thesis - KI Open Archive

Transcription

thesis - KI Open Archive
From The Department of Neurobiology, Care Sciences and Society
Karolinska Institutet, Stockholm, Sweden
THE N-METHYL-D-ASPARTATE RECEPTOR SUBUNIT
NR3A: CLONING, EXPRESSION AND INTERACTING
PROTEINS
Maria Eriksson
Stockholm 2008
All previously published papers were reproduced with the permission from the publisher.
Cover: β-tubulin III immunoreactivity in a hippocampal neuron from mouse brain.
Printed by Larserics Digital Print AB
© Maria Eriksson, 2008
ISBN 978-91-7357-554-6
Sluta stressa, du dör ändå.
Ovanligt klok kvinna
ABSTRACT
This thesis focuses on the subunit NR3A, which together with subunits NR1, NR2A-NR2D and
NR3B builds the excitatory glutamate receptor N-methyl-D-aspartate (NMDA). The receptor is
most likely a tetramer, containing at least one NR1, probably two NR2 and sometimes one NR3
subunit. NR3A is highly expressed during development and present at moderate levels in the
adult brain. Both NR3A and NR3B have an attenuating effect on NMDA receptor currents,
when incorporated into the receptor complex.
In study I we cloned and sequenced the human NR3A subunit, which at the time only had been
described in rat. We found the homology between the rat and human sequences to be high, and
most potential phosphorylation and glycosylation sites to be conserved. In contrast to rat tissue,
we did not detect the longer splice variant of NR3A in the human central nervous system. We
found NR3A mRNA to be expressed both in the developing human brain and spinal cord, with
prominent staining of the cortical plate, ventricular zone and the dorsal half of the spinal cord as
well as the neuroepithelial layer surrounding the central canal. Interestingly, the neuroepithelial
layer expressed very low levels of NR1 mRNA, suggesting that NR3A might have replaced one
of the NR1 subunits in NMDA receptors in this area.
In study II we investigated the occurrence of NR3A mRNA and protein in adult human brain,
its association with the other NMDA receptor subunits and the subunits solubility. We found
NR3A mRNA and protein to be expressed in adult human brain, especially in layers II/III and
V of cerebral cortex and in thalamus and pons. Expression was low or undetectable in caudate
nucleus, claustrum, cerebellum and spinal cord. Further we found NR3A to be associated with
NR1, NR2A and NR2B in adult human cortex, and to some extent in the spinal cord. NR3A
showed a different solubility profile to the other NMDA receptor subunits, being extracted with
milder detergents. Both NR1 and NR3A were found as monomers, dimers and tetramers, as
well as in larger protein complexes. In contrast, NR2 subunits were only found in tetramers and
in larger protein complexes. This probably reflects the presence of an intracellular pool of
unassembled NR1 and NR3A subunits.
To learn more about NR3A and its function we screened a fetal human brain cDNA library for
proteins interacting with NR3A. Among a number of potentially interesting candidates we
choose MAP1S/C19ORF5 and MAP1B for further analysis. The results are presented in study
III and IV respectively. We found MAP1S to be highly expressed throughout brain and spinal
cord, predominantly in neurons. MAP1S-EGFP over-expressed in cultured hippocampal cells,
localized both to dendritic shafts and filopodia. Colocalization of MAP1S-EGFP with β-tubulin
III immunoreactivity was prominent, with synapsin and PSD95 immunoreactivity occasional
and with NR3A immunoreactivity frequent in dendritic shafts and sparse in filopodia. Judging
from the subcellular distribution of MAP1S and NR3A their interaction might be important for
transport through shafts or regulation of NR3A-containing receptors in spines.
The distribution of MAP1B immunoreactivity resembled that of MAP1S-EGFP, with
prominent overlap with β-tubulin III immunoreactivity, sparse colocalization with synapsin and
SAP102 immunoreactivity, and frequent colocalization with NR3A immunoreactivity in
dendritic shafts and infrequent colocalization in spines. To address the function of the NR3AMAP1B interaction, we investigated the expression and distribution of NR3A in MAP1B
deficient mice. These mice expressed increased NR3A and decreased NR1 levels compared to
wild type, suggesting that NMDA receptors in the MAP1B deficient mice might have an altered
subunit composition. NR3A was equally distributed to filopodia in neurons from MAP1B
deficient and wild type mice, indicating that MAP1B is not essential for transport of NR3Acontaining NMDA receptors to synaptic sites. Instead the interaction might involve regulating
the distribution of NR3A-containing receptors between intracellular pools and the cell surface,
as well as the distribution between synaptic and extrasynaptic sites.
LIST OF PUBLICATIONS
The thesis is based on the following articles, which will be referred to in the text by
their roman numerals.
I. Maria Eriksson, Anna Nilsson, Susanne Froelich-Fabre, Elisabet Åkesson,
Jenny Dunker, Åke Seiger, Ronnie Folkesson, Eirikur Benedikz, and Erik
Sundström. Cloning and expression of the human N-methyl-D-aspartate
receptor subunit NR3A. Neurosci. Lett. (2002) 321; 177-181.
II. Anna Nilsson*, Maria Eriksson*, Chris Muly, Elisabet Åkesson, Eva-Britt
Samuelsson, Nenad Bogdanovic, Eirikur Benedikz and Erik Sundström.
Analysis of NR3A receptor subunits in human native NMDA receptors. Brain
Res. (2007) 1186; 102-112.
III. Maria Eriksson, Helena Samuelsson, Eva-Britt Samuelsson, Leyuan Liu,
Wallace L. McKeehan, Eirikur Benedikz and Erik Sundström. The NMDAR
subunit NR3A interacts with microtubule-associated protein 1S in the brain.
Biochem. Biophys. Res. Commun. (2007) 361;127-132.
IV. Maria Eriksson, Helena Samuelsson, Stefan Björklund, Elena Tortosa, Jesus
Avila, Eirikur Benedikz and Erik Sundström. The N-methyl-D-aspartate
receptor subunit NR3A interacts with microtubule-associated proteins in the
mammalian brain. Submitted manuscript.
*These authors have contributed equally to the work.
TABLE OF CONTENTS
LIST
OF
ABBREVIATIONS
SAMMANFATTNING
PÅ
SVENSKA .................................................................................................. 1
NERVSYSTEMET
OCH
NERVCELLEN .......................................................................................................................1
N‐METHYL‐D‐ASPARTAT
(NMDA)
RECEPTORN ..............................................................................................2
AVHANDLINGENS
DELARBETEN .............................................................................................................................3
SLUTSATS ....................................................................................................................................................................5
INTRODUCTION .................................................................................................................................... 6
THE
NMDA
RECEPTOR ...........................................................................................................................................7
Identification
of
the
NMDA
receptor
subunits ...................................................................................... 7
NMDA
receptor
activation.............................................................................................................................. 9
THE
NR3A
SUBUNIT .............................................................................................................................................11
Spatial
and
temporal
distribution
of
NR3A..........................................................................................11
NR3A
knock
out
mice.......................................................................................................................................11
The
atypical
NR1/NR3
glycine
receptor ................................................................................................12
NR3A
in
glial
cells..............................................................................................................................................13
NR3A
and
schizophrenia ...............................................................................................................................14
THE
POSTSYNAPTIC
DENSITY ..............................................................................................................................15
The
NMDA
receptor
complex.......................................................................................................................16
Proteins
interacting
with
NR3A .................................................................................................................19
SUBUNIT
ASSEMBLY
AND
INTRACELLULAR
TRANSPORT ...............................................................................21
Assembly
of
the
NMDA
receptor
subunits .............................................................................................21
The
exocyst
complex.........................................................................................................................................21
Transport
along
microtubules....................................................................................................................22
Receptor
trafficking
at
synapses................................................................................................................23
MICROTUBULE‐ASSOCIATED
PROTEINS ............................................................................................................24
The
MAP1
family................................................................................................................................................24
MAP1S/C19ORF5...............................................................................................................................................25
MAP1B ....................................................................................................................................................................26
AIMS
OF
THE
THESIS.........................................................................................................................30
GENERAL
AIM .........................................................................................................................................................30
SPECIFIC
AIMS .........................................................................................................................................................30
RESULTS
AND
DISCUSSION.............................................................................................................31
STUDY
I,
CLONING..................................................................................................................................................31
STUDY
II,
MAPPING ...............................................................................................................................................33
STUDY
III,
INTERACTION
WITH
MAP1S/C19ORF5....................................................................................35
STUDY
IV,
INTERACTION
WITH
MAP1B..........................................................................................................37
CONCLUSIONS
AND
FUTURE
PERSPECTIVES ...........................................................................39
MATERIALS
AND
METHODS .......................................................................................................... 41
HUMAN
TISSUE
(PAPERS
I,
II,
III
AND
IV) .......................................................................................................41
ANIMAL
TISSUE
(PAPERS
I,
II,
III
AND
IV) ......................................................................................................41
HUMAN
BRAIN
CDNA
LIBRARIES
(PAPER
I)....................................................................................................41
REVERSE
TRANSCRIPTION‐POLYMERASE
CHAIN
REACTION
(RT‐PCR)
(PAPERS
I
AND
II) .................42
SUBCLONING,
MUTAGENESIS
AND
SEQUENCING
(PAPERS
I,
III
AND
IV) ..................................................42
IN
SITU
HYBRIDIZATION
(PAPERS
I
AND
II) .....................................................................................................42
IMMUNOHISTOCHEMISTRY
(PAPER
II) .............................................................................................................42
SOLUBILIZATION
OF
RECEPTOR
SUBUNITS
(PAPER
II)..................................................................................42
SIZE
EXCLUSION
CHROMATOGRAPHY
AND
SPOT
BLOT
(PAPER
II) .............................................................43
PREPARATION
OF
TISSUE
AND
WESTERN
BLOT
(PAPERS
II,
III
AND
IV)..................................................43
CO‐IMMUNOPRECIPITATION
(PAPERS
II,
III
AND
IV)...................................................................................43
YEAST
TWO‐HYBRID
SCREEN
(PAPERS
III
AND
IV) .......................................................................................43
GST
PULL‐DOWN
(PAPERS
III
AND
IV)............................................................................................................44
CELL
CULTURES,
TRANSFECTION
AND
IMMUNOCYTOCHEMISTRY
(PAPERS
III
AND
IV)........................44
STABILIZATION
OF
MICROTUBULES
(PAPER
IV).............................................................................................44
ACKNOWLEDGEMENTS ................................................................................................................... 46
REFERENCES........................................................................................................................................ 49
LIST OF ABBREVIATIONS
5-HT
AKAP
AMPA
AMPAR
CaMKII
cAMP
CARP-1
C19ORF5
cDNA
CHAPSO
CK2
CNS
CV
DAB
DIG
DIV
DNA
Dlg
DOC
EGFP
E
ER
EZ
FMRP
GABA
GABARAP
GAN
GFP
GK
GluR
GPS2
GRIP
GST
HC
HEK293
ICC
IHC
IP3R
ISH
IgG
iGluR
KIF
KO
LC
5-hydroxy tryptamine
A kinase anchoring protein
α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid
α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor
Ca2+ calmodulin-dependent protein kinase II
Cyclic adenosine monophosphate
Cell cycle and apoptosis regulatory protein-1
Chromosome 19 open reading frame 5
Complementary DNA
3-[(3-Cholamidopropyl)dimethylammonio]-2-hydroxy-1-propanesulfonate
Casein kinase 2
Central nervous system
Clathrin-coated vesicle
3,3'-diaminobenzidine
Digoxigenin
Days in vitro
Deoxyribonucleic acid
Discs large
Sodiumdeoxycholate
Enhanced green fluorescent protein
Embryonic day
Endoplasmic reticulum
Endocytic zone
Fragile X mental retardation protein
γ-amino-butyric acid
GABAA receptor-associated protein
Giant axonal neuropathy
Green fluorescent protein
Guanylate kinase
Glutamate receptor subunit
G-protein pathway suppressor 2
Glutamate receptor-interacting protein
Glutathione-S-transferase
Heavy chain
Human embryonic kidney 293
Immunocytochemistry
Immunohistochemistry
Inositol 1,4,5-trisphosphate (IP3) receptor
In situ hybridization
Immunoglobulin G
Ionotropic glutamate receptor
Kinesin superfamily protein
Knock out
Light chain
LRPPRC
MAGUK
mGluR
MAP
MH1-3
mRNA
NMDA
NMDAR
nNOS
NP-40
NR1-3B
P
PCR
PDZ
PKA
PNS
PP1
PP2A
PR
ProSAP
PSD
PSD93
PSD95
RAB1
RNA
RT-PCR
SAM
SAP102
SAP97
SDS-PAGE
SH3
Shank
SER
VCY2IP-1
WT
ZO-1
Leucine-rich PPR-motif containing protein, AAA67549
Membrane-associated guanylate kinase
Metabotropic glutamate receptor
Microtubule-associated protein
MAP1 homology domains 1-3
Messenger RNA
N-methyl-D-aspartate
N-methyl-D-aspartate receptor
Neuronal nitric oxide synthase
Nonidet P-40
NMDA receptor subunit 1-3B
Postnatal day
Polymerase chain reaction
PSD-95/Dlg/ZO-1
cAMP-dependent protein kinase PKA
Peripheral nervous system
Protein phosphatase 1
Protein phosphatase 2A
Polyribosome
Proline-rich synapse-associated protein
Postsynaptic density
Postsynaptic density protein 93
Postsynaptic density protein 95
RAS-associated domain family protein 1A-binding protein 1
Ribonucleic acid
Reverse transcription-polymerase chain reaction
Sterile alpha motif
Synapse-associated protein 102
Synapse-associated protein 97
Sodium dodecyl sulfate-polyacrylamide gel electrophoresis
Src homology 3
SH3 and ankyrin repeat-containing protein
Smooth endoplasmic reticulum
Variable charge Y chromosome 2 interacting protein-1
Wild type
Zona occludens-1
SAMMANFATTNING PÅ SVENSKA
NERVSYSTEMET OCH NERVCELLEN
Vårt nervsystem delas upp i en central del, bestående av hjärna och ryggmärg, och en
perifer del, bestående av nervvävnad belägen utanför de centrala delarna. Den
mänskliga hjärnan består av uppskattningsvis 10 miljarder nervceller och ännu fler
gliaceller. Gliacellerna bildar till exempel blod-hjärn-barriären och de fettrika
myelinskidor som möjliggör att våra elektriska nervimpulser kan vidarebefordras i hög
hastighet längs nervcellens utskott.
Nervcellens cellkropp innehåller bland annat cellkärnan med vårt arvsanlag, DNA och
system för att bilda, bryta ner och transportera proteiner till olika platser inuti och
utanför cellen (figur 1). Från cellkroppen utgår ofta ett förgrenat nätverk av långa
utskott, kallade dendriter. Genom dessa dendriter mottar nervcellen information från ett
stort antal omkringliggande nervceller. En del nervceller kan ta emot information från
upp till 100 000 andra nervceller. Från cellkroppen utgår också ett axon, ett utskott
specialiserat för att transportera nervimpulser vidare till omkringliggande celler.
Förutom andra nervceller skickar nervsystemet signaler till våra muskler och körtlar.
Nervcellen innehåller också ett cellskelett som hjälper den att förändra eller bibehålla
sin struktur. Avhandlingen berör två olika typer av cellskelett, aktin och mikrotubuli.
Figur 1. En nervcell från mushjärna. Cellen är
tagen från regionen hippocampus som bland
annat är viktig för minnesfunktioner. Cellen är
odlad tre veckor i näringslösning och under
den tiden hinner nervcellen mogna och bilda
kontakter med de omkringliggande cellerna.
Till höger syns den runda cellkroppen och
från den sträcker sig de långa utskotten,
dendriterna och axonen. För att synliggöra
cellen har jag färgat den med en antikropp
som känner igen mikrotubuli-cellskelettet.
Nervimpulserna vidarebefordras från en cell till nästa genom att nervcellerna
kommunicerar med varandra i specialiserade kontaktzoner, synapser (figur 2). Det
finns elektriska och kemiska synapser men de kemiska synapserna är vanligast. Vid en
kemisk synaps utsöndrar nervcell 1 molekyler som med hjälp av diffusion rör sig den
korta sträckan till nervcell 2. På nervcell 2 binder molekylerna till mottagarproteiner, så
kallade receptorer och aktiverar därmed dessa. Vårt nervsystem har många olika sorters
receptorer som var och en förmedlar olika signaler till nervcellen de sitter på.
Receptorerna består av tre delar. En del som sitter utanför cellen och kan binda
molekyler utsöndrade från omkringliggande celler. En del som går genom nervcellens
yttervägg, cellmembranet, och en del som sitter inuti cellen och där kan interagera med
flera andra av nervcellens proteiner.
1
Figur 2. En synaptisk kontaktzon mellan två
nervceller. Överst i bilden syns nervcell 1
som avger molekyler från sitt axon.
Molekylerna rör sig genom synapsklyftan och
binder till mottagarproteiner, så kallade
receptorer som sitter i cellmembranet på
nervcell 2. Då molekylerna binder till
receptorerna aktiveras dessa och en rad
signaler befordras till nervcell 2.
N-METHYL-D-ASPARTAT (NMDA) RECEPTORN
Den här avhandlingen beskriver proteinet NR3A som ingår som en enhet i receptorn
NMDA (figur 3). NMDA-receptorn är utbredd i centrala nervsystemet hos däggdjur
och är bland annat inblandad i minnesfunktioner och vissa typer av celldöd. NMDAreceptorn aktiveras av att molekylerna glutamat och glycin binder till den del av
receptorn som sticker ut utanför cellen. Receptorn anpassar sig till bindningen genom
att ändra form, vilket leder till att en kanal öppnas genom receptorn och jonerna Na+,
K+ och Ca2+ kan passera från ena sidan av cellmembranet till den andra. Förändringen
av receptorns form samt jonströmmarna över cellmembranet ger en rad effekter på
nervcellen. Dels kan proteinerna som binder till receptorns inre delar påverkas, vilket
kan leda till en kaskad av händelser inuti cellen. Dels kan cellens benägenhet att skicka
vidare nervimpulser till omkringliggande celler påverkas. Mitt arbete har främst berört
de delar av NR3A som finns inuti cellen och de proteiner dessa delar kan interagera
med.
Figur 3. NMDA-receptorn och dess
enheter NR1, NR2 och NR3A.
Enheterna binder till varandra i
grupper om fyra och bilder på så sätt
en
funktionell
NMDA-receptor.
Receptorn aktiveras av att molekylerna
glutamat och glycin binder till den och
aktiveringen kan i vissa fall leda till att
nya minnen bildas.
2
AVHANDLINGENS DELARBETEN
I arbete I beskriver vi identifieringen av proteinet NR3A i människans centrala
nervsystem. Från mänsklig hjärnvävnad renade vi fram den del av arvsanlaget, DNA,
som innehåller NR3A. Alla levande organismer har DNA i sina celler och det är DNA
som bär vår genetiska kod, våra gener. DNA är uppbyggt av fyra olika beståndsdelar,
baserna A, T, C och G och alla gener har en unik följd av dessa fyra baser. Basernas
ordning avgör i sin tur hur proteinet de kodar för kommer att se ut. Våra proteiner är
uppbyggda av 20 olika aminosyror och deras inbördes ordningen är unik för varje
protein. Vi har här tagit reda på i vilken följd baserna A, T, C och G kommer i NR3Agenen, det vill säga vi har sekvensbestämt DNA. Basernas följd översätts enligt
bestämda regler till aminosyror. Då NR3A-genen sekvenserats kunde vi översätta
basernas följd till aminosyror och därmed var det humana NR3A-proteinet identifierat.
I arbete II undersökte vi i vilka delar av människans centrala nervsystem som NR3A
förekommer och fann att de högsta nivåerna av proteinet finns i hjärnbarken, thalamus
och pons, eller bryggan (figur 4). Hjärnbarken (cortex cerebri) är inblandad i högre
kognitiva funktioner och thalamus vidarebefordrar signaler från kroppen till hjärnan
och tillbaka igen. Pons förbinder bland annat lillhjärnan med resten av nervsystemet. Vi
hittade bevis för att NR3A sitter ihop med de andra komponenterna i NMDAreceptorn, NR1, NR2A och NR2B. Hos råtta har det visat sig att NR3A förekommer i
höga nivåer under tidig utveckling. Här såg vi att NR3A förekommer i betydande
nivåer även i den vuxna hjärnan hos råtta, apa och människa. Den vuxna råttan har
betydande nivåer av NR3A i sin ryggmärg men vi fann att vuxna människor har mycket
lite kvar av proteinet i ryggmärgen. Slutligen undersökte vi hur hårt NR3A sitter
förankrad i nervcellens membran och till andra proteiner jämfört med enheterna NR1,
NR2A och NR2B. Det gjorde vi genom att använda oss av diskmedelliknande
substanser, detergenter, som löser upp de feta cellmembranen och i viss mån
bindningarna mellan proteiner. Vi fann att NR3A var mer lättlösligt än NR1 och NR2A
och NR2B. NR3A förekom också i större utsträckning i en form där det inte var bundet
till de andra enheterna. Det indikerar att förutom att ingå i NMDA-receptorkomplexet
vid nervcellens yta, kan obundet NR3A finnas i en reservpool inuti cellen.
