology, 1969. 27(4): p. 563-567. Scully, C., E. MacFadyen, and A. Campbell, Oral manifestation in cyclic neutropenia. British Journal of Oral Surgery, 1982. 20(2): p. 96-101. Yamahk, N., et al., Periodical gingival bleeding as a presenting symptom of periodontitis due to underlying cyclic neutropenia. Case report. Australian dental journal, 1993. 38(4): p. 272-276. Dale, D.C., ELANE-Related neutropenia. GeneReviews [Internet]. Seattle (WA): University of Washington, Seattle, 2002. Hammond Iv, W.P., et al., Treatment of cyclic neutropenia with granulocyte colony-stimulating factor. New England Journal of Medicine, 1989. 320(20): p. 1306-1311. Bux, J., et al., Influence of granulocyte antibodies on granulocyte function. Vox sanguinis, 1993. 64(4): p. 220-225. Nagendran, J., et al., Leukocyte adhesion deficiency: a case report and review. Journal of Dentistry for Children. 79(2): p. 105-110. Etzioni, A., Adhesion molecules in host defense. Clinical and diagnostic laboratory immunology, 1994. 1(1): p. 1. Yakubenia, S. and M.K. Wild, Leukocyte adhesion deficiency II. FEBS Journal, 2006. 273(19): p. 43904398. Etzioni, A. and R. Alon, Leukocyte adhesion deficiency III: a group of integrin activation defects in hematopoietic lineage cells. Current opinion in allergy and clinical immunology, 2004. 4(6): p. 485-490. Schmidt, S., M. Moser, and M. Sperandio, The molecular basis of leukocyte recruitment and its deficiencies. Molecular immunology. 55(1): p. 49-58. Anderson, D.C. and T.A. Springer, Leukocyte adhesion deficiency: an inherited defect in the Mac-1, LFA-1, and p150, 95 glycoproteins. Annual review of medicine, 1987. 38(1): p. 175-194. Dababneh, R., et al., Periodontal manifestation of leukocyte adhesion deficiency type I. Journal of periodontology, 2008. 79(4): p. 764-768. Fischer, A., et al., Leukocyte adhesion deficiency: molecular basis and functional consequences. Immunodefic Rev, 1988. 1(1): p. 39-54. Lakshman, R. and A. Finn, Neutrophil disorders and their management. Journal of clinical pathology, 2001. 54(1): p. 7-19. 44. 45. 46. 47. 48. 49. 50. 51. 52. 53. 54. 55. 56. 57. 58. 59. 60. 61. Movahedi, M., et al., Clinical and laboratory findings in Iranian patients with leukocyte adhesion deficiency (study of 15 cases). Journal of clinical immunology, 2007. 27(3): p. 302-307. Lühn, K., et al., The gene defective in leukocyte adhesion deficiency II encodes a putative GDP-fucose transporter. Nature genetics, 2001. 28(1): p. 69-72. Lübke, T., et al., Complementation cloning identifies CDG-IIc, a new type of congenital disorders of glycosylation, as a GDP-fucose transporter deficiency. Nature genetics, 2001. 28(1): p. 73-76. McDowall, A., et al., A novel form of integrin dysfunction involving β1, β2, and β3 integrins. Journal of clinical investigation, 2003. 111(1): p. 51. Dennison, D.K. and T.E. Dyke, The acute inflammatory response and the role of phagocytic cells in periodontal health and disease. Periodontology 2000, 1997. 14(1): p. 54-78. Meyle, J., Leukocyte adhesion deficiency and prepubertal periodontitis. Periodontology 2000, 1994. 6(1): p. 26-36. Todd 3rd, R.F. and D.R. Freyer, The CD11/CD18 leukocyte glycoprotein deficiency. Hematology/oncology clinics of North America, 1988. 2(1): p. 13-31. Meyle, J. and J.R. Gonzales, Influences of systemic diseases on periodontitis in children and adolescents. Periodontology 2000, 2001. 26(1): p. 92-112. Bowen, T.J., et al., Severe recurrent bacterial infections associated with defective adherence and chemotaxis in two patients with neutrophils deficient in a cell-associated glycoprotein. The Journal of pediatrics, 1982. 101(6): p. 932-940. Haddadin, I., et al., Bone marrow transplantation for leukocyte adhesion deficiency-I: Case report. Saudi Journal of Kidney Diseases and Transplantation, 2006. 17(4): p. 564. Hattori, H., et al., Successful human leukocyte antigen one antigen-mismatched related bone marrow transplantation in a 6-year-old boy with leukocyte adhesion deficiency syndrome. Pediatrics international, 2001. 