Figur 4. Bilden visar hjärnvävnad där
proteinet NR3A är infärgat. Genom
mikroskopet kan man se de mörka nervceller
som innehåller NR3A. Cellkropparna syns
tydligast och utskotten något. Vävnaden är
tagen från främre delen av hjärnbarken från en
aphjärna. Bilden är tagen av Chris Muly.
Syftet med arbete III och IV var att undersöka vilka proteiner som binder till NR3A
inuti nervcellen. Kartläggning av proteininteraktionerna ger en ökad förståelse av de
signaler NR3A och NMDA-receptorn skickar till mottagarcellen. Kunskapen kan
eventuellt användas för att ta fram läkemedel mot sjukdomar där NR3A och NMDA-
3
receptorn är inblandad. Genom att använda NR3A som bete har vi ”fiskat” i ett
bibliotek som innehåller en stor mängd proteiner från den mänskliga hjärnan. Vår
fångst bestod av både bottennapp och riktigt intressanta proteiner. Av de intressanta
proteinerna valde vi ut två besläktade proteiner att arbeta vidare med, ”microtubuleassociated protein” (MAP) 1S, som beskrivs i arbete III och MAP1B som beskrivs i
arbete IV. Dessa två proteiner har fått sitt namn på grund av att de kan binda till
mikrotubuli-cellskelettet. Efter att vi med metoden ”GST pull-down” verifierat att
proteinerna binder till NR3A i provröret, undersökte vi om proteinerna också binder till
varandra i hjärnan. Med hjälp av en antikroppsbaserad metod, immunoprecipitering,
kunde vi se att både MAP1S och MAP1B binder till NR3A i hjärnan, vilket indikerar
att interaktionen har en funktion! I och med att få studier tidigare publicerats om
proteinet MAP1S undersökte vi i vilka organ och i vilka regioner av hjärnan som
proteinet finns. Vi kunde se att det finns stora mängder MAP1S i hjärnan men att
proteinet även förekommer i andra organ. Arbete III avslutades med att vi med hjälp av
så kallad immunocytokemi undersökte i vilka delar av nervcellen MAP1S-proteinet
finns. I denna metod tillsätts färgade antikroppar till nervceller odlade på glasplattor.
Genom mikroskopering kan man se var de färgade antikropparna har fastnat, det vill
säga den plats i nervcellen där det proteinet som antikroppen specifikt känner igen
finns. Vi använde antikroppar mot både NR3A och MAP1S och kunde se att de till viss
del förekommer i samma del av nervcellen. Genom att använda antikroppar mot två
proteiner (synapsin och PSD95) som finns i nervcellernas synapser, kunde vi se att
MAP1S till viss del förekommer i synapserna. I synapserna finns även NMDAreceptorn och NR3A, vilket kan betyda att MAP1S är ett av proteinerna som förmedlar
signaler i mottagarcellen efter att NMDA-receptorn aktiverats.
I arbete IV undersökte vi interaktionen mellan proteinerna NR3A och MAP1B. Med
hjälp av immunocytokemi såg vi att både NR3A och MAP1B förekommer i stor
utsträckning i nervcellens dendriter men även i synapserna (figur 5). För att ta reda på
funktionen av interaktionen mellan de båda proteinerna använde vi oss av möss som
saknar MAP1B-proteinet. Vi fann att dessa möss hade mer NR3A och mindre NR1 än
kontrollmössen. Det tyder på att dessa möss skulle kunna ha en annan sammansättning
av receptorenheterna i sina NMDA-receptorer än kontrollmössen. I nervceller odlade
från dessa möss och från kontrollmössen såg vi att NR3A förekom i lika stor
utsträckning i synaptiska strukturer hos båda djuren. Det tyder på att MAP1B inte krävs
för att transportera NR3A till synaptiska strukturer. Istället är det mer troligt att
interaktionen mellan MAP1B och NR3A är viktig för hur NR3A rör sig i de synaptiska
strukturerna.
Figur 5. Immunocytokemisk infärgning av en dendrit i en odlad, human nervcell med
antikropp som känner igen MAP1B, till vänster och NR3A till höger. Färgningen visar att båda
proteinerna finns i dendriterna men att NR3A förekommer i större utsträckning i de tunna
utskott där synaptiska kontakter bildas.
4
SLUTSATS
Avhandlingen visar att proteinet NR3A i människa liknar NR3A i råtta, både avseende
aminosyrasammansättning och förekomst i hjärnans olika delar. Däremot skiljer sig
förekomsten av NR3A i ryggmärg hos råtta och människa, då den vuxna människans
ryggmärg nästan helt saknar NR3A. NR3A förekommer i betydande nivåer i vuxen
hjärna hos råtta, apa och människa, vilket tyder på att proteinet spelar en viktigare roll i
den vuxna hjärnan än tidigare trott. Vi visar också att NR3A är associerad med de
andra NMDA-receptorenheterna NR1, NR2A och NR2B i den vuxna hjärnan men
även förekommer obundet. Slutligen har vi identifierat två proteiner som interagerar
med NR3A i hjärnan, MAP1S och MAP1B. Båda proteinerna är vanligt
förekommande i hjärnan och binder till nervcellernas cellskelett. Våra fynd tyder på att
interaktionerna mellan NR3A och MAP1S respektive MAP1B är inblandade i
regleringen av NR3A i nervcellens synapser. Reglering av NMDA receptorn och dess
enheter är viktig för normal mognad av nervceller under hjärnans utveckling och för
minnesfunktioner under hela livet.
5
INTRODUCTION
Our nervous system contains a complex network of neurons and glial cells that
communicate with each other in a highly regulated manner. Nerve impulses are
electrical signals, propagated through the neuron by ions, and from one neuron to the
next usually by chemical substances called neurotransmitters. When the electrical
signal reaches the neurons terminal, neurotransmitters are released from the terminal
into a narrow, extracellular space, the synaptic cleft, and diffuse to the receptors of the
receiving neuron. With respect to their position in the synaptic cleft, neuronal structures
are termed pre- and postsynaptic. The functional unit consisting of the presynaptic
membrane, the synaptic cleft and the postsynaptic membrane is called the synapse. The
amino acid glutamate is the most common excitatory neurotransmitter in the
mammalian central nervous system (CNS), affecting the postsynaptic neuron by
binding to glutamate receptors situated in the postsynaptic plasma membrane. There are
three types of ionotropic glutamate receptors in mammals, N-methyl-D-aspartate
(NMDA), α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) and
kainate receptors. This thesis focuses on the NMDA receptor subtype.
6
THE NMDA RECEPTOR
The NMDA receptor is widely distributed throughout the mammalian CNS and has
been prescribed a vast array of functions, ranging from involvement in normal brain
development and learning to cell death and psychiatric disorders. The receptor is
situated at excitatory synapses on dendritic spines, but is also localized to extrasynaptic
sites. A large number of proteins in the postsynaptic density (PSD) interact with the
NMDA receptor and mediates its signaling upon receptor activation. The receptor is
composed of different subunits, which assemble and form an ionotropic receptor
permeable to positively charged ions. Each individual NMDA receptor subunit has an
extracellular N-terminus, four membrane domains and an intracellular C-terminus
(figure 1). Membrane domains three and four are connected by a long extracellular
loop, which together with the N-terminus form a ligand-binding pocket. Membrane
domains one, three and four span the membrane while domain number two is a reentrant loop, beginning and ending on the intracellular side of the membrane. The
membrane domains are arranged such that the re-entrant loops outline the ion channel
pore.
Figure 1. Topology of the NMDA receptor
subunits. The long N-terminus, which is situated
outside the cell, is followed by four membrane
regions connected by intracellular and
extracellular loops, and an intracellular Cterminus. The N-terminus forms the ligandbinding pocket together with the extracellular
loop between membrane regions III and IV.
Abbreviation; Ligand (L).
Identification of the NMDA receptor subunits
There are three types of NMDA receptor subunits, NR1, NR2 and NR3, and they
assemble into tetramers to form functional receptors. NR1 was the first identified
NMDA receptor subunit, cloned from rat tissue in the early 1990s (Moriyoshi et al.,
1991). Cloning the NR1 subunit from mouse tissue in 1992 and from human tissue in
1993 closely followed the initial identification (Yamazaki et al., 1992; Karp et al.,
1993). The NR2 subunits, NR2A-NR2D, are encoded by four different genes and have
been identified in rat (Monyer et al., 1992), mouse (Ikeda et al., 1992; Kutsuwada et al.,
1992; Meguro et al., 1992) and human tissue (Foldes et al., 1994; Adams et al., 1995;
Lin et al., 1996). As the other NMDA receptor subunits, NR3A was initially cloned and
characterized in rat tissue (Ciabarra et al., 1995; Sucher et al., 1995), but was not
cloned from human tissue until early this century (Andersson et al., 2001). Around the
same time, the final member of the NMDA receptor family, NR3B, was identified and
characterized (Nishi et al., 2001; Matsuda et al., 2002; Bendel et al., 2005; Chatterton et
al., 2002).
7
NR1
NR1 is ubiquitously expressed in brain and spinal cord during development and
adulthood (Moriyoshi et al., 1991; Brose et al., 1993; Tölle et al., 1993; Monyer et al.,
1994). The NR1 transcript contains 22 exons and can be alternatively spliced, resulting
in eight different variants of the NR1 subunit (table 1) (Anantharam et al., 1992;
Durand et al., 1992; Nakanishi et al., 1992; Yamazaki et al., 1992; Hollmann et al.,
1993). Exon five encodes a region in the N-terminus of NR1, named the N1 cassette,
and can either be included or excluded in the protein. In the 3’ end of the transcript,
exons 21 and 22, encoding C-terminal cassettes C1, C2 and C2’, can be alternatively
spliced. The NR1 splice variants differ in their anatomical distribution and generate
NMDA receptors with different physiological and pharmacological properties
(reviewed in Hollmann and Heinemann, 1994; Zukin and Bennett, 1995; Dingledine et
al., 1999; Prybylowski and Wenthold, 2004).
Table 1. The NR1 splice variants.
E5/N1
E21/C1
E22/C2
E22/C2’
NR1-1a
+
+
+
NR1-1b
+
+
NR1-2a
+
+
NR1-2b
+
NR1-3a
+
+
+
NR1-3b
+
+
NR1-4a
+
+
NR1-4b
+
Exons (E) 5, 21 and 22 can be alternatively spliced, resulting
in either the presence or absence of N-terminal cassette N1
and C-terminal cassettes C1, C2 and C2’. Nomenclature
according to Hollmann and Heinemann, 1994.
NR2
In rat brain NR2B and NR2D mRNAs are expressed prenatally, while NR2A and
NR2C mRNAs are detected around birth (Monyer et al., 1994). NR2D mRNA peaks
around postnatal day 7 (P7), and is mainly found in midbrain structures, and in the
thalamus and brain stem of adult animals. NR2C mRNA and protein is almost
exclusively found in cerebellum, and the protein is detected around P5 in rat brain
(Wenzel et al., 1997). At birth, NR2B is expressed in most regions of the brain, but
during the first postnatal weeks its distribution becomes highly restricted to
hippocampus and forebrain. The NR2A protein levels increase after birth and in adult
rat NR2A is ubiquitously expressed throughout the brain (Petralia and Wenthold 1994;
Wenzel et al., 1995; Wenzel et al., 1997).
The NR2 subunits are also expressed in the spinal cord. Both NR2B and NR2D are
expressed in the embryonic spinal cord, while NR2A and NR2C are absent (Monyer et
al., 1994). During postnatal development the NR2A and NR2B subunits follow the
same distribution pattern as in the brain, with higher NR2B than NR2A levels
(Stegenga and Kalb, 2001). The adult rat spinal cord seems to express at least low
8
levels of all four NR2 subunits, even though the results are slightly contradictory (Tölle
et al., 1993; Petralia and Wenthold, 1994; Stegenga and Kalb, 2001). The adult human
spinal cord expresses NR2A, NR2C and NR2D (Sundström et al., 1997).
NR3
Two different genes encode the NR3 subunits, NR3A and NR3B, and in some species
the NR3A transcript can be spliced into two different variants. The longer splice variant
includes 60 base pairs in the 3’ region and has been identified in rat (Sun et al., 1998).
Both splice variants have their highest expression during the first postnatal weeks and
NR3A-l seems to be the dominating form in the entorhinal cortex and thalamus. In
mouse, NR3B mRNA is mostly found in brain stem and in motor neurons of the spinal
cord and at low levels in the cerebellum (Nishi et al., 2001). Similarly, Matsuda and coworkers found NR3B mRNA in mouse midbrain, medulla and spinal cord (Matsuda et
al., 2002). In addition, they found that in whole brain the NR3B mRNA levels were
constant during development and that NR3B protein could be co-immunoprecipitated
with NR1, NR2A and NR2B from transfected human embryonic kidney 293 (HEK293)
cells. Investigation of NR3B distribution in adult rat brain confirmed previous studies,
and showed both mRNA and protein to be restricted to motor neurons in the ventral
horn and to brain stem nuclei (Chatterton et al., 2002). In contrast, one study found
NR3B mRNA to be widely distributed throughout adult rat brain (Andersson et al.,
2001). So far there is only one study on NR3B expression in adult human CNS, and it
shows NR3B mRNA to be expressed in the hippocampus and adjacent cortex (Bendel
et al., 2005). We performed immunohistochemistry (IHC) on adult human brain and
found expression of NR3B protein in hippocampus (unpublished observation).
NMDA receptor activation
NMDA receptors are most likely tetrameric assemblies of NR1, NR2 and sometimes
NR3 subunits (figure 2) (Laube et al., 1998; Rosenmund et al., 1998). NR1 and NR3
bind glycine (Hirai et al., 1996; Yao and Mayer, 2006; Nilsson et al., 2007) and the
NR2 subunits bind glutamate (Laube et al., 1997). Recent evidence has shown that Dserine, which binds to the same site as glycine, might be as important for NMDA
receptor regulation as glycine (reviewed in Wolosker, 2007). Other endogenous
substances, such as L-aspartate, have been suggested to regulate NMDA receptor
activity, but the importance of these is still unclear (Collins and Probett, 1981; Lysko et
al., 1989). NR1 is present in all known functional NMDA receptors (Forrest et al.,
1994) and expression of functional recombinant receptors in mammalian cells require
both NR1 and NR2 subunits (McIlhinney et al., 1996). Moreover, NR1 has been shown
to be essential for transport of NR3A-containing receptors to the cell surface (PérezOtano et al., 2001).
9
Figure 2. Functional NMDA receptors
expressed at the plasma membrane are
believed to consist of four assembled
subunits, of which at least one is NR1.
NR1 and NR3A bind glycine and Dserine and the NR2 subunits bind
glutamate.
Activation of endogenous NMDA receptors requires simultaneous ligand-binding to
both the glycine and glutamate sites (figure 3) (reviewed in Dingledine et al., 1999;
Paoletti and Neyton, 2007). In addition to ligand binding, receptor activation requires
depolarization of the plasma membrane, which allows the Mg2+ ion situated in the
channel pore to be released. This depolarization can be mediated by the glutamate
receptor α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA), situated in
the same postsynaptic membrane. When activated, the NMDA allows influx of Na+ and
Ca2+ and out flux of K+ through the channel pore.
Figure 3. Activation of the NMDAR requires binding of the ligands glycine (or D-serine) and
glutamate as well as depolarization of the plasma membrane. The depolarization can be
mediated by the AMPAR, and results in release of the Mg2+ ion situated in the channel pore at
resting membrane potentials. The channel pore is permeable to positively charged ions, of
which K+, Na+ and Ca2+ endogenously are the most important. Abbreviations; N-methyl-Daspartate receptor (NMDAR), α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid
receptor (AMPAR).
10
THE NR3A SUBUNIT
Spatial and temporal distribution of NR3A
The early studies on NR3A performed in rat, showed NR3A mRNA to be elevated just
prior to birth and during the first postnatal week. The levels begin to decline between
P7 and P14 and remained at a lower, but constant level into adulthood (Ciabarra et al.,
1995; Sucher et al., 1995). The highest levels of the NR3A transcript are expressed in
spinal cord, brain stem, thalamus, hypothalamus, hippocampus, amygdala and in
cortical areas. It was also detected in hindbrain but not in cerebellum. In 2002 Wong
and co-workers published an extensive study on the temporal and spatial distribution of
NR3A protein in rat CNS (Wong et al., 2002). They found NR3A to be highly
expressed at birth, to further increase and peak at P8 and then decline to lower levels in
adult rat brain. They detected NR3A in spinal cord, brain stem, thalamus,
hypothalamus, hippocampus and amygdala, and in cerebral and cerebellar cortices.
Investigation of cerebral cortex at P16 showed NR3A immunoreactivity mainly in layer
V, and to some extent in layers II/III and VI. Further, electron microscopy studies
showed NR3A to be localized to asymmetric synapses, but not always to be directly
associated with the PSD (Wong et al., 2002). A similar temporal and spatial expression
pattern of NR3A protein was described by Al-Hallaq and co-workers, who also
investigated the association of NR3A with the other NMDA receptor subunits in rat
brain (Al-Hallaq et al., 2002). They found that approximately 80% of expressed NR3A
subunits were associated with NR1 in P10 cortex, but only 40% in adult rat cortex.
However, only about 10% of NR1 and NR2A and 5% of NR2B were associated with
NR3A at P10, decreasing to about half in adult rat cortex. This shows that in both
postnatal and adult rat cortex the majority of NMDA receptors do not contain NR3A.
Moreover, NR3A mRNA and protein has been shown to be expressed in rat and mouse
retina (Sucher et al., 2003).
An extensive study investigating the NR3A mRNA levels in adult macaque brain
revealed that the distribution in monkey brain slightly differed from that in rat brain
(Mueller and Meador-Woodruff, 2005). Just as rat brain, monkey brain expressed
NR3A mRNA in cerebral cortex, hypothalamus, thalamus, hippocampus and amygdala,
but in contrast to rat brain, high expression was also found in substantia nigra and
cerebellum. Investigation of NR3A in human brain was first performed on embryonic
tissue, and showed NR3A to be expressed in the developing cortex and in the spinal
cord (Mueller and Meador-Woodruff, 2003). Soon after, investigation of adult human
prefrontal and temporal cortices, showed NR3A mRNA also in adult human brain
(Mueller and Meador-Woodruff, 2004). A recent study showed the developmental
profile of NR3A protein in human brain to be similar to what has previously been
shown in rat (Henson et al., 2008).
NR3A knock out mice
NR3A is enriched in PSD preparations and co-immunoprecipitates with NR1, NR2A
and NR2B but not with glutamate receptor subunits (GluR) GluR2 or GluR6/7, in brain
extracts and in over-expressing heterologous cells (Das et al., 1998; Pérez-Otano et al.,
2001; Al-Hallaq et al., 2002; Sasaki et al., 2002). Mice lacking NR3A reach adulthood,
11
are fertile and do not display obvious behavioral abnormalities (Das et al., 1998).
NR3A knock out (KO) mice expressed normal NR1, NR2A and NR2B protein levels
and displayed increased NMDA-induced currents compared to wild type (WT) mice.
The increased currents observed in the KO mice are consistent with earlier studies
showing NR3A to have attenuating effects on NMDA receptor currents in Xenopus
laevis oocytes injected with NR1, NR2 and NR3A (Ciabarra et al., 1995; Sucher et al.,
1995; Das et al., 1998). The presence of NR3A in the NMDA receptor complex also
reduces Ca2+ permeability and sensitivity to Mg2+ block (Ciabarra et al., 1995; Sucher
et al., 1995; Das et al., 1998; Pérez-Otano et al., 2001; Chatterton et al., 2002; Sasaki et
al., 2002). In addition to electrophysiological alterations, neurons in the NR3A KO
mice had an increased density of dendritic spines, which displayed enlarged heads and
elongated necks (Das et al., 1998). The effect on dendritic spines was more clearly seen
in postnatal than adult mice, and preferentially in brain regions normally expressing
NR3A. Male NR3A KO mice also show enhanced prepulse inhibition of the startle
response (Brody et al., 2005). This is a measure of sensorimotor gating, which is
impaired in schizophrenic hyperactivity disorders.