43(3): p. 306-309. Engel, M.E., et al., Matched unrelated bone marrow transplantation with reduced-intensity conditioning for leukocyte adhesion deficiency. Bone marrow transplantation, 2006. 37(7): p. 717-718. Majorana, A., et al., Leukocyte adhesion deficiency in a child with severe oral involvement. Oral Surgery, Oral Medicine, Oral Pathology, Oral Radiology, and Endodontology, 1999. 87(6): p. 691-694. Wara-Aswapati, N., et al., Periodontitis in the child and adolescent. ASDC journal of dentistry for children, 1999. 66: p. 167-174. Delcourt-Debruyne, E.M.C., H.R.A. Boutigny, and H.F. Hildebrand, Features of severe periodontal disease in a teenager with Chediak-Higashi syndrome. Journal of periodontology, 2000. 71(5): p. 816-824. Nargund, A.R., et al., Accelerated Phase of Chediak Higashi Syndrome Mimicking Lymphoma—A Case Report. Journal of pediatric hematology/oncology. 32(6): p. e223-e226. Gajendra, S., et al., Accelerated phase at initial presentation in chediak-higashi syndrome: is it really uncommon? Pediatric hematology and oncology. 31(4): p. 382-385. Imran, T., et al., Chediak-Higashi syndrome presenting in accelerated phase. J Coll Physicians Surg Pak. 22(8): p. 539-41. International Dental Journal of Student’s Research, April - June 2015;3(2):93-104 {102} 62. 63. 64. 65. 66. 67. 68. 69. 70. 71. 72. 73. 74. 75. 76. 77. 78. 79. 80. Sánchez-Guiu, I., et al., Chediak–Higashi syndrome: description of two novel homozygous missense mutations causing divergent clinical phenotype. European journal of haematology. 92(1): p. 49-58. O'Leary, T.J., R.B. Drake, and J.E. Naylor, The plaque control record. Journal of periodontology, 1972. 43(1): p. 38-38. Hart, T.C. and J.C. Atkinson, Mendelian forms of periodontitis. Periodontology 2000, 2007. 45(1): p. 95112. Hanna, S. and A. Etzioni, Leukocyte adhesion deficiencies. Annals of the New York Academy of Sciences. 1250(1): p. 50-55. HR, M., Tooth mobility: a review of clinical aspects and research findings. Journal of periodontology, 1967. 38(6): p. Suppl: 686. Bailleul-Forestier, I., et al., Generalized periodontitis associated with Chediak-Higashi syndrome. Journal of periodontology, 2008. 79(7): p. 1263-1270. Kornman, K.S., et al., The influence of supragingival plaque control on clinical and microbial outcomes following the use of antibiotics for the treatment of periodontitis. Journal of periodontology, 1994. 65(9): p. 848-854. Winkel, E.G., et al., Clinical and microbiological effects of initial periodontal therapy in conjunction with amoxicillin and clavulanic acid in patients with adult periodontitis. Journal of clinical periodontology, 1999. 26(7): p. 461-468. Socransky, S.S., et al., Ecological considerations in the treatment of Actinobacillus actinomycetemcomitans and Porphyromonas gingivalis periodontal infections. Periodontology 2000, 1999. 20(1): p. 341-362. Shibutani, T., et al., Long-term follow-up of periodontitis in a patient with Chediak-Higashi syndrome. A case report. Journal of periodontology, 2000. 71(6): p. 1024-1028. Winkelstein, J.A., et al., Chronic granulomatous disease: report on a national registry of 368 patients. Medicine, 2000. 79(3): p. 155-169. Stasia, M.J. and X.J. Li. Genetics and immunopathology of chronic granulomatous disease. in Seminars in immunopathology. 2008: Springer. Giannopoulou, C., K.-H. Krause, and M. F. The NADPH oxidase NOX2 plays a role in periodontal pathologies. in Seminars in immunopathology. 2008: Springer. Smith, J. and A. Finn, Antimicrobial prophylaxis. Archives of disease in childhood, 1999. 80(4): p. 388392. Mouy, R., et al., Long-term itraconazole prophylaxis against Aspergillus infections in thirty-two patients with chronic granulomatous disease. The Journal of pediatrics, 1994. 125(6): p. 998-1003. Ezekowitz, R.A.B., et al., Partial correction of the phagocyte defect in patients with X-linked chronic granulomatous disease by subcutaneous interferon gamma. New England Journal of Medicine, 1988. 319(3): p. 146-151. Calvino, M.C., et al., Bone marrow transplantation in chronic granulomatous disease. European journal of pediatrics, 1996. 155(10): p. 877-879. Charon, J.A., S.E. Mergenhagen, and J.I. Gallin, Gingivitis and oral ulceration in patients with neutrophil dysfunction. Journal of Oral Pathology & Medicine, 1985. 14(2): p. 150-155. Cohen, M.S., P.A. Leong, and D.M. Simpson, Phagocytic Cells in Periodontal Defense: Periodontal 81. 82. 83. 84. 85. 86. 87. 88. 89. 90. 91. 92. 93. 94. 95. 96. 97. Status of Patients with Chronic Granulomatous Disease of Childhood*. Journal of periodontology, 1985. 56(10): p. 611-617. Wolf, J.E. and L.K. Ebel, Chronic granulomatous disease: report of case and review of the literature. The Journal of the American Dental Association, 1978. 96(2): p. 292-295. Landing, B.H. and H.S. Shirkey, A syndrome of recurrent infection and infiltration of viscera by pigmented lipid histiocytes. Pediatrics, 1957. 20(3): p. 431-438. De Ravin, S.S., et al., Chronic granulomatous disease as a risk factor for autoimmune disease. Journal of Allergy and Clinical Immunology, 2008. 122(6): p. 1097-1103. Scully, C., Orofacial manifestations of chronic granulomatous disease of childhood. Oral Surgery, Oral Medicine, Oral Pathology, 1981. 51(2): p. 148151. Movahedi, M., et al., Gastrointestinal manifestations of patients with chronic granulomatous disease. Iranian Journal of Allergy, Asthma and Immunology, 2004. 3(2): p. 83-88. Dusi, S., et al., Chronic granulomatous disease in an adult female with granulomatous cheilitis. Acta haematologica, 1990. 84(1): p. 49-56. Miller, R., C.M. Myer, and S. Gray, Otolaryngologic manifestations of chronic granulomatous disease. American journal of otolaryngology, 1988. 9(2): p. 7982. Buduneli, N., et al., Prepubertal periodontitis associated with chronic granulomatous disease. Journal of clinical periodontology, 2001. 28(6): p. 589-593. Hasui, T.H.E., Chronic granulomatous disease in Japan: incidence and natural history. Pediatrics international, 1999. 41(5): p. 589-593. Ahlin, A., et al., Prevalence, genetics and clinical presentation of chronic granulomatous disease in Sweden. Acta Paediatrica, 1995. 84(12): p. 1386-1394. Beertsen, W., et al., Impaired phagosomal maturation in neutrophils leads to periodontitis in lysosomalassociated membrane protein-2 knockout mice. The journal of immunology, 2008. 180(1): p. 475-482. Kinane, D.F. and P. Mark Bartold, Clinical relevance of the host responses of periodontitis. Periodontology 2000, 2007. 43(1): p. 278-293. Jirapongsananuruk, O., et al., Diagnostic paradigm for evaluation of male patients with chronic granulomatous disease, based on the dihydrorhodamine 123 assay. Journal of Allergy and Clinical Immunology, 2003. 111(2): p. 374-379. Baehner, R.L. and D.G. Nathan, Quantitative nitroblue tetrazolium test in chronic granulomatous disease. New England Journal of Medicine, 1968. 278(18): p. 971976. Heyworth, P.G., A.R. Cross, and J.T. Curnutte, Chronic granulomatous disease. Current opinion in immunology, 2003. 15(5): p. 578-584. Grimbacher, B., et al., Hyper-IgE syndrome with recurrent infections--an autosomal dominant multisystem disorder. New England Journal of Medicine, 1999. 340(9): p. 692-702. Renner, E.D., et al., Autosomal recessive hyperimmunoglobulin E syndrome: a distinct disease entity. The Journal of pediatrics, 2004. 144(1): p. 9399. International Dental Journal of Student’s Research, April - June 2015;3(2):93-104 {103} 98. 99. 100. 101. 102. 103. 104. 105. 106. 107. 108. 109. 110. 111. 112. 113. 114. 115. Grimbacher, B., B.H. Belohradsky, and S.M. Holland, Immunoglobulin E in primary immunodeficiency diseases. Allergy, 2002. 57(11): p. 995-1007. Borges, W.G., et al., The face of Job. The Journal of pediatrics, 1998. 133(2): p. 303-305. Engelhardt, K.R., et al., Large deletions and point mutations involving the dedicator of cytokinesis 8 (DOCK8) in the autosomal-recessive form of hyperIgE syndrome. Journal of Allergy and Clinical Immunology, 2009. 124(6): p. 1289-1302. e4. Tsang, P., et al., Severe periodontitis in a 5-year-old girl with hyperimmunoglobulin E syndrome. Pediatric dentistry, 2005. 