The atypical NR1/NR3 glycine receptor
NR3A has been reported to form an atypical excitatory glycine receptor together with
NR1 when injected into Xenopus laevis oocytes (figure 4). This novel receptor subtype,
which possesses low Ca2+ permeability and low sensitivity to Mg2+ block, lacks the
NR2 subunit, and accordingly is not affected by NR2-binding substances such as
glutamate and NMDA (Chatterton et al., 2002; Awobuluyi et al., 2007; Madry et al.,
2007). When measuring whole-cell currents from cultured cerebrocortical neurons,
Chatterton and co-workers recorded glycine-evoked currents with a similar
pharmacological profile as those measured from the NR1/NR3A receptors in oocytes.
This result suggests that the atypical glycine receptor is expressed endogenously in
neurons (Chatterton et al., 2002). Moreover, over-expression of NR1 together with
NR3A in HEK293 cells was reported to result in functional NR1/NR3A glycine
receptors (Pina-Crespo and Heinemann, Soc. Neurosci. Abstr. 957.1, 2004). These
results were presented at the Neuroscience meeting 2004 but have not yet been
published.
Figure 4. The atypical glycine receptor is
composed of NR1 and NR3A subunits and
lacks NR2 subunits. The receptor is
activated by glycine in the absence of
glutamate, possesses low Ca2+ permeability
and low sensitivity to Mg2+ block. The
endogenous expression of this atypical
receptor is debated.
12
Earlier studies on HEK293 cells showed NR1-1a/NR3A complexes to be transported to
the cell surface and properly inserted into the plasma membrane, but not to form
functional receptors in the absence of NR2 (Pérez-Otano et al., 2001). However,
Smothers and Woodward recently showed that co-expression of NR1, NR3A and
NR3B in HEK293 cells produced glycine-activated currents (Smothers and Woodward,
2007). In contrast to the studies performed in oocytes and by Pina-Crespo and
Heinemann, expression of both NR3A and NR3B together with NR1, was required for
functional receptors to be formed in HEK293 cells. Analysis of NMDA receptor
pharmacology in Xenopus laevis oocytes is complicated by the fact that oocytes express
a glutamate binding subunit XenU1 (Soloviev and Barnard, 1997). In addition, a recent
study found oocytes to express mRNA encoding orthologs of all four NR2 subunits,
XenNR2A-XenNR2D, as well as XenNR2B protein (Schmidt and Hollmann, 2008).
The presence of these endogenously expressed glutamate-binding subunits needs to be
taken into account when interpreting data from the Xenopus system.
Another recent study investigated the presence of the NR1/NR3A glycine receptor in
both WT and hippocampal neurons from transgenic mice over-expressing the NR3A
subunit (Tong et al., 2008). They could not detect any glycine-evoked currents in either
WT or NR3A over-expressing neurons, suggesting that the atypical NR1/NR3A
receptor is not expressed endogenously. This conclusion is contradictory to the
previous report by the same group, mentioned above, which conclude that the atypical
NR1/NR3A receptor is present in neurons (Chatterton et al., 2002). In Tong’s study the
measurements were performed on cultured neurons derived from hippocampus, while
cerbrocortical neurons were used in Chatterton’s study. Thus, one possibility for the
contradicting results could be that hippocampal and cerebrocortical neurons differ in
their subunit composition. Cerebrocortical neurons might express functional
NR1/NR3A receptors, while hippocampal neurons do not, despite over-expression of
NR3A.
NR3A in glial cells
One of the first studies reporting NMDA receptors in glial cells was performed on
Schwann cells (Evans et al., 1992) and was followed by a study illustrating the
occurrence of NMDA receptors in cortical astrocytes (Conti et al., 1996). NMDA
receptors on glial cells are thought to mediate some of the white matter pathology
associated with disorders such as stroke, vascular dementia, multiple sclerosis and brain
and spinal cord trauma (reviewed in Lipton 2006; Stys and Lipton, 2007; Verkhratsky
and Kirchhoff, 2007).
There are a number of recent studies illustrating the existence of NMDA receptors and
NR3A in oligodendrocytes (Káradóttir et al., 2005; Salter et al., 2005; Micu et al.,
2006). NMDA receptors are present both in precursors, immature and mature
oligodendrocytes in cerebellum and corpus callosum (Káradóttir et al., 2005) and in the
optic nerve NR1, NR2A, NR2B, NR2C, NR2D and NR3A, but not NR3B protein was
detected (Salter et al., 2005; Micu et al., 2006). Double-labeling revealed colocalization of NR1 and NR2C immunoreactivity and NR2C and NR3A
immunoreactivity, suggesting that NR1/NR2C/NR3A complexes might be formed
(Káradóttir et al., 2005). The NMDA receptors were relatively insensitive to Mg2+
13
block, indicating that the receptor complex contains NR3A. Moreover, NMDA
receptors were mainly localized to oligodendrocytes processes, and injury to the
processes could be prevented by blocking NMDA receptors (Salter et al., 2005). These
findings imply that drugs targeting NMDA receptors, and maybe specifically NR3A,
might prove useful in preventing and treating white matter disorders.
NR3A and schizophrenia
Dysregulation of the NMDA receptor has been implicated in schizophrenia (reviewed
in Olney et al., 1999) and a few studies have addressed the possible involvement of
NR3A in the disorder. Mueller and Woodruff found the expression of NR3A mRNA to
be elevated in the dorsolateral prefrontal cortex of postmortem brains from individuals
with diagnosed schizophrenia (Mueller and Meador-Woodruff, 2004). Moreover, they
found NR3A mRNA levels to be decreased in the dorsolateral prefrontal cortex of
brains from people with bipolar disorder. They did not detect any change in NR3A
mRNA levels in the inferior temporal cortex for either schizophrenia or bipolar
disorders. Male NR3A KO mice exhibit an increased inhibition of the startle response
(Brody et al., 2005). This response is altered in some schizophrenic patients (reviewed
in Braff et al., 1992) and has been linked to the NMDA receptor (reviewed in Geyer et
al., 2001). Another study addressed whether single nucleotide polymorphism in the
NR3A gene were more frequent in schizophrenia patients compared to matched
controls, but did not find any significant correlations (Gulli et al., 2007). Finally, a
recent study investigating the NR1 and NR3A protein levels in dorsolateral prefrontal
cortex from schizophrenic patients, reported the NR1 and NR3A levels to be normal
(Henson et al., 2008). This suggests that the elevated NR3A mRNA levels reported by
Mueller and Meador-Woodruff do not reflect an increase of NR3A protein levels, and
accordingly is unlikely to affect NMDA receptor composition and signaling.
14
THE POSTSYNAPTIC DENSITY
The PSD was first described in an electron-microscopy study as an electron-dense area
facing the presynaptic active zone (Palay, 1958). Studies of PSD morphology in 1959,
led Gray to distinguish synapses with a prominent PSD from those with a less
prominent PSD, and classify them into type 1 and type 2 synapses. (Gray, 1959). The
type 1 synapses were later shown to be glutamatergic, excitatory synapses, which
usually are localized to dendritic spine heads (Peters et al., 1991) (figure 5). The PSD
contains membrane-bound proteins such as neurotransmitter receptors and cell
adhesion molecules, signaling molecules including kinases and phosphatases,
scaffolding proteins such as Shank and the postsynaptic density protein 95 (PSD95)
family-members and cytoskeletal components, especially actin and its associated
proteins (figure 6) (reviewed in Scannevin and Huganir, 2000; Okabe, 2007; Sheng and
Hoogenraad, 2007). This broad array of proteins work in concert to regulate complex
mechanisms, for instance receptor trafficking, synaptic strength and the stability of
dendritic spines.
The dendritic spine head provides a micro-compartment allowing local changes of ion
concentrations and signal transductions (figure 5). The spines contain ER,
polyribosomes (PR) and molecules required for protein synthesis. Local protein
synthesis is believed to be regulated by synaptic activity and to be involved in synapsespecific forms of long-term synaptic plasticity (reviewed in Martin et al., 2000).
Proteins are mainly inserted and removed from the plasma membrane at the
extrasynaptic endocytic zones (EZ), situated on each side of the synaptic membrane,
which faces the presynaptic terminal. There is a continuous, activity-dependent
exchange of proteins between extrasynaptic and synaptic sites (reviewed in Chouquet
and Triller, 2003; Sheng and Hoogenraad, 2007).
Figure 5. A dendritic spine forming the
postsynaptic half of an excitatory synapse. The
proteins of the PSD, colored in blue, are
continuous with the postsynaptic membrane
through the interactions with transmembrane
proteins. Actin, indicated by brown lines, is
abundant in dendritic spines and smooth
endoplasmic
reticulum
(SER)
and
polyribosomes (PR) are present. The endocytic
zone (EZ) is located in the extrasynaptic region
and clathrin-coated vesicles (CV) are indicated.
Adapted from Sheng and Hoogenraad, 2007.
15
The NMDA receptor complex
The NMDA receptor complex is a major constituent of the PSD at glutamatergic
synapses, with a large number of proteins directly and indirectly associated with the
complex (reviewed in Grant and Husi, 2001; Boeckers, 2006). High throughput studies,
examining the PSD composition have estimated the NMDA receptor complex to
contain at least 70 different proteins (Husi et al., 2000; Walikonis et al., 2000; Jordan et
al., 2004; Colllins et al., 2005; Cheng et al., 2006), but as new protein-interactions are
identified the number grows. Below are a few examples of proteins associated with the
NMDA receptor complex.
Figure 6. Schematic picture illustrating some of the components of the NMDA receptor
complex described in the text. Abbreviations; NMDA receptor subunits NR1, NR2 and NR3A
(1, 2 and 3), Ca2+ calmodulin-dependent protein kinase II (CaMKII), protein phosphatase 2A
(PP2A), postsynaptic density protein 95 (PSD95), guanylate kinase-associated protein (GKAP),
Src homology 3 (SH3) and ankyrin repeat-containing protein (Shank), neuronal nitric oxide
synthase (nNOS), inositol 1,4,5-trisphosphate (IP3) receptor (IP3R), smooth endoplasmic
reticulum (SER) and metabotropic glutamate receptor (mGluR).
Scaffolding proteins
The membrane-associated guanylate kinase (MAGUK) family of scaffolding proteins
comprises PSD95 (Cho et al., 1992; Kistner et al., 1993), synapse-associated protein
(SAP) SAP97/human discs large (hDlg) (Lue et al., 1994; Muller et al., 1995),
PSD93/Chapsyn110 (Brenman et al., 1996a; Kim et al., 1996) and SAP102 (Muller et
al., 1996) (figure 7). All MAGUK proteins are abundant in the PSD, except SAP97,
which is mainly found at presynaptic sites and in axons. The MAGUK members
contain the protein-protein interaction domains PSD-95/Dlg/zona occludens-1 (ZO-1)
(PDZ), Src homology 3 (SH3) and gunaylate kinase (GK) through which they connect
proteins with each other (reviewed in Sheng and Pak, 2000; Kim and Sheng, 2004).
The NR2 subunits bind to PDZ domains through a conserved amino acid motif in their
outermost C-terminal tails, -ESXV (figure 6). A similar PDZ binding motif, -STVV, is
present in NR1 splice variants containing the C2’ cassette (Kornau et al., 1995). PSD95
16
was the first MAGUK member shown to bind to the NMDA receptor and is also the
most studied (Kornau et al., 1995; Niethammer et al., 1996). The interactions between
NMDA receptors and MAGUK proteins have been implicated in several functions, for
instance assembly of signaling complexes, localization and clustering of NMDA
receptors at postsynaptic sites and anchoring of NMDA receptors to the cytoskeleton
(reviewed in Sheng and Pak, 2000). However PSD95, which is the most abundant
MAGUK protein at the PSD, outnumbers NMDA receptors by approximately 20-fold
and binds to several proteins in the PSD in addition to the NMDA receptor subunits
(Cheng et al., 2006).
Figure 7. Schematic picture of a membrane-associated guanylate kinase (MAGUK) protein
with its protein-protein interaction domains indicated. Abbreviations; PSD-95/Dlg/ZO-1 (PDZ),
Src homology 3 (SH3) and gunaylate kinase (GK). The picture is not drawn to scale.
Another family of scaffolding proteins, comprising proline-rich synapse-associated
protein/SH3 and ankyrin repeat-containing protein 1-3 (ProSAP)/(Shank1-3), contains
the protein-protein interaction domains PDZ, SH3, an ankyrin repeat and a sterile alpha
motif (SAM). The Shank family is involved in morphological and functional
maturation of dendritic spines (Naisbitt et al., 1999; Boeckers et al., 1999; Sala et al.,
2001). The Shank proteins bind both to the PSD95 interacting protein guanylate kinaseassociated protein (GKAP) and to the metabotropic glutamate receptor (mGluR)
binding protein Homer, thus providing a link between ionotropic and metabotropic
glutamate receptors at the PSD (Xiao et al., 1998; Tu et al., 1999). Moreover, Homer
binds to the inositol 1,4,5-trisphosphate (IP3) receptor (IP3R) situated in the membrane
of the SER, linking the receptor complex to the intracellular Ca2+ stores the SER
provides (Tu et al., 1998).
Regulatory proteins
A large number of kinases and phosphatases reside in the PSD. One of the NMDA
receptor-binding kinases found to be present in very high amounts in the PSD is Ca2+
calmodulin-dependent protein kinase II (CaMKII) (Kennedy et al., 1983; Cheng et al.,
2006). When Ca2+ ions flow into the postsynaptic cell through activated NMDA
receptors, they bind to calmodulin and thereby regulate CaMKII activity. This
regulation has been shown to be involved in certain types of NMDA receptor mediated
plasticity (reviewed in Malenka and Bear, 2004; Colbran and Brown, 2004). Based on
the abundance of CaMKII at the PSD, CaMKII has been suggested to, besides its
enzymatic role, play a structural role at the synapse. CaMKII is linked to the actin
cytoskeleton through an interaction with α-actinin (Walikonis et al., 2001) and also
interacts with the actin-associated motor protein Myosin Va (Costa et al., 1999).
NMDA receptor signaling also activates the mitogen-activated kinase (MAPK)
pathway (Bading and Greenberg, 1991), which just like CaMKII signaling, is involved
17
in synaptic plasticity (reviewed in Adams and Sweatt, 2002; Miyamoto, 2006). Other
kinases and phosphatses present in the PSD include casein kinase 2 (CK2), cyclic
adenosine monophosphate (cAMP)-dependent protein kinase (PKA), protein kinase C
(PKC), protein phosphatase 1 (PP1) and protein phosphatase 2A (PP2A). PP1 and PKA
are linked to NR1 through their interactions with the A kinase anchoring protein
(AKAP) Yotiao (Lin et al., 1998; Westphal et al., 1999). The physical link provided by
Yotiao brings PP1 and PKA in close proximity to the NMDA receptor and both
enzymes regulate NMDA receptor activity (Westphal et al., 1999).
The enzyme neuronal nitric oxide synthase (nNOS) is coupled to the NMDA receptor
through its interaction with PSD95 and PSD93 (figure 6) (Brenman et al., 1996b;
Sattler et al., 1999). When the NMDA receptor is to highly activated, for instance
during stroke-induced ischemia, excessive Ca2+ influx leads to excitotoxicity and
neuronal death (reviewed in Dirnagl et al, 1999; Hardinghamn and Bading, 2003). The
excitotoxic cell damage can be mediated by signaling molecule nitric oxide (NO),
which is synthesized by nNOS following NMDA receptor activation (Dawson et al.,
1991). Blocking the interaction between the NMDA receptor and PSD95 prevents the
nNOS-mediated excitotoxic cell death both in neurons and in vivo models of focal
ischemia (Aarts et al., 2002). Disrupting the interaction between the NMDA receptor
and PSD95, with a peptide, has been suggested as a possible strategy for treating stroke
(Aarts et al., 2002; Cui et al., 2007).
Cytoskeletal proteins
Dendritic spines are actin-rich structures, which contain intermediate filaments but little
or no microtubules (reviewed in Sekino et al., 2007). A number of actin-associated
proteins, including α-actinin, neurabin, drebrin, spectrin and cortactin (Gray et al.,
2005), have been identified in the PSD and so has tubulin, microtubule-associated
protein (MAP) MAP1A, MAP1B and MAP2. In addition, the actin-based motor protein
myosin and the microtubule-associated motor protein dynein, are present in the PSD.
While the kinesin superfamily (KIF) of microtubule-based motor proteins have not
been detected (reviewed in Sheng and Hoogenraad, 2007).
The dynamic actin network is connected to several proteins in the PSD and is involved
in the regulation of spine morphology (reviewed in Sekino et al., 2007). α-actinin 2
binds both to NR1 and to NR2B, and competes for the same binding site as calmodulin
on NR1 (Ehlers et al., 1996; Wyszynski et al., 1997). Calmodulin and α-actinin 2 are
involved in the regulation of the interaction between NR1 and the actin cytoskeleton
(Zhang et al., 1998). An additional link between NMDA receptors and the cytoskeleton
is provided by the interaction between NR1 and neurofilaments. This interaction is
splice variant-specific and requires the C1 cassette in the NR1 C-terminus (Ehlers et al.,
1998). Moreover, NMDA receptors are linked to the microtubule cytoskeleton through
the interactions between MAP1A and PSD93 (Brenman et al., 1998) and PSD95
(Reese et al., 2007). And MAP1A is involved in activity-dependent remodeling of
dendritic spines (Szebenyi et al., 2005).
18
Proteins interacting with NR3A
A number of proteins have been shown to interact with NR3A, and these are
summarized in table 2.
Table 2. Proteins binding to NR3A.
Interaction-partner
Protein-type
Reference
Protein phosphatase 2A
Enzyme
Chan and Sucher, 2001
PACSIN1/Syndapin1
Accessory *
Pérez-Otano et al., 2006
Plectin
Cytoskeletal
Eriksson et al., 2007
CARP-1
Signaling
Eriksson et al., 2007
GPS2
Signaling
Eriksson et al., 2007
*PACSIN1 has also been called a scaffolding and adaptor protein (reviewed in
Kessels et al., 2004). Abbreviations; cell cycle and apoptosis regulatory
protein-1 (CARP-1) and G-protein pathway suppressor 2 (GPS2).
PP2A
The first NR3A-interacting protein to be identified was the catalytic subunit of PP2A
(Chan and Sucher, 2001). The interaction was found through a yeast two-hybrid screen
of a human fetal brain cDNA library, using the intracellular C-terminus of NR3A as
bait. By truncating the bait, the membrane proximal 37 amino acids of NR3A were
found to be required for the interaction with PP2A. Further analysis of these 37 amino
acids by alanine-mutagenesis, revealed one histidine and two leucine residues to be
highly involved in the interaction between NR3A and PP2A (Ma and Sucher, 2004).
Chan and Sucher found the interaction between NR3A and PP2A to be activitydependant. Activation by NMDA of NR1/NR2B/NR3A receptors expressed in
HEK293 cells resulted in the loss of binding between NR3A and PP2A. Further, the
binding of PP2A to NR3A regulated the phosphorylation of Ser897 in the NR1 Cterminus, and breaking the interaction between PP2A and NR3A resulted in a
phosphorylated NR1 subunit (Chan and Sucher, 2001).
PACSIN1/Syndapin1
Another NR3A-interacting protein recently found through a yeast two-hybrid screen is
PACSIN1 (Pérez-Otano et al., 2006). PACSIN1 is a neuron-specific adaptor protein
involved in membrane fission during endocytosis (Kessels and Qualmann, 2002;
reviewed in Kessels and Qualmann, 2004). It is localized to synapses, interacts with
dynamin and is highly expressed during neuronal development (Plomann et al., 1998;
Qualmann et al., 1999).
NR3A was found to be part of a protein complex containing PACSIN1, dynamin and
clathrin in rat brain, and PACSIN1 regulated the surface expression of NR3A in
neurons in an activity-dependant manner (Pérez-Otano et al., 2006). Blocking synaptic
transmission or NMDA receptor signaling, inhibited the PACSIN1-mediated removal
of NR3A from the cell surface. The interaction of PACSIN1 is specific for NR3A, as
PACSIN1 did not bind to NR1, NR2A or NR2B. Accordingly, PACSIN1 can
19
specifically remove NR3A-containing receptors from the plasma membrane and this
mechanism might be involved in the maturation of synapses during development
(Pérez-Otano et al., 2006).