27(1): p. 68-73. Woellner, C., et al., Mutations in STAT3 and diagnostic guidelines for hyper-IgE syndrome. Journal of Allergy and Clinical Immunology. 125(2): p. 424432. e8. AC O'Connell, A.C., et al., Delayed eruption of permanent teeth in hyperimmunoglobulinemia E recurrent infection syndrome. Oral Surgery, Oral Medicine, Oral Pathology, Oral Radiology, and Endodontology, 2000. 89(2): p. 177-185. Grimbacher, B., S.M. Holland, and J.M. Puck, HyperIgE syndromes. Immunological reviews, 2005. 203(1): p. 244-250. Grimbacher, B., et al., Genetic linkage of hyper-IgE syndrome to chromosome 4. The American Journal of Human Genetics, 1999. 65(3): p. 735-744. Cunningham-Rundles, C., How I treat common variable immune deficiency. Blood. Salzer, U., et al., Mutations in TNFRSF13B encoding TACI are associated with common variable immunodeficiency in humans. Nature genetics, 2005. 37(8): p. 820-828. Salzer, U., S. Unger, and K. Warnatz, Common variable immunodeficiency (CVID): exploring the multiple dimensions of a heterogeneous disease. Annals of the New York Academy of Sciences. 1250(1): p. 41-49. Batista, E.L., et al., Necrotizing ulcerative periodontitis associated with severe congenital immunodeficiency in aprepubescent subject: clinical findings and response to intravenous immunoglobulin treatment. Journal of clinical periodontology, 1999. 26(8): p. 499-504. Dalla Torre, D., et al., Necrotizing Periodontitis as a possible manifestation of common variable immunodeficiency. International journal of oral and maxillofacial surgery. 41(12): p. 1546-1549. Vukas, E., et al., Common variable immunodeficiency case report. Journal of Health Sciences. 3(2): p. 170172. Adurogbangba, M.I., et al., Oro-facial lesions and CD4 counts associated with HIV/AIDS in an adult population in Oyo State, Nigeria. Oral diseases, 2004. 10(6): p. 319-326. Patton, L.L., Sensitivity, specificity, and positive predictive value of oral opportunistic infections in adults with HIV/AIDS as markers of immune suppression and viral burden. Oral Surgery, Oral Medicine, Oral Pathology, Oral Radiology, and Endodontology, 2000. 90(2): p. 182-188. Pakfetrat, A., et al., Oral Manifestations of Human Immunodeficiency Virus-Infected Patients. Iranian journal of otorhinolaryngology. 27(78): p. 43. Glick, M., et al., Oral manifestations associated with HIV-related disease as markers for immune suppression and AIDS. Oral Surgery, Oral Medicine, Oral Pathology, 1994. 77(4): p. 344-349. 116. 117. 118. 119. 120. 121. 122. 123. 124. 125. 126. 127. 128. 129. 130. 131. 132. 133. 134. 135. Margiotta, V., et al., HIV infection: oral lesions, CD4+ cell count and viral load in an ltalian study population. Journal of Oral Pathology & Medicine, 1999. 28(4): p. 173-177. Patton, L.L., et al., Oral manifestations of HIV in a southeast USA population. Oral diseases, 1998. 4(3): p. 164-169. Patton, L.L., et al., Changing prevalence of oral manifestations of human immuno-deficiency virus in the era of protease inhibitor therapy. Oral Surgery, Oral Medicine, Oral Pathology, Oral Radiology, and Endodontology, 2000. 89(3): p. 299-304. Arendorf, T.M., et al., Oral manifestations of HIV infection in 600 South African patients. Journal of Oral Pathology & Medicine, 1998. 27(4): p. 176-179. Bodhade, A.S., S.M. Ganvir, and V.K. Hazarey, Oral manifestations of HIV infection and their correlation with CD4 count. Journal of oral science. 53(2): p. 203211. Moniaci, D., et al., Epidemiology, clinical features and prognostic value of HIV-1 related oral lesions. Journal of Oral Pathology & Medicine, 1990. 19(10): p. 477481. Nittayananta, W. and S. Chungpanich, Oral lesions in a group of Thai people with AIDS. Oral diseases, 1997. 3(S1): p. S41-S45. Laskaris, G., M. Hadjivassiliou, and J. Stratigos, Oral signs and symptoms in 160 Greek HIV-infected patients. Journal of Oral Pathology & Medicine, 1992. 21(3): p. 120-123. Reznik, D.A., Oral manifestations of HIV disease. Topics in HIV medicine: a publication of the International AIDS Society, USA, 2004. 