Plectin, CARP-1 and GPS2
Plectin, cell cycle and apoptosis regulatory protein-1 (CARP-1) and G-protein pathway
suppressor 2 (GPS2) were also identified as NR3A-binding proteins through a yeast
two-hybrid screen (Eriksson et al., 2007). The interactions with NR3A have been
verified in vitro but not in vivo. Plectin is a large scaffolding protein, containing several
conserved protein domains and is expressed as different isoforms (Wiche et al., 1982;
reviewed in Allen and Shah, 1999). CARP-1 is involved in apoptosis through its
interaction with protein 14-3-3 and alters the expression of cell cycle regulatory genes
(Rishi et al., 2003). GPS2 interferes with G-protein signaling and has been shown to be
involved in cell survival and to modulate p53 activity (Spain et al., 1996; Peng et al.,
2001)
20
SUBUNIT ASSEMBLY AND INTRACELLULAR TRANSPORT
As mentioned above, motor proteins can move either along actin filaments or
microtubules in order to transport cargo to the correct subcellular compartment. The
myosin family of motor proteins move along actin filaments and is most likely
important in compartments devoid of microtubules, such as the neck and head of
dendritic spines. The dynein and kinesin superfamilies (KIF) of motor proteins are both
microtubule-based and transport cargo through axons and dendrites. Axonal transport
comprises transport of membrane-bound vesicles, containing for instance components
of synaptic vesicles, ion channels and adhesion molecules from, the cell body to
synaptic terminals. Molecules transported through dendrites include components of the
PSD, neurotransmitter receptors and mRNA (reviewed in Hirokawa and Takemura,
2005; Caviston and Holzbaur, 2006).
Assembly of the NMDA receptor subunits
The NMDA receptor subunits assemble in the ER and unassembled subunits do not exit
from the ER. For instance, both NR1 and NR2 are required to be expressed
simultaneously in order to be transported to the plasma membrane, and NR1 is required
for NR3 to exit the ER (McIlhinney et al., 1996; Pérez-Otano 2001). The retention of
unassembled subunits in the ER is believed to be mediated by an ER retention motif,
RXR (Zerangue et al., 1999). This motif is found in the NR1 C1 cassette, in NR3A,
NR3B and in NR2B. Subunit assembly most likely masks the ER retention signal and
thereby allows the assembled subunits to exit the ER (McIlhinney et al., 1996;
reviewed in Pérez-Otano and Ehlers 2003; Wenthold et al., 2003). However, NR1
splice variants expressing the C2’cassette, which contains the PDZ-binding motif STVV, overcome the ER retention mediated by the C1 cassette and exit the ER
(Standley et al., 2000; reviewed in Prybylowski and Wenthold, 2004).
A recent publication showed that NR1 and NR2, and NR1 and NR3A form
heterodimers that can assemble two and two into tetramers (Schuler et al., 2008). The
authors suggest the formation of heterodimers to be the initial step in NMDA receptor
assembly. In addition they find that NR2 and NR3A do not form heterodimers,
implying that receptors containing NR3A also contain one NR2 and two NR1 subunits.
This study was performed in Xenopus oocytes, thus it is not yet clear if endogenous
NMDA receptor subunits follow the same pattern of assembly and display the same
stoichiometry.
The exocyst complex
The exocyst, or Sec6/8, complex is an assembly of eight proteins, Sec3, Sec5, Sec6,
Sec8, Sec10, Sec15, Exo70 and Exo84, and was originally identified in yeast (Bowser
et al., 1992; Hsu et al., 1996). The exocyst is associated with ER and Golgi and directs
intracellular membranes to sites where they fuse with the plasma membrane (Hsu et al.,
1999; Yeaman et al., 2001). A novel function of the exocyst was reported by Sans and
co-workers, who found the complex to be involved in NMDA receptor trafficking
(Sans et al., 2003). Through a yeast two-hybrid screen they found SAP102 to bind to
21
the exocyst component Sec8. Co-immunoprecipitation showed that SAP102 and Sec8
binds to NR1 and NR2B in rat brain, and disrupting the interaction between SAP102
and Sec8 blocked the delivery of NMDA receptors to the cell surface. However,
deletion of the PDZ-binding motifs in both NR2B and Sec8 allowed delivery of
NMDA receptors to the cell surface. This suggests that there are alternative routes for
NMDA receptor delivery to the cell surface, and perhaps different mechanisms for
delivery of synaptic and extrasynaptic receptors. In addition, Sans and co-workers
determined that the complex including SAP102, Sec8 and the NMDA receptor is
formed in the ER.
Transport along microtubules
NMDA receptors are transported along microtubules in transport packages that are
rapidly recruited to sites of axodendritic contact (Washbourne et al., 2002). The NMDA
receptor clusters were more mobile than clusters containing GluR1 and rarely
colocalized with PSD95. This suggests that PSD95 is not transported together with the
NMDA receptor, but associates with the receptor at a later stage (Washbourne et al.,
2002).
NR2B-containing receptors are transported along microtubules in membrane-bound
vesicles, by the dendrite-specific kinesin motor protein KIF17 (Setou et al., 2000). The
authors found NR2B to be linked to KIF17 through a protein complex consisting of
mLin-7, mLin-2 and mLin-10, also known as Mint1/X11. Moreover, KIF17
colocalized with mLin-10, NR2B and NR1 in dendrites, indicating that NR2B is
associated with NR1 during transport by KIF17. A later study from the same group
reported transgenic mice over-expressing KIF17 to exhibit enhanced spatial and
working memory (Wong et al., 2002).
The KIF17 vesicles move along dendrites and are associated with extrasynaptic NR2B
(Guillaud et al., 2003). However, Guillaud and co-workers determined that KIF17 does
not move into the synapse, a finding consistent with the fact that KIF17 has not been
detected in the PSD (Cheng et al., 2006). Knock-down of KIF17 reduced the amount of
NR2B, and increased the amount of NR2A at synapses (Guillaud et al., 2003). This
implies that NR2A- and NR2B-containing receptors are delivered to synapses by
separate mechanisms, and that the loss of NR2B-containing receptors at synapses is
compensated by an increased amount of NR2A-contining NMDA receptors. NR2A
replaces NR2B during neuronal development, resulting in mature NMDA receptors
with different pharmacological properties from those expressed postnatally (reviewed
in Cull-Candy et al., 2001). Regulation of KIF17 activity might be one way to control
the NMDA receptor composition at synapse. As expected, depolymerization of
microtubules blocks the movement of KIF17 and impairs the delivery of NR2B to the
cell surface (Guillaud et al., 2003). Moreover, an intact microtubule cytoskeleton is
required for proper NMDA receptor-mediated currents in cortical neurons (Yuen et al.,
2005a).
Transport along microtubules is one way for assembled NMDA receptors to reach the
correct subcellular compartment. Another possibility is that the receptors are inserted to
the plasma membrane soon after the exit from ER and Golgi, and diffuse in the plasma
22
membrane to the correct location. A study by Washbourne and co-workers show that
both ways of trafficking occur in the neuron (Washbourne et al., 2004). Prior to
synapse formation, receptor transport packets move along microtubules and are actively
exocytosed and endocytosed through the dendritic plasma membrane. The transport
packets colocalized with early endosomal antigen 1 and SAP102 (Washbourne et al.,
2004) and SAP102 might provide a link to the exocyst complex.
Receptor trafficking at synapses
There is a continuous exchange of receptors between synaptic and extrasynaptic sites,
as well as between the plasma membrane and intracellular stores. Receptors move by
lateral diffusion in the plasma membrane and extrasynaptic receptors are more mobile
than synaptic receptors. Insertion and removal of receptors is believed to occur mainly
at endocytic zones (EZ) located in the extrasynaptic region (figure 5) (reviewed in
Choquet and Triller, 2003; Kennedy and Ehlers, 2006). The endocytotic proteins AP-2,
clathrin and dynamin are localized to EZ lateral to the PSD (Petralia et al., 2003; Racz
et al., 2004).
The composition of NMDA receptors expressed at extrasynaptic and synaptic sites
differ. The subunits NR2B, NR2D and NR3A, which have their expression peaks
during development, are preferentially expressed at extrasynaptic sites. While NR2A
and NR2C subunits predominate at synaptic sites (reviewed in Pérez-Otano and Ehlers,
2004). This preferential targeting requires subunit-specific regulation and can for
instance be mediated by proteins as PACSIN1, which specifically removes NR3Acontaining receptors from the plasma membrane (Pérez-Otano et al., 2006).
23
MICROTUBULE-ASSOCIATED PROTEINS
There are a number of MAPs expressed in neurons (Sloboda et al., 1975) and some of
the most studied are MAP1A, MAP1B, MAP2, tau, doublecortin and LIS1. The MAPs
bind to microtubules, often in a phosphorylation dependent manner, and regulate their
stability. This thesis focuses on the MAP1 family, comprising MAP1A, MAP1B and
MAP1S. An ortholog to the mammalian MAP1 proteins, named Futsch, has been
identified in Drosophila (Hummel et al., 2000).
The MAP1 family
MAP1A, MAP1B and MAP1S are translated as polyproteins, which undergo
posttranslational cleavage into a heavy chain (HC) and a light chain (LC) (figure 8). In
addition to the heavy chains, the MAP1A polyprotein comprises LC2 and the MAP1B
polyprotein comprises LC1. There is also a smaller light chain, LC3, translated
independently. The MAP1B-HC can associate both with LC1 and LC3 and the
MAP1A-HC can bind all three light chains, LC1, LC2 and LC3 (reviewed in Halpain
and Dehmlet, 2006). The HC:LC ratio has been shown to vary between 1:2 and 1:8,
depending on cell type and sub-cellular fraction, with a higher proportion of LC in
whole brain homogenates than in microtubule preparations (Mei et al., 2000a; Mei et
al., 200b).
Both MAP1A and MAP1B are large proteins comprising approximately 3000 and 2500
amino acids respectively, with small variations between species. MAP1S is
considerably smaller, the human protein comprises only 1059 amino acids, mainly due
to a shorter exon 5 (Orbán-Németh et al., 2005). In addition to its smaller size, the high
expression of MAP1S in organs outside the nervous system distinguishes MAP1S from
MAP1A and MAP1B. The homology between the three proteins is high predominately
in three regions, termed MAP1 homology domains 1-3 (MH1-3) (Orbán-Németh et al.,
2005). MAP1A contains three microtubule-binding regions, MAP1B contains two, and
only one has been identified in MAP1S. All three proteins contain one actin bindingregion situated in their light chains. In addition, one actin-binding region has recently
been identified in the MAP1B-HC (Cueille et al., 2007) (figure 8).
Figure 8. Schematic picture showing the three members of the MAP1 family, MAP1A,
MAP1B and MAP1S. The MAP1 family members undergo post-translational proteolytic
cleavage into a heavy and a light chain. The shaded boxes MH1, 2 and 3, indicate the MAP1
homology domains, which are regions with highly conserved sequence. The picture is not
drawn to scale. Abbreviations; microtubule-binding region (MT) and actin-binding region (A).
24
MAP1S/C19ORF5
Identification
MAP1S is the most recently identified member of the MAP1 family, and its function in
the nervous system is still unclear. MAP1S was first identified as an interaction-partner
of a fibroblast growth factor-associated protein called leucine-rich PPR-motif
containing protein, AAA67549 (LRPPRC), through a yeast two-hybrid screen of a liver
cDNA library (Liu and McKeehan, 2002a). The protein was named chromosome 19
open reading frame 5 (C19ORF5), and on basis of its high homology to MAP1A and
MAP1B, suggested to be a member of the MAP1 family. The same cDNA was later
isolated from testis cDNA libraries, through yeast two-hybrid screening, and given the
names variable charge Y chromosome 2 interacting protein-1 (VCY2IP-1) (Wong et
al., 2004), and RAS-associated domain family protein 1A-binding protein 1 (RAB1)
(Song et al., 2005). Variable charge Y chromosome 2 is a testis-specific protein of
unknown function (Stuppia et al., 2001) and RAS-associated domain family protein 1A
is a tumor suppressor, implemented in a number of different cancer forms. The names
VCY2IP-1 and RAB1, both refer to the proteins interaction-partners, while the name
C19ORF5 refers to the chromosomal location of the gene. Despite C19ORF5 being the
original name, we have chosen to refer to the protein by its fourth, and most recent
name MAP1S (Orbán-Németh et al., 2005). This name indicates the microtubulebinding property of the protein, as well as its homology to MAP1A and MAP1B.
Function
MAP1S was identified in 2002 and there are a limited number of publications
concerning the protein, and of these, only the study by Orbán-Németh and co-workers
involves nervous tissue (Orbán-Németh et al., 2005). In adult mouse tissue MAP1S is
expressed in kidney, lung, spleen, liver and heart, but the highest levels are found in
brain and testis. The protein is expressed already at birth, and the levels increase
slightly during the first postnatal weeks (Orbán-Németh et al., 2005).
Immunoprecipitaion from brain tissue shows that the MAP1S heavy and light chains
bind to each other in vivo. Deletion of the microtubule-binding domain in the light
chain causes MAP1S to lose its microtubule-binding property, indicating that the
protein only contains one microtubule-binding region (Orbán-Németh et al., 2005).
A yeast two-hybrid screen of a liver cDNA library, with MAP1S as bait, revealed that
MAP1S binds to mitochondria-associated NADH dehydrogenas I, cytochrome c
oxidase I and RASSF1 (Liu et al., 2002b). When over-expressed in the cell line HepG2,
which is derived from liver, MAP1S causes aggregation of mitochondria and genome
destruction (Liu et al., 2005b). The interaction between the microtubule-binding tumor
suppressor RASSF1 and MAP1S has been reported by several groups (Liu et al.,
2002b; Dallol et al., 2004; Liu et al., 2005a; Song et al., 2005). These studies suggest
MAP1S to be involved in apoptosis and cell cycle mechanisms through its interaction
with the microtubular cytoskeleton. In addition, there is one study reporting that
MAP1S binds to double stranded DNA, but the function of this interaction is not
known (Liu et al., 2005c).
25
MAP1B
MAP1B was identified as a microtubule-binding protein in the 1970s (Sloboda et al.,
1975) and is involved in microtubular stabilization, axon generation and growth cone
motility (reviewed in González-Billault et al., 2004; Riederer, 2007). MAP1B was
cloned in rat in 1989 and its expression was found to be higher during development
than in the adult rat brain. During postnatal development MAP1B is replaced by
MAP1A, which is highly expressed in the adult nervous system (Riederer and Matus
1985; Safaei and Fischer, 1989). MAP1B expression is maintained in areas undergoing
neurogenesis in adult animals such as the olfactory bulb. Moreover, MAP1B expression
is induced following injury to the peripheral nervous system (PNS), suggesting a role
for MAP1B both in development and regeneration (reviewed in Emery et al., 2003).
Full-length MAP1B is encoded by seven exons but the gene includes two additional
exons, 3A and 3U (Lien et al., 1994; Kutschera et al., 1998). If translated, the 3A and
3U gives rise to a MAP1B protein with a truncated N-terminus. Transcripts including
either 3A or 3U have been detected both in mouse and rat tissue, and estimated to give
rise to 1-10% of total MAP1B transcripts (Lien et al., 1994; Kutschera et al., 1998).
MAP1B knock out mice
In contrast to MAP1S, the function of MAP1B in the nervous system has been
extensively investigated, with no less than four different KO mice generated (Edelmann
et al., 1996; Takei et al., 1997; González-Billault et al., 2000; Meixner et al., 2000).
However, interpreting the function of MAP1B from these studies has not been straight
forward since the phenotypes of the KO mice differ significantly. The first KO mouse
generated by Edelmann and co-workers displayed a severe phenotype, and died in utero
prior to embryonic day 8.5. Therefore, the study describes the heterozygous mice,
which have reduced body weight, visual and motor impairments. Morphological
examination revealed malformations of cerebellum but unaffected hippocampus, and
immunohistochemical analysis showed the Purkinje cells to possess an altered
morphology and retinal layers to be disorganized. (Edelmann et al., 1996).
The second KO mouse was generated by Takei and co-workers and displayed a much
milder phenotype (Takei et al., 1997). The heterozygous mice did not exhibit either
developmental or behavioral abnormalities and could not be distinguished from WT
mice. The homozygous mice had slightly decreased body and brain weight and delayed
nervous system development. No abnormalities in hippocampus, retina, olfactory bulb
or spinal cord were observed by IHC, either at postnatal day 8 or in adult mice.
However, myelination of the optic nerve seemed to be delayed, with reduced
myelination at postnatal day 8, but normal myelination in the adult. Moreover, they
investigated the number and density of microtubules in axons of KO mice but did not
find any alterations compared to WT mice.
In the year 2000, studies on two additional MAP1B deficient mice were published
(González-Billault et al., 2000; Meixner et al., 2000). Both these strains displayed
intermediate, but slightly different phenotypes, compared to the previously generated
KO mice. The heterozygous mice generated by Meixner and co-workers were
indistinguishable from WT mice, while homozygous mice displayed decreased body
26
weight as well as a number of developmental abnormalities. The abnormalities
included a total lack of corpus callosum in 80% of the homozygous animals, and
severely malformed corpus callosum in the remaining. Moreover, the animals
demonstrated a reduced thickness of myelin sheaths around axons in the PNS, which
resulted in attenuated conduction velocity. To address the visual impairments and
disorganized retinal layers reported in heterozygous mice by Edelmann and co-workers,
Meixner and co-workers investigated the retina by IHC, but did not detect any
abnormalities. Moreover, they did not find any alterations of GABAc receptor
clustering, which might have been expected since MAP1B has been reported to be
involved in GABAc receptor clustering in the retina (Hanley et al., 1999). In addition,
they claim that the clustering of a number of glutamate receptor subunits, including
NR1 and NR2B, as well as PSD95 is unchanged in the homozygous mice.
The phenotype of the fourth MAP1B deficient mouse generated, most closely
resembles the mouse generated by Meixner and co-workers. The homozygous mice die
at postnatal day 1, exhibit an abnormal limb posture and lack of response to physical
stimuli applied to the limbs (González-Billault et al., 2000). Brains from homozygous
mice were analyzed by IHC at embryonic day 19. The analysis revealed an increased
ventricular volume of 60-70%, an interrupted corpus callosum and abnormal
development of cerebellum. The hippocampus and olfactory bulb displayed abnormal
lamination and disorganized cells. Moreover, the thickness of cerebral cortex was
reduced and its lamination absent. In contrast, heterozygous mice did not have any
alterations of either cerebral cortex or corpus callosum. By the gene-trapping approach
used in this study, β-galactosidase was produced under the control of the Map1b
promoter, allowing the normal distribution of MAP1B expression to be detected. X-gal
staining of heterozygous 13 day-old embryos, revealed MAP1B expression to be
restricted to the nervous system, with high expression throughout both the CNS and
PNS.
Despite the varying phenotypes the different KO mice display, these studies give
valuable insight into the functions of MAP1B and its involvement in CNS and PNS
development. The reason for the discrepancies between mice has been discussed and
one contributing factor could be that the mice have different genetic background. Mice
with the more severe phenotype are derived from strain 129 (Edelmann et al., 1996;
González-Billault et al., 2000) and mice with milder phenotype are derived from strain
C57 (Takei et al., 1997; Meixner et al., 200). However, even though genetic
background might contribute, it is unlikely that the large variations between the mice
generated by Edelmann and Takei are generated solely by that parameter. The KO mice
generated by Edelmann and co-workers express a N-terminal fragment of MAP1B,
which has been suggested to work in a dominant-negative manner and explain the
severe abnormalities observed even in heterozygous mice (Meixner et al., 2000; Tögel
et al., 1998). The KO mice generated by Takei and co-workers, on the other hand,
express residual levels of a slightly truncated MAP1B protein (Takei et al., 1997). This
might compensate for the loss of full-length MAP1B and explain the very mild effect
they observe.
In study IV, we used brains and hippocampal neurons from the MAP1B deficient mice
generated by González-Billault and co-workers (González-Billault et al., 2000).
27
Hippocampal neurons from these mice have been reported to display delayed axonal
outgrowth and abnormal growth cones, when examined after 2 days in vitro (DIV)
(González-Billault et al., 2001). Moreover the axons from cells lacking MAP1B had
increased MAP2 immunoreactivity in their axons, compared to cells derived from WT
mice, suggesting a certain redundancy between MAP1B and MAP2. Tau
immunoreactivity, on the other hand, was similar in neurons lacking MAP1B and WT
(González-Billault et al., 2001). The neurons used in study IV where analyzed after 21
DIV and at this time point the neurites morphological difference are much less
pronounced. Instead there is a difference in the morphology of filopodia, with longer
and thinner filopodia in neurons from MAP1B deficient animals than in WT neurons
(E. Tortosa, unpublished observation).
MAP1B phosphorylation mode I and mode II
MAP1B is a large protein under the regulation of numerous kinases and phosphatases,
with as many as 33 sites found to be phosphorylated in vivo (Collins et al., 2005). Two
different modes of MAP1B phosphorylation have been described, mode I and mode II
(Ulloa et al., 1993, Ulloa et al., 1994; Garcia-Perez et al., 1998). Mode I is directed by
proline-dependent kinases and is present in growing axons during early development.
Mode II is mediated by casein kinase II and is present both in axons and dendrites and
persist throughout postnatal development.
MAP1B in disease
MAPIB has been implicated in a number of pathological conditions of the nervous
system, including fragile X mental retardation syndrome, giant axonal neuropathy and
cancer. There is also one study describing MAP1B protein levels in post mortem brains
from patients with schizophrenia, bipolar disorder and major depression. The authors
found decreased MAP1B levels in anterior cingulate cortex of brains from patients with
bipolar disorder, but unaltered MAP1B levels in brains from patients with
schizophrenia and major depression (Bouras et al., 2001). Even though this is just one
single study, parallels can be drawn to the report showing NR3A mRNA levels to be
elevated in dorsolateral prefrontal cortex, of brains from patients with schizophrenia
(Mueller and Meador-Woodruff, 2004). However, to determine the significance and
possible relationship between these altered MAP1B and NR3A levels additional studies
need to be performed.
Of the pathological conditions mentioned above, the involvement of MAP1B is
probably most studied in the fragile X mental retardation syndrome. The syndrome,
which is inherited, involves cognitive, behavioral and emotional dysfunction, in many
patients resembling autistic symptoms (reviewed in Bernardet and Crusio, 2006;
Koukoui and Chaudhuria, 2007). The syndrome is caused by lack of expression of the
fragile X mental retardation protein (FMRP). FMRP is localized to axons, dendrites
growth cones, and dendritic spines (Antar et al., 2006). FMRP binds to different
mRNAs and regulate their translation, and lack of FMRP results in misregulated
MAP1B translation and increased microtubule stability (Lu et al., 2004). Moreover,
FMRP KO mice exhibit an increased number of filopodia and a reduced number of
mature dendritic spines (Antar et al., 2006).
28
Giant axonal neuropathy (GAN) is a disorder causing axonal degeneration and
aggregation of cytoskeletal components in sensory and motor neurons. The GAN gene
encodes gigaxonin, which is a ubiquitously expressed protein that binds to the MAP1BLC1 (Ding et al., 2002), and over-expression of gigaxonin causes degradation of LC1
(Allen et al., 2005).
One of the groups identifying MAP1S as a binding partner of the tumor suppressor
RASSF1A, also found MAP1B to bind to RASSF1A through the same yeast twohybrid screen (Dallol et al., 2004). The authors showed that RASSF1A is involved in
regulating the stability of microtubules, and thereby cell cycle dynamics. However, the
possible connection between MAP1B and cancer is still far from clear.
29
AIMS OF THE THESIS
GENERAL AIM
The aim of this thesis was to determine the function of the human NMDA receptor
subunit NR3A, since NMDA receptor signaling is involved in normal brain
development as well as number of pathological conditions. Further knowledge about
the receptor and its subunits gives insight into developmental processes, and may also
open for new treatment-strategies in disorders where NMDA receptor signaling is
affected.
SPECIFIC AIMS
•
•
•
•
•
•
30
To clone and sequence the human NR3A subunit.
To investigate the occurrence of NR3A splice variants in human and rat
CNS tissue.
To determine the expression and distribution of NR3A mRNA and protein
in embryonic and adult human CNS tissue.
To determine the solubility of NR3A compared to the other NMDA
receptor subunits.
To establish whether NR3A is associated with the other NMDA receptor
subunits in adult human brain and spinal cord.
To identify and analyze novel NR3A-binding proteins.
RESULTS AND DISCUSSION
STUDY I, CLONING
When this project was started NR3A had only been cloned from and described in rat.
Therefore we set out to clone the human NR3A, and this is described in study I. We
found that human NR3A comprises 1115 amino acids and has a calculated molecular
weight of 122 kD after removal of the first 33 amino acids, which are predicted to be a
signal peptide for intracellular targeting. The amino acid sequence of the human
subunit was 93% homologous to that of the rat, and most of the potential glycosylation
and phosphorylation sites are conserved. So far NR3A has not been shown to be either
glycosylated or phosphorylated, therefore it is not clear if the variations in such sites
between species are of any significance. Sites that are essential for protein function
and/or regulation are likely to be conserved between species.
The hydrophobicity profile of NR3A is similar to the other NMDA receptor subunits,
predicting four membrane regions, an extracellular N-terminus and an intracellular Cterminus. The highest homology between the NMDA receptor subunits was observed in
the membrane regions and in the extracellular loop between membrane regions three
and four. This extracellular loop forms the ligand-binding pocket together with the Nterminus.
The long splice variant of NR3A (NR3A-l) described in rat (Sun et al., 1998), was not
identified in either embryonic or adult human CNS tissue. Sun and co-workers report
that mRNAs encoding NR3A and NR3A-l have slightly different distribution patterns
in adult rat brain, with NR3A-l being the predominant form in the entorhinal cortex and
thalamus. Both splice variants present similar developmental regulation, with the
highest mRNA levels during the first two postnatal weeks. NR3A-l contains a 20 amino
acid insert in the intracellular C-terminus and this can potentially affect both
interactions with intracellular proteins and signaling at the PSD, as well as trafficking
through the endocytic machinery. Both NR3A splice variants contain the ER retention
motif RRR, which is also found in the C1 cassette of the NR1 C-terminus. The
retention motif in NR1 has been shown to prevent unassembled NR1 subunits to exit
the ER (Prybylowski and Wenthold, 2004). The retention can be overcome in NR1
splice variants that express the C2’ cassette, which contains the PDZ-binding motif –
STVV. Assembly with PDZ-containing proteins facilitates the exit of NMDA receptor
from the ER. NR3A does not contain a PDZ-binding motif corresponding to those
found in the NR1 C2’ cassette and NR2 subunits, and no PDZ-containing proteins have
been shown to bind directly to NR3A.
In situ hybridization performed on fetal human CNS tissue revealed that NR3A mRNA
is expressed both in the ventricular zone and cortical plate of the developing cerebral
cortex and in the fetal spinal cord, where the expression was especially prominent in
the dorsal half and around the central canal. Interestingly, in the fetal spinal cord,
NR3A mRNA levels were relatively low in regions with high NR1 expression. This
suggests that NR3A might replace one of the NR1 subunits in immature NMDA
31
receptor complexes during brain development, resulting in receptors less permeable to
Ca2+ and less sensitive to Mg2+ block.
32
STUDY II, MAPPING
Study II describes the expression of NR3A mRNA and protein in adult human CNS
tissue. In addition to studying the spatial distribution of NR3A, we investigated the
association of NR3A with the other NMDA receptor subunits, and examined the
solubility of the subunits. Reverse transcription-polymerase chain reaction (RT-PCR)
and western blot showed that NR3A mRNA and protein is expressed in cortices from
all four cerebral lobes as well as in thalamus and pons of the adult human brain. In the
caudate nucleus, claustrum and cerebellum the levels of NR3A mRNA and protein
were low or undetectable. The spatial distribution of NR3A was confirmed by in situ
hybridization performed on adult human CNS tissue, which together with IHC on
macaque brain revealed that NR3A was mainly present in cortical layers II/III and V.
No NR3A-containing cells could be detected in layer I or in subcortical white matter,
and few NR3A-containing cells were seen in the caudate nucleus. We saw very low
levels of NR3A mRNA and protein in the adult human spinal cord, and in situ
hybridization demonstrated that NR3A mRNA was highly confined to motor neurons
in the ventral horn. RT-PCR and western blot showed that both postnatal and adult rat
spinal cord expressed significant levels of NR3A, and so did embryonic human spinal
cord. Except for the different occurrence of NR3A in adult human and rodent spinal
cord, the spatial distribution of NR3A in adult human CNS resembled what has
previously been published for NR3A in rodent CNS tissue (Al-Hallaq et al., 2002;
Wong et al., 2002).
Adult human brain expresses NR3A mRNA (Mueller and Meador-Woodruff, 2004;
Bendel et al., 2005) but the presence of NR3A protein had not been reported prior to
study II. Recently, a study describing the developmental expression of NR3A mRNA
and protein in human prefrontal cortex was published (Henson et al., 2008). The
authors found the levels of both NR3A mRNA and protein to peak during the first year
of life, decline gradually during the following years and at 25 years of age the levels
were comparatively low, but still higher than during gestation. This implies that the
temporal expression of NR3A in humans is similar to what has previously been
suggested for rodents, with exception for the spinal cord. However, our study revealed
that both adult human and rodent brain express significant levels of NR3A protein, the
levels corresponding to 20-30% of the levels seen during early postnatal life in both
rodents and humans (Al-Hallaq et al., 2002; Wong et al., 2002; Henson et al., 2008).
Accordingly, we conclude that in addition to the developmental processes NR3A has
been implicated in, the subunit has a significant function in adult brain.
Furthermore, we investigated the association of NR3A with the other NMDA receptor
subunits in adult human brain and spinal cord by co-immunoprecipitation. NR3A was
associated with NR1, NR2A and NR2B in brain and to some extent in spinal cord. The
weaker immunoreactive bands seen in western blots of spinal cord samples, most likely
reflect the very low NR3A levels in the tissue. We did not detect any association
between NR3A and GluR2, demonstrating that the interaction between NR3A and the
other NMDA receptor subunits is specific. The incorporation of NR3A in NMDA
receptor complexes in adult CNS further illustrates that NR3A has a functional role in
the adult.
33
We also examined the solubility of NR3A compared to the other NMDA receptor
subunits in both human and rat brain. By centrifugation, we prepared a P2-pellet
containing plasma membranes and membrane-bound proteins, from which we extracted
proteins with different detergents. NR3A showed a remarkably different profile from
the other subunits, being readily extracted by the milder detergents Triton-X 100,
nonidet P-40 (NP-40) and 3-[(3-cholamidopropyl)dimethylammonio]-2-hydroxy-1propanesulfonate (CHAPSO). These milder detergents did not extract detectable levels
of either NR1 or NR2A, which both required sodiumdeoxycholate (DOC) for effective
solubilization. Despite the differences regarding milder detergents, the largest amounts
of NR3A were extracted with DOC treatment. NR3A has been reported to be more
easily extracted from plasma membranes than the other NMDA receptor subunits
(Pérez-Otano et al., 2006). Their study was performed on rat brain, and is in accordance
with what we see for both human and rat preparations. The unusually high solubility of
NR3A indicates that NR3A-containing receptors are less firmly associated with the
PSD, and are probably localized to extrasynaptic sites to a higher degree than NMDA
receptor complexes lacking NR3A. This is interesting since synaptic and extrasynaptic
NMDA receptors are involved in different intracellular protein-interactions and
signaling-pathways. For instance, extrasynaptic NMDA receptors have been shown to
mediate excitotoxic cell death to a higher degree than synaptic NMDA receptors
(reveiwed in Hardinghamn and Bading, 2003)
The solubility of the NMDA receptor subunits was further investigated by size
exclusion chromatography of solubilized plasma membranes. This demonstrated that
NR3A, NR1 and NR2A are all present in large protein complexes, containing both the
subunits themselves and associated proteins. In addition, all three subunits are found in
tetrameric NMDA receptor complexes, but in contrast to NR2A, both NR1 and NR3A
are also found as dimers and monomers. The NR1 subunits have previously been
shown to be localized both to intracellular membranes, were they reside as monomers,
and to the cell surface where they are part of functional NMDA receptor assemblies
(Huh and Wenthold, 1999). The intracellular pool of NR1 has been estimated to contain
60% of the total amount of NR1, and over-expression of NR1 did not have any
significant effect on the number of NMDA receptors present at the cell surface
(Prybylowski et al., 2002). In contrast, only about 10% of NR2A and NR2B subunits
are localized to intracellular pools and over-expression of either of these resulted in an
increase of NMDA receptor subunits at the cell surface, predominantly at extrasynaptic
sites (Prybylowski et al., 2002). Our results suggest that NR3A, just like NR1, is
present as monomers in intracellular pools. Thus, the amount of NR3A subunits is
probably not limiting for the number of NR3A-containig NMDA receptors at the cell
surface.
34
STUDY III, INTERACTION WITH MAP1S/C19ORF5
The cellular response of NMDA receptor activation is mediated by ion fluxes through
the channel pore, as well as by protein-protein interactions. Identifying NR3A-binding
proteins can give insight into which signaling pathways NR3A-containing receptors are
involved in. Apart from mediating cellular responses, protein-protein interactions are
essential for transport of NMDA receptors to appropriate subcellular locations, and the
local trafficking of receptors between synaptic and extrasynaptic sites of dendritic
spines. Both study III and study IV are the result of a yeast two hybrid-screen
performed in order to find novel NR3A-binding proteins. We screened a fetal, human
brain cDNA library, using the intracellular C-terminus of human NR3A as bait. Among
a number of interesting candidates, we chose MAP1S/C19ORF5 and MAP1B for
further investigation, and this is described in study III and IV respectively. We found
MAP1B interesting since it had been shown to be involved in the regulation of other
neurotransmitter receptors (Hanley et al., 1999; Yuen et al., 2005b), and just like
NR3A, it is developmentally regulated with its highest expression levels during
postnatal development (Riederer and Matus, 1985; Safaei and Fischer, 1989). Since
very little is known about the function of MAP1S in the nervous system, we wanted to
investigate this protein and its interaction with NR3A further. Based on the homology
between MAP1S and MAP1B, it could be envisioned that they are involved in similar
mechanisms regarding NR3A.
The 3’ part of MAP1S cDNA, encoding amino acids 705-1059/stop was isolated in the
screen, and the interaction between MAP1S and NR3A was verified in vitro by GST
pull-down experiments. We determined that the interaction between MAP1S and
NR3A also takes place in the brain, by demonstrating co-immunoprecipitation from
solubilized rat brain membranes. In addition, we showed that the membrane-proximal
part of the NR3A C-terminus is required for the interaction with MAP1S.
There are a limited number of publications on MAP1S and only one of them concerns
neuronal tissue (Orbán-Németh et al., 2005). This led us to investigate the occurrence
of MAP1S in different organs and brain regions. We found MAP1S to be highly
expressed in cerebral cortex and cerebellum of both adult and eight day-old rats. Adult
rat also expressed MAP1S in testis, heart and skeletal muscle and postnatal rat
expressed MAP1S in heart, muscle and liver. Interestingly, in brain and testis the posttranslationally cleaved heavy chain dominated over the full-length MAP1S,
distinguishing them from the other organs. In addition, we saw that the levels of
MAP1S are higher in adult than in embryonic human forebrain and higher in postnatal
than adult rat forebrain. However, the heavy chain predominated over the full-length
MAP1S in all forebrain samples, and the dominance was increased with age. MAP1B
is almost exclusively expressed in its cleaved form, and the fact that we detect so much
full-length MAP1S suggests that the posttranslational cleavage of MAP1B and MAP1S
is regulated differently. The MAP1B heavy chain has been shown to associate with 2 to
8 light chains, which suggests that the MAP1B light chain has a longer half-life than
the heavy chain since they are synthesized in a 1:1 ratio. (Mei et al., 2000a; Mei et al.,
200b). The ratio between MAP1S heavy and light chain has not been investigated so far
35
and it is not clear if the function of full-length, uncleaved protein is different from that
of cleaved and re-assembled protein.
Next we compared the levels of MAP1S in human cortical cells cultured under
conditions adapted to obtain cultures of mainly neurons, or cultures of mainly glial
cells. We found the MAP1S levels to be higher in neurons than in glial cells. We also
examined the occurrence of MAP1S in different CNS regions and in dorsal root ganglia
of postnatal rat, and found MAP1S to be expressed in all regions examined. Previous
publications on MAP1S have focused on its role in regulation of cell cycle events and
cell death (Liu et al., 2005b; Liu et al., 2005a; Song et al., 2005). Therefore, it is of
significant interest to find that the MAP1S levels are high in post-mitotic neurons, a
fact that opens for novel functions of MAP1S, one being regulation of NR3Acontaining NMDA receptors.
We studied the subcellular distribution of MAP1S in primary rat hippocampal and
human cortical cells cultured for 14 DIV. Due to different fixation requirements, we
were not able to successfully perform double-staining with the MAP1S antibody.
Therefore, we chose to transfect the neurons at 10 DIV with EGFP-tagged MAP1S, and
after fixation at 14 DIV we performed immunocytochemistry (ICC) on the cells. We
found MAP1S-EGFP to be localized throughout dendritic shafts, and the distribution
resembled that of β-tubulin III immunoreactivity. But, MAP1S-EGFP was also
localized to filopodia-like protrusions which lacked β-tubulin III immunoreactivity.
Since filopodia and spines contain little or no microtubules, MAP1S in these structures
might be associated with actin, NR3A or other so far unidentified binding-partners.
Moreover, we found a partial colocalization of MAP1S-EGFP and the presynaptic
marker synapsin and postsynaptic marker PSD95, indicating that MAP1S can be
localized to synaptic structures. MAP1S-EGFP and NR3A immunoreactivity
colocalized in dendritic shafts and occasionally in filopodia-like processes. An
interaction between MAP1S and NR3A in dendritic shafts might involve regulation of
the transport of NR3A-containing NMDA receptors along microtubules. However, the
interaction between the two proteins in filopodia or spines, more likely involves
regulation of expression of NR3A-containing NMDA receptors at synaptic and
extrasynaptic sites.
36
STUDY IV, INTERACTION WITH MAP1B
Study IV focuses on the interaction between NR3A and MAP1B. MAP1B was
identified as a potential binding-partner of NR3A in the yeast two-hybrid screen
described above. The interaction between NR3A and MAP1B was verified in vitro by
GST pull-down experiments and in vivo by co-immunoprecipitations from solubilized
rat brain membranes. Moreover, the microtubule-binding region of the MAP1B light
chain was shown to be required for the interaction with NR3A.
Truncation analysis showed that the microtubule-binding region of the MAP1B light
chain is required for the interaction with NR3A.
To examine the subcellular distribution of MAP1B and NR3A we performed ICC on
human cortical cells cultured for 21 DIV. This analysis showed that MAP1B was
highly colocalized with both β-tubulin III and actin in dendritic shafts and to some
extent with actin in filopodia. Colocalization between MAP1B and the synaptic
proteins synapsin and SAP102 was sparse. The infrequent occurrence of MAP1B
immunoreactivity at synaptic sites of cultured neurons is in accordance with two
recently published papers (Davidkova and Carroll, 2007; Kitamura et al., 2007).
However, a much higher presence of MAP1B in synapses was suggested in an electronmicroscopy study on adult rat brain, estimating MAP1B to be present in as many as
50% of cerebral cortex synapses (Kawakami et al., 2003). NR3A immunoreactivity was
frequently seen throughout dendritic shafts where it colocalized with β-tubulin III
immunoreactivity, but also in actin-containing filopodia and spines. The distribution of
NR3A and MAP1B resembled each other, consequently colocalization between NR3A
and MAP1B immunoreactivity was more frequently seen throughout dendritic shafts,
than in filopodia-like processes.
Through truncation-analysis of MAP1B we determined the microtubule-binding region
in MAP1B-LC1 to be required for the interaction with NR3A. Thus, it is possible that
by binding to MAP1B-LC1, NR3A blocks the interaction between MAP1B-LC1 and
microtubules. However, since the MAP1B-HC contains an additional microtubulebinding region, MAP1B might be able to bind to microtubules and NR3A
simultaneously.
To learn more about the function of the interaction between NR3A and MAP1B, we
investigated the protein levels of NR3A and NR1 in MAP1B deficient mice. Compared
to WT mice, the levels of NR3A were increased and the levels of NR1 decreased in
MAP1B deficient animals, with a 35% increase in the NR3A/NR1 ratio. We did not see
any effect on GluR2 protein levels, indicating that the absence of MAP1B does not
have a general effect on glutamate receptors but is specifically affecting the NMDA
receptor subtype. The fact that NR3A and NR1 seem to be mutually regulated
strengthens our hypothesis that NR3A can replace at least one of the NR1 subunits in
NMDA receptor assemblies. The elevated NR3A levels in MAP1B deficient mice
might result in an increased amount of NR3A-containing NMDA receptors at the cell
surface, which would lead to an attenuation of NMDA receptor signaling in these
animals.
37
Next we studied the subcellular distribution of NR3A in 21 DIV cultured hippocampal
neurons from MAP1B deficient and WT mice. We visualized filopodia/spines by
phalloidin staining of actin filaments and counted the number of NR3Aimmunoreactive filopodia/spines. The fraction of filopodia/spines containing NR3A
was equal in MAP1B deficient and WT animals, which implies that MAP1B is not
required for transport of NR3A-containing NMDA receptors to filopodia/spines.
Instead we postulate that MAP1B might be involved in regulating the expression of
NR3A-containing receptors at the cell surface. The subcellular distribution of both
GABAc and 5-HT1A and 5-HT3 is regulated by MAP1B (Hanley et al., 1999; Yuen et
al., 2005b; Sun et al., 2008) and similar mechanisms might underlie the regulation of
NR3A-containing NMDA receptors.
Finally, we compared the microtubular-binding properties of MAP1S and MAP1B. We
found that both MAP1S and MAP1B bind strongly to paclitaxel-stabilized
microtubules, showing that the microtubule-binding properties of MAP1S and MAP1B
are shared. Study III and IV suggest that MAP1S and MAP1B have similar functions in
neurons, one being involvement in the regulation of NR3A-containing NMDA
receptors.