13(5): p. 143148. Patton, L.L., Oral lesions associated with human immunodeficiency virus disease. Dental clinics of North America. 57(4): p. 673-698. Demirer, S., et al., Gingival hyperplasia as an early diagnostic oral manifestation in acute monocytic leukemia: a case report. European journal of dentistry, 2007. 1(2): p. 111. Lim, H.-C. and C.-S. Kim, Oral signs of acute leukemia for early detection. Journal of periodontal & implant science. 44(6): p. 293-299. Bennett, J.M., et al., Proposed revised criteria for the classification of acute myeloid leukemia: a report of the French-American-British Cooperative Group. Annals of internal medicine, 1985. 103(4): p. 620-625. Williams, W.J., et al., Haematology. 1983, New York: McGraw-Hill. Little, J.W., Dental management of the medically compromised patient. 1997: Mosby. Orbak, R. and Z. Orbak, Oral condition of patients with leukemia and lymphoma. The Journal of Nihon University School of Dentistry, 1997. 39(2): p. 67-70. Barrett, A.P., Gingival Lesions in Leukemia: A Classification*. Journal of periodontology, 1984. 55(10): p. 585-588. Fatahzadeh, M. and A.M. Krakow, Manifestation of acute monocytic leukemia in the oral cavity: a case report. Special Care in Dentistry, 2008. 28(5): p. 190194. Reenesh, M., S. Munishwar, and S.K. Rath, Generalised leukaemic gingival enlargement: a case report. Journal of oral & maxillofacial research. 3(3). Wu, J., J.E. Fantasia, and R. Kaplan, Oral manifestations of acute myelomonocytic leukemia: a case report and review of the classification of International Dental Journal of Student’s Research, April - June 2015;3(2):93-104 {104} 136. 137. 138. 139. 140. leukemias. Journal of periodontology, 2002. 73(6): p. 664-668. Bergmann, O.J., H.P. Philipsen, and J. Ellegaard, Isolated gingival relapse in acute myeloid leukaemia. European journal of haematology, 1988. 40(5): p. 473476. Anil, S., et al., Gingival enlargement as a diagnostic indicator in leukaemia. Case report. Australian dental journal, 1996. 41(4): p. 235-237. Hou, G.-L. and C.-C. Tsai, Primary gingival enlargement as a diagnostic indicator in acute myelomonocytic leukemia: a case report. Journal of periodontology, 1988. 59(12): p. 852-855. Long, R.G., L. Hlousek, and J.L. Doyle, Oral manifestations of systemic diseases. The Mount Sinai journal of medicine, New York, 1997. 65(5-6): p. 309315. Felix, D.E. and J. Lukens, Oral symptoms as a chief sign of acute monoblastic leukemia: report of case. 141. Journal of the American Dental Association (1939), 1986. 113(6): p. 899-900. Stoopler, E.T., D.T. Vogl, and E.A. Stadtmauer, Medical management update: multiple myeloma. Oral Surgery, Oral Medicine, Oral Pathology, Oral Radiology, and Endodontology, 2007. 103(5): p. 599609. International Dental Journal of Student’s Research, April - June 2015;3(2):93-104 {105} CLINICAL CASE REPORT Autogenous Connective Tissue Graft for the Treatment of Localized Gingival Recession: A Case Report Dr Zohaib Akram1, Dr Haroon Rashid2, Dr Fahim Vohra3 1 BDS, Department of Restorative Dentistry, University of Malaya, Kuala Lumpur, Malaysia. 2 MDSc, BDS, Faculty, Department of Prosthodontics, College Of Dentistry, Ziauddin University, Karachi Pakistan; 3 MRDRCS, M Clin Dent, MFDS, BDS, Department of Prosthetic Dental Science (SDS), College of Dentistry, King Saud University, Riyadh, Saudi Arabia Corresponding Author: Dr. Fahim Vohra Email: [email protected] Access this Article Online www.idjsr.com Use the QR Code scanner to access this article online in our database Article Code: IDJSR SE 0173 Quick Response Code Introduction Gingival recession is defined as the apical migration of the gingival margin in relation to the cementoenamel junction causing root exposure. The negative effects of exposed roots may lead to dentine hypersensitivity, root caries, and poor esthetics(1). One of the reasons of mucogingival surgery is to gain root coverage with procedures that warrant sound predictability and good esthetics. There are several periodontal plastic surgery proceduresto cover exposed root surfaces