38
CONCLUSIONS AND FUTURE PERSPECTIVES
The homology between the rat and human NMDA receptor subunit NR3A is high, 93%
at the amino acid level. The spatial and temporal distribution of NR3A is similar in
mammals, with the exception of the spinal cord and cerebellum. Adult rats express
comparable NR3A levels in the spinal cord and cerebral cortex, whereas NR3A levels
in the adult human spinal cord have declined markedly, compared to earlier
developmental stages and to the levels in the adult cerebral cortex. Moreover, we could
not detect NR3A mRNA or protein in adult human cerebellum, in contrast to earlier
studies reporting expression of NR3A in rodent cerebellum.
NR3A is less firmly bound to plasma membranes than the other NMDA receptor
subunits. Just as NR1, NR3A is incorporated into tetrameric NMDA receptor
complexes but also exists as dimers and monomers. Both NR1 and NR3A can bind
glycine and it is likely that NR3A replaces one of the NR1 subunits in certain NMDA
receptor complexes, especially during development when the NR3A levels are high.
NR3A binds to the C-terminal regions of MAP1S and MAP1B. Both MAPs are highly
expressed in neurons, with prominent distribution throughout dendritic shafts and with
occasional localization to dendritic filopodia/spines. In addition to the previously
reported interaction with actin, we showed that MAP1S and MAP1B both bind to
stabilized microtubules. Little is known about the function of MAP1S in the nervous
system, whereas MAP1B is known to be essential for normal brain development and is
involved in axon guidance and in myelinization, both in the CNS and PNS. Moreover,
MAP1B is involved in the regulation of several neurotransmitter receptors, such as
GABAc and 5-HT.
Based on our findings in study IV, I find it unlikely that MAP1B is involved in
transport of NR3A-containing NMDA receptors along microtubules. I favor the idea
that interaction between the MAPs and NR3A is involved in regulation of the local
distribution of NR3A-containing NMDA receptors between intracellular pools and the
plasma membrane and/or the distribution between synaptic and extrasynaptic sites. To
see whether the MAPs have an effect on the number of NR3A-containing receptors at
the cell surface, the MAPs could either be over-expressed in heterologous cells, or
knocked down by antisense treatment of neurons. The cells could then be analyzed by
ICC.
When analyzing the distribution of NR3A in neurons from MAP1B deficient and WT
mice, we found that the localization of NR3A to filopodia was not affected by the lack
of MAP1B. However, it would be interesting to examine the morphology of the NR3Acontainining spines in MAP1B deficient and WT mice. Dendritic spines in the MAP1B
deficient mice have a more immature morphology than spines in WT mice (E. Tortosa,
unpublished observation). NR3A is associated with development and might not be
equally distributed between immature filopodia and more mature, mushroom shaped
spines. As a complement to the morphological examination, it would be interesting to
stain the cells for synaptic proteins, for example PSD95 and synapsin and compare
their localization to the localization of NR3A immunoreactivity.
39
The MAP1B deficient mice have increased NR3A protein levels. However, at the
moment it is not clear if these additional NR3A subunits are part of functional NMDA
receptor assemblies expressed at the cell surface. Since the presence of NR3A in the
NMDA receptor complex attenuates receptor currents, this might be elucidated by
electrophysiological measurements. Decreased NMDA receptor currents would
indicate the presence of NR3A in receptor assemblies at the cell surface, while currents
similar to those in WT cells would suggest that the excess NR3A seen in MAP1B
deficient animals is mainly localized to intracellular pools. More than half of the NR1
subunits expressed in a neuron have been estimated to be localized to intracellular
pools, where they have a high turnover rate compared to NR1 subunits incorporated
into receptors and expressed at the cell surface. Our data suggests that NR3A has a
similar distribution between intracellular membranes and the cell surface as NR1.
Both MAPs bind to actin and to microtubules, and their distribution is dependent on
cytoskeletal integrity. If the MAPs are associated with the cytoskeleton when
interacting with NR3A, cytoskeletal depolymerization would also affect the distribution
of NR3A. This could be investigated by exposing cultured neurons to actin or
microtubule-depolymerizing drugs and by ICC determine if the distribution of the
proteins is altered.
The interaction between NMDA receptor subunits and intracellular proteins are often
activity dependent. For instance, NR3As interaction with both PACSIN1 and PP2A has
been shown to be regulated in an activity-dependent manner. Whether there is such a
relationship between NR3A and its interaction with the MAPs could be tested by
treating cultured neurons with NMDA receptor agonists or antagonists. The effect of
the treatment could then be evaluated by co-immunoprecipitation and ICC.
The studies I have presented are part of the characterization of NR3A and NR3Ainteracting proteins, performed both by us as well as other research groups. In this
thesis I discuss the role of NR3A-interacting proteins in receptor transport, trafficking
and signaling. It is my belief that these efforts will give us a greater understanding of
NR3A, and possibly also NR3B, which both have profound effects on NMDA
receptors. When such understanding is reached, we will also be able to evaluate NR3Arelated processes in the brain and spinal cord, and their influence on health and disease.
40
MATERIALS AND METHODS
HUMAN TISSUE (PAPERS I, II, III AND IV)
Adult human post mortem CNS tissue, from individuals without any record of a
neurological or psychiatric disorder, was provided by Netherlands Brain Bank (papers II
and III) and by Huddinge Brain Bank, Sweden (paper II). Adult tissue was used for in
situ hybridization, IHC, immunoprecipitation and western blot in paper II and for
western blot in paper III. Embryonic human CNS tissue was collected at elective,
routine, first trimester abortions and written consent had been given by the pregnant
women. The tissue was used for reverse transcription-polymerase chain reaction (RTPCR) and in situ hybridization in paper I, for RT-PCR in paper II, for western blot in
paper III and culturing cells used for ICC in papers III and IV. The use of human tissue
was performed in compliance with Dutch and Swedish law and the procedures were
approved by the Regional Ethical Committee in Stockholm.
ANIMAL TISSUE (PAPERS I, II, III AND IV)
Eight-day old Sprague-Dawley rats were decapitated, CNS tissue dissected and used for
RT-PCR in papers I and II, for western blot and solubilization-experiments in paper II,
for immunoprecipitation and western blot in papers III and IV and for microtubulebinding experiments in paper IV. Adult rats were anestitized, decapitated, CNS tissue
dissected and used for RT-PCR in paper II and for western blot in papers II and III.
Hippocampi were dissected from embryonic day 18 rats and dissociated cells cultured
and used for ICC in papers III and IV. The handling and procedures were approved by
the Ethical Committee on Animal Research in Southern Stockholm.
Female macaque monkeys ranging from 6 months to 13 years of age were used for IHC
in paper II. The care of the animals and all anesthesia and euthanasia procedures in this
study were performed according to the National Institutes for Health Guide for the Care
and Use of Laboratory Animals, and were approved by the Institutional Animal Care
and Use Committee at Emory University, Atlanta USA.
Cortices from newborn Swiss-Webster mice were dissected, the cells dissociated and
cultured under conditions to form a monolayer of astrocytes. Hippocampi were
dissected from 18 day-old WT and MAP1B deficient R1/NMRI mouse fetuses, the
dissociated cells plated on cover slips and after adhering combined with the astrocyte
monolayer. The hippocampal cells were used for ICC in paper IV. Newborn WT and
MAP1B deficient R1/NMRI mice were decapitated, the brains dissected and used for
western blot in paper IV. The handling and procedures were approved by the Ministry
of Agriculture, Spain.
HUMAN BRAIN CDNA LIBRARIES (PAPER I)
We isolated the first fragment of human NR3A from an adult, human hippocampal
cDNA library by PCR-amplification using degenerate primers. The isolated fragment
was highly homologous to the rat NR3A sequence and used as a template to synthesize
a digoxigenin (DIG)-labeled probe. The probe was then used to screen a fetal, human
brain cDNA library, and an additional part of the human NR3A sequence obtained.
41
However, the full-length human NR3A cDNA was not retrieved from the cDNA
libraries.
REVERSE TRANSCRIPTION-POLYMERASE CHAIN REACTION (RT-PCR)
(PAPERS I AND II)
Complementary DNA (cDNA) was reverse transcribed from mRNA prepared from rat
or human brain CNS tissue, by gene specific primers. In paper I, the full-length human
NR3A cDNA was amplified by PCR and then sequenced. The cDNA obtained was also
used to investigate the occurrence of NR3A splice variants in both paper I and II. In
paper II we also used this method to investigate in which regions of the adult human
brain NR3A mRNA was present.
SUBCLONING, MUTAGENESIS AND SEQUENCING (PAPERS I, III AND IV)
In paper I the full-length human NR3A cDNA was obtained by RT-PCR and the
nucleotide sequences determined. In papers III and IV different DNA constructs have
been prepared by subcloning NR3A, MAP1S and MAP1B cDNA into yeast, bacterial
and eukaryotic expression vectors. Some of the DNA constructs were further modified
by inserting mutations into their nucleotide sequence by PCR-based mutagenesis. The
sequence of all prepared DNA constructs were verified by sequence-analysis.
IN SITU HYBRIDIZATION (PAPERS I AND II)
To investigate the expression of NR3A mRNA in embryonic human CNS tissue in
paper I, we performed in situ hybridization using [35S]-labeled oligonucleotide probes.
In paper II we investigated the expression of NR3A mRNA in different regions of the
adult human brain using first a full-length rat NR3A (rNR3A) [35S]-riboprobe and then
verifying the result using a mixture of two human-specific [35S]-labeled NR3A
oligonucleotide probes. The [35S]-labeled sections were exposed to film for 7-50 days,
depending on the strength of the signal and then developed. As negative control,
sections were hybridized in the presence of a 100-fold excess of unlabeled probe. For
visualization of cells, a week counterstaining with cresol violet was used.
IMMUNOHISTOCHEMISTRY (PAPER II)
To investigate the expression of NR3A protein in brains from higher mammals we
performed IHC on macaque monkeys using a NR3A antibody. NR3A-labeled brain
sections were incubated with biotinylated goat anti-rabbit IgG and visualized using 3,3'diaminobenzidine (DAB) as chromogen. Control sections, processed as above except
for the omission of the primary immunoreagent, did not contain DAB label.
SOLUBILIZATION OF RECEPTOR SUBUNITS (PAPER II)
We investigated the solubility of the NR3A subunit, by preparing a membrane fraction
(P2-pellet) of rat or human brain tissue by centrifugation and then extracting
membrane-bound proteins from the fraction. We used different detergents, varying in
their strength and a total of six different conditions for the extractions. We loaded equal
volumes of solubilized protein on SDS-gels and compared the occurrence of NR3A
protein between the different extraction conditions. We also investigated which
extraction conditions solubilized the other NMDA receptor subunits. To avoid that
42
solubility of other cellular proteins would affect our results, the same volume of each
supernatant, rather than a fixed amount of total solubilized protein, was loaded in each
well. This will more accurately allow a comparison of solubility.
SIZE EXCLUSION CHROMATOGRAPHY AND SPOT BLOT (PAPER II)
To analyze the presence of NR3A in protein complexes we separated the membrane
fraction (P2-pellet), solubilized with either the mild detergent CHAPSO or with
CHAPSO followed by the stronger detergent DOC, on a Suprose 6 column. The
solubilization buffer was used to elute protein monomers, dimers, tetramers and larger
complexes from the column. High molecular weight complexes passed through the
column at higher speed than low molecular weight complexes and were accordingly
collected in earlier fractions. The fractions were filtered through a nitrocellulose
membrane and the membranes probed for NMDA receptor subunit antibodies to
determine which subunits were present in different fractions. The protein spots were
detected using a CCD-camera and a standard set of known proteins were used to
identify the molecular weights of proteins in the different fractions.
PREPARATION OF TISSUE AND WESTERN BLOT (PAPERS II, III AND IV)
Human or animal CNS tissue was homogenized in cold buffer and protein
concentrations determined. Samples were separated by SDS-PAGE on Tris-glycine
gels, transferred to nitrocellulose membranes, incubated with primary antibody over
night and with secondary antibody for 1 hr, both at room temperature, se table 2. In
paper II protein bands were detected on film and in papers III and IV with a CCDcamera. In papers III and IV the optical density of protein bands were determined and
quantified. In paper IV the relative optical density of protein bands from MAP1B
deficient animals, compared to WT animals was calculated and values analyzed with
Mann-Whitney U-test. α-Tubulin was used as an internal control to ensure that the
same amount of protein was loaded in all wells.
CO-IMMUNOPRECIPITATION (PAPERS II, III AND IV)
To determine if two proteins can bind to each other in vivo we performed a number of
co-immunoprecipitations, from adult human brain in paper II and from eight-day old rat
brain in papers III and IV. In either case, a membrane fraction (P2-pellet) was prepared
by centrifugation and then solubilized with DOC at pH 9. To remove excess detergent,
solubilized samples were dialyzed over night and also diluted 1:10 before use for
immunoprecipitation. After preclearing, the samples were incubated with primary
antibody over night, precipitated with Dynabeads and the precipitated and
coprecipitated proteins analyzed by western blot.
YEAST TWO-HYBRID SCREEN (PAPERS III AND IV)
To find novel proteins interacting with NR3A we performed a yeast two-hybrid screen
of a fetal, human brain cDNA library. As bait, we used the full-length intracellular Cterminus of human NR3A, comprising amino acids 952-1115. The initial screen
resulted in more than 88 interacting clones. To reduce the number of false positive
binding-partners, all clones were exposed to more stringent selection conditions, which
resulted in 10 remaining clones. These clones were sequenced and one of them encoded
MAP1S and three encoded MAP1B.
43
GST PULL-DOWN (PAPERS III AND IV)
To determine whether the different clones interacting with NR3A in yeast, also bind to
NR3A in a different in vitro system, we performed glutathione-S-transferase (GST)
pull-down experiments. In addition, in paper IV we performed GST pull-down
experiments with truncated MAP1B fragments, to determine which part of MAP1B is
required for binding to NR3A.
CELL CULTURES, TRANSFECTION AND IMMUNOCYTOCHEMISTRY
(PAPERS III AND IV)
HEK 293 cells were either transiently or stably transfected with NR3A cDNA. The
cells were lysed on ice with Triton X-100 and NP-40, unsolubilized material pelleted
and a known amount of total protein used for GST pull-down experiments.
The subcellular distribution of NR3A, MAP1S and MAP1B was investigated by
immunocytochemical staining of primary neurons cultured in Neurobasal media
supplemented with glutamine, glutamate and B27. In paper III we used rat hippocampal
and human cortical cells transfected with MAP1S-EGFP at10 DIV and fixed at 14 DIV.
The localization of MAP1S-EGFP was then compared to that of β-tubulin III, the
presynaptic marker synapsin, the postsynaptic marker PSD95 and NR3A. In paper IV
we investigated the location of MAP1B and NR3A in relation to β-tubulin III, actin,
synapsin and the postsynaptic protein SAP102 in human cortical cells cultured for 21
DIV. In paper IV we also examined the distribution of NR3A in hippocampal neurons
from MAP1B deficient and WT mice cultured for 21 DIV. The actin cytoskeleton was
visualized by Alexa Fluor® 488-conjugated phalloidin, and the number of NR3A
immunoreactive filopodia counted.
STABILIZATION OF MICROTUBULES (PAPER IV)
To investigate the microtubule-binding properties of MAP1S, we treated rat brain
homogenate with the microtubule-stabilizing drug paclitaxel. The stabilized
microtubules were then pelleted by high-speed centrifugation and the distribution of
MAP1S between pellet and supernatant analyzed by western blot. As comparison, we
looked at the distribution of MAP1B in the different fractions, since the microtubulebinding property of MAP1B is well documented.
44
Table 2. Primary antibodies employed in the studies.
Antibody
Species
Dilution WB
Dilution ICC
Supplier
Actin, AC-40
Mouse
1:1 000
-
Abcam
α-tubulin, YL1/2
Rat
1:2 000
-
Abcam
β-tubulin type III
Rabbit
1:2 000
1:500
Biosite
β-tubulin type III,
Mouse
-
1:1 200
Sigma
GFAP
Rabbit
1:1 000
-
Promega
GluR2, MAB397
Mouse
1:500
-
Chemicon
GST
Goat
1:10 000
-
GE Healthcare
MAP1B, AA6
Mouse
1:500
1:300
Abcam
MAP1B
Mouse
1:250
-
BD Pharmingen
MAP1S, mAb4G1
Mouse
1:2 000
-
(Liu et al., 2005b)
NR1, 54.1
Mouse
1:500
1:300
BD Pharmingen
NR2A
Rabbit
1:500
-
Merck Biosciences
NR2B
Rabbit
1:500
-
Invitrogen
NR3A
Rat
1:4 000
1:500
Custom made by Zymed Invitrogen
PSD95, K28/43
Mouse
-
1:500
SDL.3D10
Corporation
Upstate Cell Signaling Solutions
Abbreviations; Western blot (WB) and Immunocytochemistry (ICC).
45
ACKNOWLEDGEMENTS
I would like to thank the following people for help and support during my postgraduate
studies:
Erik Sundström, my main supervisor, for guiding and supporting me through these
years. You have thought me project-planning and critical thinking. The fact that you
have trusted me to plan and organize my projects and to try my own ideas has been
very important to me. At the same time you have always been there to give advice and
to discuss my good and bad suggestions, and this has been essential both to the project
and me.
Eirikur Benedikz, my cosupervisor for always being there and taking the time to
discuss big problems as well as small, regarding science and life. Your techniqual
knowledge and critical feedback has been invaluable.
Åke Seiger, head of our department. You have been a great source of inspiration and
your enthusiasm is contagious. At our annual lab-day meetings you always bring fresh
views and ideas to our projects and plans.
Elisabet Åkesson, for being such a knowledgeable and enthusiastic person, always
enriching our discussions, and also for being a great “förebild” giving advice on
everything from science to handling children, husbands and supervisors. I enjoyed
teaching together with you and learnt a great deal from both the classes and you. Thank
you for reading this thesis and giving me constructive feed back.
Jenny Odeberg and Mia Emgård for sharing your knowledge and experience and giving
me feedback on my ideas. Your participation has meant a lot to the scientific climate in
our group and your positive and friendly attitudes have chaired everyone up.
Anna Öhman and Jinghua Piao, the other two Ph D students in our group, Anna who is
soon defending her thesis and Dr. Piao who did an excellent job at her defense a few
months back. The past years we have not only shared rooms but also the ups and downs
of research and life as a Ph D student. Thanks for all the fun we have had both in the
lab and on conferences in Sweden and abroad. Good luck to both of you on future
positions and in life!
Eva-Britt Samuelsson, LiLi Mo and Lena Holmberg, our outstanding technicians. Your
long experience of lab work and different techniques is an invaluable source of
knowledge in the group. Thanks also for organizing and convincing the rest of us to
participate in everything from KI-loppet and Blodomloppet to guided tours of
Stockholm. LiLi, we miss you and hope you will be back with us soon!
Per-Henrik Anderson and Helena Aineskog, the two newest members of our group. PH,
thanks for being an invaluable resource at the lunch table, sorting out the different seeds
and weeds we consume and for professional advice on alfa-alfa seed culturing. Helena
thanks for teaching me everything there is to know about primary urine, and more
46
importantly, for giving me supportive and positive feedback at times when I have
needed it. I suspect that both of you are slightly crazy and this will most likely be of
great value for surviving in science, good luck!
Helena Samuelsson and Stefan Björklund, who have worked together with me while
doing their master thesis projects in our group. Helena, you impressed me with how
easily you comprehended new knowledge and methods and it was a pleasure to work
with someone so talented and skillful as you. I understand that you are happy at your
new job at AstraZeneca, but we still miss you in the group! Stefan, it has been a lot of
fun working with you, I have learnt a lot and I hope you have too. I think your
determination, positive attitude and careful planning will bring you far. Good luck with
your future positions and with your training-program to obtain perfect body shape and
composition! And a huge thanks for helping me with the references for this thesis after
the Endnote break down!
Ronnie Folkesson, though you have not been my formal supervisor you spent a lot of
time teaching me when I was new in the lab. Even after those first months we spent in
the inspiring cellar environment of the labs underneath the hospital, you have always
been supportive and helpful, sharing experience and knowledge.
My coauthors on study I Dr. Susanne Froelich-Fabre and Jenny Dunker, and Dr. Chris
Muly and Dr. Nenad Bogdanovic on study II. Dr. Leyuan Liu and Professor Wallace L
McKeehan, with whom we cooperated on study III. Thanks for sharing your experience
and knowledge on MAP1S/C19ORF5, as well as on space travel. Professor Jesus
Avila, Dr. Filip Lim and Elena Tortosa, with whom we cooperated on study IV. Thanks
for letting me visit your lab, and for taking so well care of me during my visit. Your
knowledge on MAP1B has been invaluable to this project. Thanks also to all the nice
and friendly people in Jesus Avila’s group, who made my visit such a pleasant one.