which include pedicle grafts (2), free gingival grafts (3), connective tissue grafts (4, 5), membrane barrier guided tissue regeneration technique (6), and acellular dermal matrix allografts (7, 8). However, the predictability of such surgical procedures may be associated with different conditions. Out of number of procedures, subepithelial connective tissue graft can be considered as the ‘gold standard’ technique for treating teeth with gingival recessions (9). A number of systematic reviews (9-11)have validated the use of connective tissue graft (CTG) as the most suitable procedure for Millers Class I and II gingival recession lesions. Besides improving clinical attachment, probing depths, thickness of keratinized mucosa and root coverage, these procedures offer better uniformity of color between the surgical grafted area and adjacent tissues Therefore, this manuscript presents a case report detailing the successful use of autogenous CTG in the management of a Class II gingival recession lesion. Case Report Clinical case presentation A 28-year-old male with no medical problems was referred to the Department of Periodontology with complain of an un-aesthetic mandibular incisor tooth (tooth #41) with sensitivity to hot and cold stimulus from approximately 12 months. Clinical evaluation revealed gingival recession on the labial surface extending 2 mm apical to the cemento-enamel junction (CEJ) and narrow zone of attached gingiva measuring approximately 1 mm (Fig 1). There was no loss of interdental papillary height on the distal aspect of the incisor and mild loss of papilla on the mesial aspect. Plaque control and oral hygiene was good with no apparent staining on the teeth. There was no evidence of interdental bone loss (i.e. the distance between the crestal bone and CEJ was not greater than 2 mm). The case was diagnosed to be sensitivity associated with Class II Miller recession. The goal of the treatment was to restore harmonious appearance of the gingiva by covering the root surface to the height similar to the adjacent tooth and to increase the zone of attached gingiva. Figure 1: Intraoral image showing gingival recession on the labial surface extending 2 mm apical to the CEJ and a narrow zone of attached gingiva. International Dental Journal of Student’s Research, April - June 2015;3(2):105-108 {106} Autogenous Connective Tissue Graft (ACTG) Following local anesthesia with 2% lidocaine, epinephrine 1:100,000, the exposed root surface was thoroughly planed and scaled first with ultrasonic instrument and then manually with the use of hand instruments to remove plaque, accretions and root surface irregularities. The exposed root surface was then conditioned with a saturated solution of tetracycline-HCL for 2 minutes (100mg tetracyclineHCL/1 ml of sterile distilled water). A sulcular incision was made at both sides through the bottom of the crevice allowing dissection of the papillae adjacent to the site of recession defect until the proximal line angles of the adjacent teeth. Afterwards two vertical releasing incisions were placed both mesial and distal involving adjacent teeth, distant from the main defect. A full thickness flap providing a broader surgical bed was elevated in an apical direction exposing the alveolar plate of bone until the mucogingival junction (MGJ). The periosteum was released and blunt dissection into the vestibular lining mucosa was performed to eliminate tension to help re-position the flap coronal at the level of CEJ. The interdental papilla of the adjacent teeth were not involved (Fig 2). The donor site for the sub-epithelial connective tissue graft was the palate in the bicuspid region of the same subject (Fig 3). Donor palatal tissue was harvested in the following way: a horizontal incision was placed in the palate 2 to 3 mm from the free gingival margin, and two parallel internal vertical incisions, one superficial and one deep, were made and connected mesially and distally. The underlying connective tissue was released at its base and removed (Fig 4). The wound was closed with simple interrupted 3-0 silk sutures. The donor site on the palate healed by primary intention after two week of suture removal. The graft was shaped to fit the recipient site and secured to the wound bed (Fig 5) with a continuous sling suture using 5-0 vicryl material to the papilla on either side of the graft. Silk sutures were removed after 15 days; visible portions of the vicryl suture were removed after 3 weeks. Figure 2: Full thickness flap elevated exposing the alveolar plate of bone. The interdental papilla of the adjacent teeth were not involved. Figure 3: Subepithelial connective tissue graft harvested from the palate of the same subject. Figure 4: Resected palatal subepithelial connective tissue graft Figure 5: Graft secured to the papilla on either side using continuous sling suture 5-0 vicryl. International Dental Journal of Student’s Research, April - June 2015;3(2):105-108 {107} Postsurgical Care Patient was instructed to avoid trauma and to discontinue tooth brushing at the surgical site during the first 15 days. Patient was instructed to use 0.12% chlorhexidinedigluconate solution rinse for 60 seconds twice daily for 2 weeks. After 15 days, a modified brushing technique was advised in order to minimize apically directed trauma to the soft tissue around the surgical site. Throughout the treatment, recall visits for prophylaxis treatment were arranged at 1, 3, 5, 8, 12, 16 and 32 weeks. Healing was uneventful. At 2nd week, the gingiva at the surgical site was still edematous (Fig 6). Only erythema could be observed along the border of attached gingiva which improved at 8th week of follow-up. At 9 months postoperatively, the amount of attached gingiva was approximately 3 mm, and the gingiva was firmly attached. Probing depth at the mid buccal site was less than 1 mm and the free gingival margin was located less than 1 mm apically to the apical border of the CEJ (Fig 7). Figure 6: 2nd week postoperative. Gingiva is still edematous and erythema could be observed along the border of the attached gingiva. Figure 7: At 2 months postoperatively the periodontal tissue is less edematous and improvement in erythema is clinically evident. Figure 8: At 9 months the probing depth at mid buccal site was 1 mm and free gingival margin was located 1 mm apical to CEJ. Discussion This case report evaluated the treatment of localized gingival recession by using palatal connective tissue graft for the treatment of Class II Miller recession. The present clinical result is encouraging and indicates significant coverage of the exposed root with the palatal connective tissue graft that has provided restoration of clinical attachment. Overall surgical procedure was aimed to reduce any risks involved with no harmful events in the healing process along with patient comfort. The connective tissue graft for the restoration of root defect was harvested from the palate of the same subject. Horizontal incisions were placed in the palatal tissue with two parallel vertical incisions along each side of the horizontal incision to remove adequate tissue from the underlying connective tissue. The incisions were placed to ensure primary intention healing and comfort for the patient. Multiple factors can effect the degree of root coverage including, biocompatibility of root surface, sufficient vascularization of the surgical bed, surgical manipulation, tissue width and ideal plaque control (12). Meticulous root planing on the exposed root surface was performed with the use of hand instruments to remove plaque and accretions and further increasing the surface biocompatibility. Special care was taken to prepare recipient surgical bed. Sutures were performed without stretching the graft tissue, preventing the displacement of graft without tension, thereby avoiding impaired vascularization. The grafts were also compressed to promote the tensile strength and stability of the wound. Coronally advanced flaps (CAF) with or without enamel matrix derivatives have been recommended as an alternative to CTG