Your Christmas dinner was fabulous and the turon has provided me with all the sugar I
needed to write this thesis!
Dr. Patrick Allan, Dr. Haleh Razani and Derek Haspeslagh at the Connecticut Mental
Health Center, in New Haven, USA. Thanks for letting me visit your lab and for taking
the time to share your knowledge on phosphorylation. I enjoyed our discussions and
learnt a lot during my stay. Thanks also to Dr. Peter Olausson and Dilja Krueger for
sharing your monkey brain samples with us, even though they ended up as “results not
shown”. Haleh, thanks for inviting me to your great house-warming party and for
taking me to some of New Havens nice restaurants.
The senior scientist in the lab for creating a good scientific atmosphere: Marianne
Schultzberg, Zhu Jie, Bengt Winblad, Maria Ankarcrona, Jan Näslund, Caroline Graf,
Helena Karlström, Lars Tjernberg, Jing-Jing Pei, Susanne Frykman, Homira
Behbahani, Håkan Thonberg. Special thanks to Angel Cedazo-Minguez for reading this
thesis and giving me constructive feed back.
Former and present Ph D students and post docs, thanks for all the nice company and
discussions, during lunches and in the gym.
47
The administration staff, especially Jessica Asplund, for your fantastic helpful and
positive attitude, even when there has been something wrong with my invoices! Monica
Erlandsson at Stockholms Sjukhem, for being so patient with the absent-minded people
in the lab. I suspect organizing us is far from easy. Kristina Lundh for all help with
LADOK, especially at the “alla kurser klara” stage.
My classmates from the biomedical research school and our coordinator Eva
Severinsson. Thanks for an excellent year, a fantastic trip to Iceland, Canada and USA
and for our monthly pub-evenings. I’m glad I still have contact with so many of you!
Thanks also to all the nice people the year ahead of me, especially Magnus, and all the
fun at the SNiB conferences.
Family and friends: First of all my Mother Ann-Marie, for your support and for always
taking the time to listen, I do not know how this would have gone without you. Kalle,
for encouraging me to go to the gym and to look after myself. My terrific sister
Karolina and her Jonas. Helena and Johanna, for bringing some culture into my life
every now and then. Hasse and Kersti, Martina and Jonas, for all nice talks and
discussions, and for all our wonderful midsummer parties on Gräskö. My Father
Ronald, Dawn and the little fellows David and Daniel. André for taking care of me
while visiting in the US. I miss our discussions and have heaps of new topics ready for
you! Thanks to Ann-Marie and Karolina for feed back on the Swedish introduction.
Holger and I thank Grandmother Ann-Marie, Grandfather Hasse, and Aunt Johanna for
picking Holger up at kindergarten so often. Without your help there would not have
been any thesis this year. And probably a lot less dinners in front of Bolibompa, so I
think both Holger and I have been quite happy with the arrangement.
Anna, I know its difficult to believe but this autumn it will be 20 years since we started
in the same school and we have experienced a lot together. Thank you for being such a
fantastic friend. Linus and Emelie for all the fun, especially during summer holidays.
Louise, Maria S, Maria K, Camilla, Anna P, Maggan and Pia. With a home full of
crazy boys our girl-nights have been essential for my well-being. Louise and Anders for
an unforgettable trip to Nepal. Thanks for taking care of us Louise, and for being
terrific company during our trek in Spain.
Patrick, Maria L, Peter, Hanna, Julian and Maria N, for all nice discussions at our
Novum-lunches. Dating you has chaired me up during long weeks in the lab.
Anders, Maria and Jakob, Mathias, Alice and Hilding, for all nice walks and talks,
dinners and discussions, midsummer and New Year parties.
Henrik and Holger my favorite boys. Henrik, for constructive feed back on the list of
abbreviations, it really took the thesis to a new level! But more importantly, for
struggling with me during these years. Yes, you may carry my suitcase next time we
travel as well. Holger, for giving me a new perspective on life and for always trusting
me to manage every situation with your ”Det kan du, mamma!”.
48
REFERENCES
Aarts M, Liu Y, Liu L, Besshoh S, Arundine M, Gurd JW, Wang YT, Salter MW,
Tymianski M (2002) Treatment of ischemic brain damage by perturbing
NMDA receptor- PSD-95 protein interactions. Science (New York, NY
298:846-850.
Adams JP, Sweatt JD (2002) Molecular psychology: roles for the ERK MAP kinase
cascade in memory. Annu Review Pharmacology Toxicol 42:135-163.
Adams SL, Foldes RL, Kamboj RK (1995) Human N-methyl-D-aspartate receptor
modulatory subunit hNR3: cloning and sequencing of the cDNA and primary
structure of the protein. Biochimica et biophysica acta 1260:105-108.
Al-Hallaq RA, Jarabek BR, Fu Z, Vicini S, Wolfe BB, Yasuda RP (2002) Association
of NR3A with the N-methyl-D-aspartate receptor NR1 and NR2 subunits. Mol
Pharmacol 62:1119-1127.
Allen E, Ding J, Wang W, Pramanik S, Chou J, Yau V, Yang Y (2005) Gigaxonincontrolled degradation of MAP1B light chain is critical to neuronal survival.
Nature 438:224-228.
Allen PG, Shah JV (1999) Brains and brawn: plectin as regulator and reinforcer of the
cytoskeleton. Bioessays 21:451-454.
Anantharam V, Panchal RG, Wilson A, Kolchine VV, Treistman SN, Bayley H (1992)
Combinatorial RNA splicing alters the surface charge on the NMDA receptor.
FEBS letters 305:27-30.
Andersson O, Stenqvist A, Attersand A, von Euler G (2001) Nucleotide sequence,
genomic organization, and chromosomal localization of genes encoding the
human NMDA receptor subunits NR3A and NR3B. Genomics 78:178-184.
Antar LN, Li C, Zhang H, Carroll RC, Bassell GJ (2006) Local functions for FMRP in
axon growth cone motility and activity-dependent regulation of filopodia and
spine synapses. Mol Cell Neurosci 32:37-48.
Awobuluyi M, Yang J, Ye Y, Chatterton JE, Godzik A, Lipton SA, Zhang D (2007)
Subunit-specific roles of glycine-binding domains in activation of NR1/NR3 Nmethyl-D-aspartate receptors. Mol Pharmacol 71:112-122.
Bading H, Greenberg ME (1991) Stimulation of protein tyrosine phosphorylation by
NMDA receptor activation. Science (New York, NY 253:912-914.
Bendel O, Meijer B, Hurd Y, von Euler G (2005) Cloning and expression of the human
NMDA receptor subunit NR3B in the adult human hippocampus. Neurosci Lett
377:31-36.
Bernardet M, Crusio WE (2006) Fmr1 KO Mice as a Possible Model of Autistic
Features. Scientif World J 6:1164-1176.
Boeckers TM (2006) The postsynaptic density. Cell and Tissue Res 326:409-422.
Boeckers TM, Kreutz MR, Winter C, Zuschratter W, Smalla KH, Sanmarti-Vila L,
Wex H, Langnaese K, Bockmann J, Garner CC, Gundelfinger ED (1999)
Proline-rich synapse-associated protein-1/cortactin binding protein 1
(ProSAP1/CortBP1) is a PDZ-domain protein highly enriched in the
postsynaptic density. J Neurosci 19:6506-6518.
Bouras C, Kovari E, Hof PR, Riederer BM, Giannakopoulos P (2001) Anterior
cingulate cortex pathology in schizophrenia and bipolar disorder. Acta
Neuropathol (Berl) 102:373-379.
Bowser R, Muller H, Govindan B, Novick P (1992) Sec8p and Sec15p are components
of a plasma membrane-associated 19.5S particle that may function downstream
of Sec4p to control exocytosis. J Cell Biol 118:1041-1056.
Braff DL, Grillon C, Geyer MA (1992) Gating and habituation of the startle reflex in
schizophrenic patients. Arch Gen Psychiatry 49:206-215.
Brenman JE, Christopherson KS, Craven SE, McGee AW, Bredt DS (1996a) Cloning
and characterization of postsynaptic density 93, a nitric oxide synthase
interacting protein. J Neurosci 16:7407-7415.
Brenman JE, Chao DS, Gee SH, McGee AW, Craven SE, Santillano DR, Wu Z, Huang
F, Xia H, Peters MF, Froehner SC, Bredt DS (1996b) Interaction of nitric oxide
49
synthase with the postsynaptic density protein PSD-95 and alpha1-syntrophin
mediated by PDZ domains. Cell 84:757-767.
Brenman JE, Topinka JR, Cooper EC, McGee AW, Rosen J, Milroy T, Ralston HJ,
Bredt DS (1998) Localization of Postsynaptic Density-93 to dendritic
microtubules and interaction with Microtubule-Associated Protein 1A. J
Neurosci 18:8805-8813.
Brody SA, Nakanishi N, Tu S, Lipton SA, Geyer MA (2005) A developmental
influence of the N-methyl-D-aspartate receptor NR3A subunit on prepulse
inhibition of startle. Biol Psychiatry 57:1147-1152.
Brose N, Gasic GP, Vetter DE, Sullivan JM, Heinemann SF (1993) Protein chemical
characterization and immunocytochemical localization of the NMDA receptor
subunit NMDA R1. J Biol Chem 268:22663-22671.
Caviston JP, Holzbaur EL (2006) Microtubule motors at the intersection of trafficking
and transport. Trends Cell Biol 16:530-537.
Chan SF, Sucher NJ (2001) An NMDA receptor signaling complex with protein
phosphatase 2A. J Neurosci 21:7985-7992.
Chatterton JE, Awobuluyi M, Premkumar LS, Takahashi H, Talantova M, Shin Y, Cui
J, Tu S, Sevarino KA, Nakanishi N, Tong G, Lipton SA, Zhang D (2002)
Excitatory glycine receptors containing the NR3 family of NMDA receptor
subunits. Nature 415:793-798.
Cheng D, Hoogenraad CC, Rush J, Ramm E, Schlager MA, Duong DM, Xu P,
Wijayawardana SR, Hanfelt J, Nakagawa T, Sheng M, Peng J (2006) Relative
and absolute quantification of postsynaptic density proteome isolated from rat
forebrain and cerebellum. Mol Cell Proteomics 5:1158-1170.
Cho KO, Hunt CA, Kennedy MB (1992) The rat brain postsynaptic density fraction
contains a homolog of the Drosophila discs-large tumor suppressor protein.
Neuron 9:929-942.
Choquet D, Triller A (2003) The role of receptor diffusion in the organization of the
postsynaptic membrane. Nature Rev 4:251-265.
Ciabarra AM, Sullivan JM, Gahn LG, Pecht G, Heinemann S, Sevarino KA (1995)
Cloning and characterization of chi-1: a developmentally regulated member of a
novel class of the ionotropic glutamate receptor family. J Neurosci 15:64986508.
Colbran RJ, Brown AM (2004) Calcium/calmodulin-dependent protein kinase II and
synaptic plasticity. Current Opin Neurobiol 14:318-327.
Collins GG, Probett GA (1981) Aspartate and not glutamate is the likely transmitter of
the rat lateral olfactory tract fibres. Brain Res 209:231-234.
Collins MO, Yu L, Coba MP, Husi H, Campuzano I, Blackstock WP, Choudhary JS,
Grant SG (2005) Proteomic analysis of in vivo phosphorylated synaptic
proteins. J Biol Chem 280:5972-5982.
Conti F, DeBiasi S, Minelli A, Melone M (1996) Expression of NR1 and NR2A/B
subunits of the NMDA receptor in cortical astrocytes. Glia 17:254-258.
Costa MC, Mani F, Santoro W, Jr., Espreafico EM, Larson RE (1999) Brain myosin-V,
a calmodulin-carrying myosin, binds to calmodulin-dependent protein kinase II
and activates its kinase activity. J Biol Chem 274:15811-15819.
Cueille N, Blanc CT, Popa-Nita S, Kasas S, Catsicas S, Dietler G, Riederer BM (2007)
Characterization of MAP1B heavy chain interaction with actin. Brain Res Bull
71:610-618.
Cui H, Hayashi A, Sun HS, Belmares MP, Cobey C, Phan T, Schweizer J, Salter MW,
Wang YT, Tasker RA, Garman D, Rabinowitz J, Lu PS, Tymianski M (2007)
PDZ protein interactions underlying NMDA receptor-mediated excitotoxicity
and neuroprotection by PSD-95 inhibitors. J Neurosci 27:9901-9915.
Cull-Candy S, Brickley S, Farrant M (2001) NMDA receptor subunits: diversity,
development and disease. Current Opin Neurobiol 11:327-335.
Dallol A, Agathanggelou A, Fenton SL, Ahmed-Choudhury J, Hesson L, Vos MD,
Clark GJ, Downward J, Maher ER, Latif F (2004) RASSF1A interacts with
50
microtubule-associated proteins and modulates microtubule dynamics. Cancer
Res 64:4112-4116.
Das S, Sasaki YF, Rothe T, Premkumar LS, Takasu M, Crandall JE, Dikkes P, Conner
DA, Rayudu PV, Cheung W, Chen HS, Lipton SA, Nakanishi N (1998)
Increased NMDA current and spine density in mice lacking the NMDA
receptor subunit NR3A. Nature 393:377-381.
Davidkova G, Carroll RC (2007) Characterization of the role of microtubule-associated
protein 1B in metabotropic glutamate receptor-mediated endocytosis of AMPA
receptors in hippocampus. J Neurosci 27:13273-13278.
Dawson VL, Dawson TM, London ED, Bredt DS, Snyder SH (1991) Nitric oxide
mediates glutamate neurotoxicity in primary cortical cultures. Proc Natl Acad
Sci U S A 88:6368-6371.
Ding J, Liu JJ, Kowal AS, Nardine T, Bhattacharya P, Lee A, Yang Y (2002)
Microtubule-associated protein 1B: a neuronal binding partner for gigaxonin. J
Cell iol 158:427-433.
Dingledine R, Borges K, Bowie D, Traynelis SF (1999) The Glutamate Receptor Ion
Channels. Pharmacol Rev 51:7-62.
Dirnagl U, Iadecola C, Moskowitz MA (1999) Pathobiology of ischaemic stroke: an
integrated view. Trends Neurosci 22:391-397.
Durand GM, Gregor P, Zheng X, Bennett MV, Uhl GR, Zukin RS (1992) Cloning of
an apparent splice variant of the rat N-methyl-D-aspartate receptor NMDAR1
with altered sensitivity to polyamines and activators of protein kinase C. Proc
Natl Acad Sci USA 89:9359-9363.
Edelmann W, Zervas M, Costello P, Roback L, Fischer I, Hammarback JA, Cowan N,
Davies P, Wainer B, Kucherlapati R (1996) Neuronal abnormalities in
microtubule-associated protein 1B mutant mice. Proc Natl Acad Sci USA
93:1270-1275.
Ehlers MD, Zhang S, Bernhadt JP, Huganir RL (1996) Inactivation of NMDA
receptors by direct interaction of calmodulin with the NR1 subunit. Cell
84:745-755.
Ehlers MD, Fung ET, O'Brien RJ, Huganir RL (1998) Splice variant-specific
interaction of the NMDA receptor subunit NR1 with neuronal intermediate
filaments. J Neurosci 18:720-730.
Emery DL, Royo NC, Fischer I, Saatman KE, McIntosh TK (2003) Plasticity following
injury to the adult central nervous system: is recapitulation of a developmental
state worth promoting? J Neurotrauma 20:1271-1292.
Eriksson M, Nilsson A, Samuelsson H, Samuelsson EB, Mo L, Akesson E, Benedikz
E, Sundström E (2007) On the role of NR3A in human NMDA receptors.
Physiol Behav 92:54-59.
Evans PD, Reale V, Merzon RM, Villegas J (1992) N-methyl-D-aspartate (NMDA)
and non-NMDA (metabotropic) type glutamate receptors modulate the
membrane potential of the Schwann cell of the squid giant nerve fibre. J Exp
Biol 173:229-249.
Foldes RL, Adams SL, Fantaske RP, Kamboj RK (1994) Human N-methyl-D-aspartate
receptor modulatory subunit hNR2A: cloning and sequencing of the cDNA and
primary structure of the protein. Biochimica et biophysica acta 1223:155-159.
Forrest D, Yuzaki M, Soares HD, Ng L, Luk DC, Sheng M, Stewart CL, Morgan JI,
Connor JA, Curran T (1994) Targeted disruption of NMDA receptor 1 gene
abolishes NMDA response and results in neonatal death. Neuron 13:325-338.
Garcia-Perez J, Avila J, Diaz-Nido J (1998) Implication of cyclin-dependent kinases
and glycogen synthase kinase 3 in the phosphorylation of microtubuleassociated protein 1B in developing neuronal cells. J Neurosci Res 52:445-452.
Geyer MA, Krebs-Thomson K, Braff DL, Swerdlow NR (2001) Pharmacological
studies of prepulse inhibition models of sensorimotor gating deficits in
schizophrenia: a decade in review. Psychopharmacology (Berl) 156:117-154.
González-Billault C, Avila J (2000) Molecular genetic approaches to microtubuleassociated protein function. Histol Histopathol 15:1177-1183.
51
González-Billault C, Avila J, Caceres A (2001) Evidence for the role of MAP1B in
axon formation. Mol Biol Cell 12:2087-2098.
González-Billault C, Jimenez-Mateos EM, Caceres A, Diaz-Nido J, Wandosell F, Avila
J (2004) Microtubule-associated protein 1B function during normal
development, regeneration, and pathological conditions in the nervous system. J
Neurobiol 58:48-59.
Grant SG, Husi H (2001) Proteomics of multiprotein complexes: answering
fundamental questions in neuroscience. Trends Biotechnol 19:S49-54.
Gray EG (1959) Axo-somatic and axo-dendritic synapses of the cerebral cortex: an
electron microscope study. J Anat 93:420-433.
Gray NW, Kruchten AE, Chen J, McNiven MA (2005) A dynamin-3 spliced variant
modulates the actin/cortactin-dependent morphogenesis of dendritic spines. J
Cell Sci 118:1279-1290.
Guillaud L, Setou M, Hirokawa N (2003) KIF17 dynamics and regulation of NR2B
trafficking in hippocampal neurons. J Neurosci 23:131-140.
Gulli R, Masnata B, Bonvicini C, Tura GB, Manglaviti L, Vaggi M, Mollica M,
Bellone E, Mandich P, Gennarelli M, Di Maria E (2007) A putative regulatory
subunit (NR3A) of the NMDA receptor complex as candidate gene for
susceptibility to schizophrenia: a case-control study. Psychiatric Genetics
17:355-356.
Halpain S, Dehmelt L (2006) The MAP1 family of microtubule-associated proteins.
Genome Biol 7:224.221-224.227.
Hanley JG, Koulen P, Bedford F, Gordon-Weeks PR, Moss SJ (1999) The protein
MAP-1B links GABA(C) receptors to the cytoskeleton at retinal synapses.
Nature 397:66-69.
Hardingham GE, Bading H (2003) The Yin and Yang of NMDA receptor signalling.
Trends Neurosci 26:81-89.
Henson MA, Roberts AC, Salimi K, Vadlamudi S, Hamer RM, Gilmore JH, Jarskog
LF, Philpot BD (2008) Developmental Regulation of the NMDA Receptor
Subunits, NR3A and NR1, in Human Prefrontal Cortex. Cereb Cortex.
Hirai H, Kirsch J, Laube B, Betz H, Kuhse J (1996) The glycine binding site of the Nmethyl-D-aspartate receptor subunit NR1: identification of novel determinants
of co-agonist potentiation in the extracellular M3-M4 loop region. Proc Natl
Acad Sci USA 93:6031-6036.
Hirokawa N, Takemura R (2005) Molecular motors and mechanisms of directional
transport in neurons. Nat Rev Neurosci 6:201-214.
Hollmann M, Boulter J, Maron C, Beasley L, Sullivan J, Pecht G, Heinemann S (1993)
Zinc potentiates agonist-induced currents at certain splice variants of the
NMDA receptor. Neuron 10:943-954.
Hollmann M, Heinemann S (1994) Cloned glutamate receptors. Ann Rev Neurosci
17:31-108.
Hsu SC, Ting AE, Hazuka CD, Davanger S, Kenny JW, Kee Y, Scheller RH (1996)
The mammalian brain rsec6/8 complex. Neuron 17:1209-1219.
Hsu SC, Hazuka CD, Foletti DL, Scheller RH (1999) Targeting vesicles to specific
sites on the plasma membrane: the role of the sec6/8 complex. Trends Cell Biol
9:150-153.