in the management of Class I & II recession lesions. Nemcovsky et al, (13) compared the clinical outcome of CAF and CTG in the management of recession defects, concluding that CTG was superior to CAF in the percentage of coverage and increase in width of keratinized tissue. International Dental Journal of Student’s Research, April - June 2015;3(2):105-108 {108} In addition, soft tissue allografts have been used as an alternate to autogenous CTG to provide root coverage without the need of a second surgical site intra-orally (14).However CTG additional increases the gingival tissue thickness and width of keratinized tissue, two critical features, which warrant the use of connective tissue graft over allografts (15). Conclusion This surgical technique aided complete root coverage as well as improved the thickness of attached gingiva. The interpretations made in the present case report indicate that connective tissue graft can be a successful treatment option in achieving soft tissue root coverage and gain of clinical attachment in Miller’s class II root defects. systematic review. Annals of Periodontology 2003; 8: 303-320. 12. Burkhardt R, Lang NP. Coverage of localized gingival recessions: comparison of micro‐and macrosurgical techniques. Journal of clinical periodontology 2005; 32: 287-293. 13. Nemcovsky CE, Artzi Z, Tal H, Kozlovsky A, Moses O.A multicenter comparative study of two root coverage procedures: coronally advanced flap with addition of enamel matrix proteins and subpedicle connective tissue graft. J Periodontol 2004;75:600-7. 14. Saadoun AP. Root coverage with Emdogain/Alloderm: a new way to treat gingival recessions. Eur J Esthet Dent 2008 ;3:46. 15. Silva, R.C.D., et al., Root coverage using the coronally positioned flap with or without a subepithelial connective tissue graft. J Periodontol 2004;75:413-19. __________________________________________________ References 1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. Bouchard P, Malet J, Borghetti A. Decision‐making in aesthetics: root coverage revisited. Periodontology 2000 2001; 27: 97-120. Harris RJ, Harris AW. The coronally positioned pedicle graft with inlaid margins: a predictable method of obtaining root coverage of shallow defects. The International journal of periodontics & restorative dentistry 1994; 14: 228-241. Miller Jr PD. Root coverage with the free gingival graft: factors associated with incomplete coverage. Journal of Periodontology 1987; 58: 674-681. Langer B, Langer L. Subepithelial connective tissue graft technique for root coverage. Journal of periodontology 1985; 56: 715-720. Bruno JF. Connective tissue graft technique assuring wide root coverage. The International journal of periodontics & restorative dentistry 1994; 14: 126-137. Müller H-P, Stahl M, Eger T. Root coverage employing an envelope technique or guided tissue regeneration with a bioabsorbable membrane. Journal of periodontology 1999; 70: 743-751. Tal H, Moses O, Zohar R, Meir H, Nemcovsky C. Root coverage of advanced gingival recession: A comparative study between acellular dermal matrix allograft and subepithelial connective tissue grafts. Journal of periodontology 2002; 73: 1405-1411. Harris RJ. Root Coverage With a Connective Tissue With Partial Thickness Double Pedicle Graft and an Acellular Dermal Matrix Graft: A Clinical and Histological Evaluation of a Case Report*. Journal of periodontology 1998; 69: 1305-1311. Chambrone L, Chambrone D, Pustiglioni FE, Chambrone LA, Lima LA. Can subepithelial connective tissue grafts be considered the gold standard procedure in the treatment of Miller Class I and II recession-type defects? Journal of dentistry 2008; 36: 659-671. Roccuzzo M, Bunino M, Needleman I, Sanz M. Periodontal plastic surgery for treatment of localized gingival recessions: a systematic review. Journal of Clinical Periodontology 2002; 29: 178-194. Oates TW, Robinson M, Gunsolley JC. Surgical therapies for the treatment of gingival recession. A International Dental Journal of Student’s Research, April - June 2015;3(2):105-108