Huh KH, Wenthold RJ (1999) Turnover analysis of glutamate receptors identifies a
rapidly degraded pool of the N-methyl-D-aspartate receptor subunit, NR1, in
cultured cerebellar granule cells. J Biol Chem 274:151-157.
Hummel T, Krukkert K, Roos J, Davis G, Klambt C (2000) Drosophila Futsch/22C10
is a MAP1B-like protein required for dendritic and axonal development.
Neuron 26:357-370.
Husi H, Ward MA, Choudhary JS, Blackstock WP, Grant SG (2000) Proteomic
analysis of NMDA receptor-adhesion protein signaling complexes. Nat
Neurosci 3:661-669.
Ikeda K, Nagasawa M, Mori H, Araki K, Sakimura K, Watanabe M, Inoue Y, Mishina
M (1992) Cloning and expression of the epsilon 4 subunit of the NMDA
receptor channel. FEBS lett 313:34-38.
52
Jordan BA, Fernholz BD, Boussac M, Xu C, Grigorean G, Ziff EB, Neubert TA (2004)
Identification and verification of novel rodent postsynaptic density proteins.
Mol Cell Proteomics 3:857-871.
Káradóttir R, Cavelier P, Bergersen LH, Attwell D (2005) NMDA receptors are
expressed in oligodendrocytes and activated in ischaemia. Nature 438:11621166.
Karp SJ, Masu M, Eki T, Ozawa K, Nakanishi S (1993) Molecular cloning and
chromosomal localization of the key subunit of the human N-methyl-Daspartate receptor. J Biol Chem 268:3728-3733.
Kawakami S, Muramoto K, Ichikawa M, Kuroda Y (2003) Localization of
microtubule-associated protein (MAP) 1B in the postsynaptic densities of the
rat cerebral cortex. Cell Mol Neurobiol 23:887-894.
Kennedy MB, Bennett MK, Erondu NE (1983) Biochemical and immunochemical
evidence that the "major postsynaptic density protein" is a subunit of a
calmodulin-dependent protein kinase. Proc Natl Acad Sci USA 80:7357-7361.
Kennedy MJ, Ehlers MD (2006) Organelles and trafficking machinery for postsynaptic
plasticity. Annu Rev Neurosci 29:325-362.
Kessels MM, Qualmann B (2002) Syndapins integrate N-WASP in receptor-mediated
endocytosis. EMBO J 21:6083-6094.
Kessels MM, Qualmann B (2004) The syndapin protein family: linking membrane
trafficking with the cytoskeleton. J Cell Sci 117:3077-3086.
Kim E, Cho KO, Rothschild A, Sheng M (1996) Heteromultimerization and NMDA
receptor-clustering activity of Chapsyn-110, a member of the PSD-95 family of
proteins. Neuron 17:103-113.
Kim E, Sheng M (2004) PDZ domain proteins of synapses. Nature reviews 5:771-781.
Kistner U, Wenzel BM, Veh RW, Cases-Langhoff C, Garner AM, Appeltauer U, Voss
B, Gundelfinger ED, Garner CC (1993) SAP90, a rat presynaptic protein related
to the product of the Drosophila tumor suppressor gene dlg-A. Journal Biol
Chem 268:4580-4583.
Kitamura C, Shirai K, Inoue M, Tashiro T (2007) Changes in the subcellular
distribution of microtubule-associated protein 1B during synaptogenesis of
cultured rat cortical neurons. Cell Mol Neurobiol 27:57-73.
Kornau HC, Schenker LT, Kennedy MB, Seeburg PH (1995) Domain interaction
between NMDA receptor subunits and the postsynaptic density protein PSD-95.
Science 269:1737-1740.
Koukoui SD, Chaudhuri A (2007) Neuroanatomical, molecular genetic, and behavioral
correlates of fragile X syndrome. Brain Res Rev 53:27-38.
Kutschera W, Zauner W, Wiche G, Propst F (1998) The mouse and rat MAP1B genes:
genomic organization and alternative transcription. Genomics 49:430-436.
Kutsuwada T, Kashiwabuchi N, Mori H, Sakimura K, Kushiya E, Araki K, Meguro H,
Masaki H, Kumanishi T, Arakawa M, et al. (1992) Molecular diversity of the
NMDA receptor channel. Nature 358:36-41.
Laube B, Hirai H, Sturgess M, Betz H, Kuhse J (1997) Molecular determinants of
agonist discrimination by NMDA receptor subunits: analysis of the glutamate
binding site on the NR2B subunit. Neuron 18:493-503.
Laube B, Kuhse J, Betz H (1998) Evidence for a Tetrameric Structure of Recombinant
NMDA Receptors. J Neurosci 18:2954-2961.
Lien LL, Feener CA, Fischbach N, Kunkel LM (1994) Cloning of human microtubuleassociated protein 1B and the identification of a related gene on chromosome
15. Genomics 22:273-280.
Lin YJ, Bovetto S, Carver JM, Giordano T (1996) Cloning of the cDNA for the human
NMDA receptor NR2C subunit and its expression in the central nervous system
and periphery. Brain Res Mol Brain Res 43:57-64.
Lin JW, Wyszynski M, Madhavan R, Sealock R, Kim JU, Sheng M (1998) Yotiao, a
novel protein of neuromuscular junction and brain that interacts with specific
splice variants of NMDA receptor subunit NR1. J Neurosci 18:2017-2027.
Lipton SA (2006) NMDA receptors, glial cells, and clinical medicine. Neuron 50:9-11.
Liu L, McKeehan WL (2002a) Sequence analysis of LRPPRC and its SEC1 domain
interaction partners suggests roles in cytoskeletal organization, vesicular
53
trafficking, nucleocytosolic shuttling, and chromosome activity. Genomics
79:124-136.
Liu L, Vo A, Liu G, McKeehan WL (2002b) Novel complex integrating mitochondria
and the microtubular cytoskeleton with chromosome remodeling and tumor
suppressor RASSF1 deduced by in silico homology analysis, interaction
cloning in yeast, and colocalization in cultured cells. In Vitro Cell Dev Biol
Anim 38:582-594.
Liu L, Vo A, McKeehan WL (2005a) Specificity of the methylation-suppressed A
isoform of candidate tumor suppressor RASSF1 for microtubule
hyperstabilization is determined by cell death inducer C19ORF5. Cancer Res
65:1830-1838.
Liu L, Vo A, Liu G, McKeehan WL (2005b) Distinct structural domains within
C19ORF5 support association with stabilized microtubules and mitochondrial
aggregation and genome destruction. Cancer Res 65:4191-4201.
Liu L, Vo A, Liu G, McKeehan WL (2005c) Putative tumor suppressor RASSF1
interactive protein and cell death inducer C19ORF5 is a DNA binding protein.
Biochem Biophys Res Commun 332:670-676.
Lu R, Wang H, Liang Z, Ku L, O'Donnell W T, Li W, Warren ST, Feng Y (2004) The
fragile X protein controls microtubule-associated protein 1B translation and
microtubule stability in brain neuron development. Proc Natl Acad Sci USA
101:15201-15206.
Lue RA, Marfatia SM, Branton D, Chishti AH (1994) Cloning and characterization of
hdlg: the human homologue of the Drosophila discs large tumor suppressor
binds to protein 4.1. Proc Natl Acad Sci USA 91:9818-9822.
Lysko PG, Cox JA, Vigano MA, Henneberry RC (1989) Excitatory amino acid
neurotoxicity at the N-methyl-D-aspartate receptor in cultured neurons:
pharmacological characterization. Brain Res 499:258-266.
Ma OK, Sucher NJ (2004) Molecular interaction of NMDA receptor subunit NR3A
with protein phosphatase 2A. Neuroreport 15:1447-1450.
Madry C, Mesic I, Bartholomaus I, Nicke A, Betz H, Laube B (2007) Principal role of
NR3 subunits in NR1/NR3 excitatory glycine receptor function. Biochem
Biophys Res Commun 354:102-108.
Malenka RC, Bear MF (2004) LTP and LTD: an embarrassment of riches. Neuron
44:5-21.
Martin KC, Barad M, Kandel ER (2000) Local protein synthesis and its role in
synapse-specific plasticity. Current opinion in neurobiology 10:587-592.
Matsuda K, Kamiya Y, Matsuda S, Yuzaki M (2002) Cloning and characterization of a
novel NMDA receptor subunit NR3B: a dominant subunit that reduces calcium
permeability. Brain Res Mol Brain Res 100:43-52.
McIlhinney RA, Molnar E, Atack JR, Whiting PJ (1996) Cell surface expression of the
human N-methyl-D-aspartate receptor subunit 1a requires the co-expression of
the NR2A subunit in transfected cells. Neurosci 70:989-997.
Meguro H, Mori H, Araki K, Kushiya E, Kutsuwada T, Yamazaki M, Kumanishi T,
Arakawa M, Sakimura K, Mishina M (1992) Functional characterization of a
heteromeric NMDA receptor channel expressed from cloned cDNAs. Nature
357:70-74.
Mei X, Sweatt AJ, Hammarback JA (2000a) Regulation of microtubule-associated
protein 1B (MAP1B) subunit composition. J Neurosci Res 62:56-64.
Mei X, Sweatt AJ, Hammarback JA (2000b) Microtubule-associated protein 1 subunit
expression in primary cultures of rat brain. Brain Res Bull 53:801-806.
Meixner A, Haverkamp S, Wassle H, Fuhrer S, Thalhammer J, Kropf N, Bittner RE,
Lassmann H, Wiche G, Propst F (2000) MAP1B is required for axon guidance
and Is involved in the development of the central and peripheral nervous
system. J Cell Biol 151:1169-1178.
Micu I, Jiang Q, Coderre E, Ridsdale A, Zhang L, Woulfe J, Yin X, Trapp BD,
McRory JE, Rehak R, Zamponi GW, Wang W, Stys PK (2006) NMDA
receptors mediate calcium accumulation in myelin during chemical ischaemia.
Nature 439:988-992.
54
Miyamoto E (2006) Molecular mechanism of neuronal plasticity: induction and
maintenance of long-term potentiation in the hippocampus. J Pharmacol Sci
100:433-442.
Monyer H, Burnashev N, Laurie DJ, Sakmann B, Seeburg PH (1994) Developmental
and regional expression in the rat brain and functional properties of four
NMDA receptors. Neuron 12:529-540.
Monyer H, Sprengel R, Schoepfer R, Herb A, Higuchi M, Lomeli H, Burnashev N,
Sakmann B, Seeburg PH (1992) Heteromeric NMDA receptors: molecular and
functional distinction of subtypes. Science 256:1217-1221.
Moriyoshi K, Masu M, Ishii T, Shigemoto R, Mizuno N, Nakanishi S (1991) Molecular
cloning and characterization of the rat NMDA receptor. Nature 354:31-37.
Mueller HT, Meador-Woodruff JH (2003) Expression of the NR3A subunit of the
NMDA receptor in human fetal brain. Ann N Y Acad Sci 1003:448-451.
Mueller HT, Meador-Woodruff JH (2004) NR3A NMDA receptor subunit mRNA
expression in schizophrenia, depression and bipolar disorder. Schizophr Res
71:361-370.
Mueller HT, Meador-Woodruff JH (2005) Distribution of the NMDA receptor NR3A
subunit in the adult pig-tail macaque brain. J Chem Neuroanat 29:157-172.
Muller BM, Kistner U, Veh RW, Cases-Langhoff C, Becker B, Gundelfinger ED,
Garner CC (1995) Molecular characterization and spatial distribution of SAP97,
a novel presynaptic protein homologous to SAP90 and the Drosophila discslarge tumor suppressor protein. J Neurosci 15:2354-2366.
Muller BM, Kistner U, Kindler S, Chung WJ, Kuhlendahl S, Fenster SD, Lau LF, Veh
RW, Huganir RL, Gundelfinger ED, Garner CC (1996) SAP102, a novel
postsynaptic protein that interacts with NMDA receptor complexes in vivo.
Neuron 17:255-265.
Naisbitt S, Kim E, Tu JC, Xiao B, Sala C, Valtschanoff J, Weinberg RJ, Worley PF,
Sheng M (1999) Shank, a novel family of postsynaptic density proteins that
binds to the NMDA receptor/PSD-95/GKAP complex and cortactin. Neuron
23:569-582.
Nakanishi N, Axel R, Shneider NA (1992) Alternative splicing generates functionally
distinct N-methyl-D-aspartate receptors. Proc Natl Acad Sci USA 89:85528556.
Niethammer M, Kim E, Sheng M (1996) Interaction between the C terminus of NMDA
receptor subunits and multiple members of the PSD-95 family of membraneassociated guanylate kinases. J Neurosci 16:2157-2163.
Nilsson A, Duan J, Mo-Boquist LL, Benedikz E, Sundström E (2007) Characterisation
of the human NMDA receptor subunit NR3A glycine binding site.
Neuropharmacol 52:1151-1159.
Nishi M, Hinds H, Lu HP, Kawata M, Hayashi Y (2001) Motoneuron-specific
expression of NR3B, a novel NMDA-type glutamate receptor subunit that
works in a dominant-negative manner. J Neurosci 21:RC185.
Okabe S (2007) Molecular anatomy of the postsynaptic density. Mol Cell Neurosci
34:503-518.
Olney JW, Newcomer JW, Farber NB (1999) NMDA receptor hypofunction model of
schizophrenia. J Psychiatr Res 33:523-533.
Orbán-Németh Z, Simader H, Badurek S, Trancikova A, Propst F (2005) Microtubuleassociated protein 1S, a short and ubiquitously expressed member of the
microtubule-associated protein 1 family. J Biol Chem 280:2257-2265.
Palay SL (1958) The morphology of synapses in the central nervous system. Exp Cell
Res 14:275-293.
Paoletti P, Neyton J (2007) NMDA receptor subunits: function and pharmacology. Curr
Opin Pharmacol 7:39-47.
Peng YC, Kuo F, Breiding DE, Wang YF, Mansur CP, Androphy EJ (2001) AMF1
(GPS2) modulates p53 transactivation. Mol Cell Biol 21:5913-5924.
Peters A, Palay SL, Webster HD (1991) The Fine Structure of the Nervous System:
Neurons and Their Supporting Cells (3rd ed.), Oxford University Press, New
York (1991).
55
Petralia RS, Wang YX, Wenthold RJ (1994) The NMDA receptor subunits NR2A and
NR2B show histological and ultrastructural localization patterns similar to those
of NR1. J Neurosci 14:6102-6120.
Pérez-Otano I, Ehlers MD (2004) Learning from NMDA receptor trafficking: clues to
the development and maturation of glutamatergic synapses. Neuro-Signals
13:175-189.
Pérez-Otano I, Schulteis CT, Contractor A, Lipton SA, Trimmer JS, Sucher NJ,
Heinemann SF (2001) Assembly with the NR1 subunit is required for surface
expression of NR3A-containing NMDA receptors. J Neurosci 21:1228-1237.
Pérez-Otano I, Lujan R, Tavalin SJ, Plomann M, Modregger J, Liu X-B, Jones EG,
Heinemann SF, Lo DC, Ehlers MD (2006) Endocytosis and synaptic removal of
NR3A-containing NMDA receptors by PACSIN1/syndapin1. Nature
neuroscience 9:611-621.
Petralia RS, Wang YX, Wenthold RJ (2003) Internalization at glutamatergic synapses
during development. Europ J Neurosci 18:3207-3217.
Plomann M, Lange R, Vopper G, Cremer H, Heinlein UA, Scheff S, Baldwin SA,
Leitges M, Cramer M, Paulsson M, Barthels D (1998) PACSIN, a brain protein
that is upregulated upon differentiation into neuronal cells. Europ J Biochem /
FEBS 256:201-211.
Prybylowski K, Fu Z, Losi G, Hawkins LM, Luo J, Chang K, Wenthold RJ, Vicini S
(2002) Relationship between availability of NMDA receptor subunits and their
expression at the synapse. J Neurosci 22:8902-8910.
Prybylowski K, Wenthold RJ (2004) N-Methyl-D-aspartate receptors: subunit
assembly and trafficking to the synapse. J Biol Chem 279:9673-9676.
Qualmann B, Roos J, DiGregorio PJ, Kelly RB (1999) Syndapin I, a synaptic dynaminbinding protein that associates with the neural Wiskott-Aldrich syndrome
protein. Mol Biol Cell 10:501-513.
Racz B, Blanpied TA, Ehlers MD, Weinberg RJ (2004) Lateral organization of
endocytic machinery in dendritic spines. Nat Neurosci 7:917-918.
Reese ML, Dakoji S, Bredt DS, Dotsch V (2007) The guanylate kinase domain of the
MAGUK PSD-95 binds dynamically to a conserved motif in MAP1a. Nat
Struct Mol Biol 14:155-163.
Riederer B, Matus A (1985) Differential expression of distinct microtubule-associated
proteins during brain development. Proc Natl Acad Sci USA 82:6006-6009.
Riederer BM (2007) Microtubule-associated protein 1B, a growth-associated and
phosphorylated scaffold protein. Brain Res Bull 71:541-558.
Rishi AK, Zhang L, Boyanapalli M, Wali A, Mohammad RM, Yu Y, Fontana JA,
Hatfield JS, Dawson MI, Majumdar AP, Reichert U (2003) Identification and
characterization of a cell cycle and apoptosis regulatory protein-1 as a novel
mediator of apoptosis signaling by retinoid CD437. Journal Biol Chem
278:33422-33435.
Rosenmund C, Stern-Bach Y, Stevens CF (1998) The tetrameric structure of a
glutamate receptor channel. Science 280:1596-1599.
Safaei R, Fischer I (1989) Cloning of a cDNA encoding MAP1B in rat brain:
regulation of mRNA levels during development. J Neurochem 52:1871-1879.
Sala C, Piech V, Wilson NR, Passafaro M, Liu G, Sheng M (2001) Regulation of
dendritic spine morphology and synaptic function by Shank and Homer.
Neuron 31:115-130.
Salter MG, Fern R (2005) NMDA receptors are expressed in developing
oligodendrocyte processes and mediate injury. Nature 438:1167-1171.
Sans N, Prybylowski K, Petralia RS, Chang K, Wang YX, Racca C, Vicini S,
Wenthold RJ (2003) NMDA receptor trafficking through an interaction
between PDZ proteins and the exocyst complex. Nat Cell Biol 5:520-530.
Sasaki YF, Rothe T, Premkumar LS, Das S, Cui J, Talantova MV, Wong HK, Gong X,
Chan SF, Zhang D, Nakanishi N, Sucher NJ, Lipton SA (2002)
Characterization and comparison of the NR3A subunit of the NMDA receptor
in recombinant systems and primary cortical neurons. J Neurophysiol 87:20522063.
56
Sattler R, Xiong Z, Lu WY, Hafner M, MacDonald JF, Tymianski M (1999) Specific
coupling of NMDA receptor activation to nitric oxide neurotoxicity by PSD-95
protein. Science 284:1845-1848.
Scannevin RH, Huganir RL (2000) Postsynaptic organization and regulation of
excitatory synapses. Nat Rev Neurosci 1:133-141.
Schmidt C, Hollmann M (2008) Apparent homomeric NR1 currents observed in
Xenopus oocytes are caused by an endogenous NR2 subunit. J Mol Biol
376:658-670.
Schuler T, Mesic I, Madry C, Bartholomaus I, Laube B (2008) Formation of NR1/NR2
and NR1/NR3 heterodimers constitutes the initial step in N-methyl-D-aspartate
receptor assembly. The J Biol Chem 283:37-46.
Sekino Y, Kojima N, Shirao T (2007) Role of actin cytoskeleton in dendritic spine
morphogenesis. Neurochem Internat 51:92-104.
Setou M, Nakagawa T, Seog DH, Hirokawa N (2000) Kinesin superfamily motor
protein KIF17 and mLin-10 in NMDA receptor-containing vesicle transport.
Science 288:1796-1802.
Sheng M, Pak DT (2000) Ligand-gated ion channel interactions with cytoskeletal and
signaling proteins. Annu Rev Physiol 62:755-778.
Sheng M, Hoogenraad CC (2007) The postsynaptic architecture of excitatory synapses:
a more quantitative view. Annu Rev Biochem 76:823-847.
Sloboda RD, Rudolph SA, Rosenbaum JL, Greengard P (1975) Cyclic AMP-dependent
endogenous phosphorylation of a microtubule-associated protein. Proc Natl
Acad Sci USA 72:177-181.
Smothers CT, Woodward JJ (2007) Pharmacological Characterization of GlycineActivated Currents in HEK 293 Cells Expressing NMDA NR1 and NR3
Subunits. J Pharmacol Exp Ther:jpet.107.123836.
Soloviev MM, Barnard EA (1997) Xenopus oocytes express a unitary glutamate
receptor endogenously. J Mol Biol 273:14-18.
Song MS, Chang JS, Song SJ, Yang TH, Lee H, Lim DS (2005) The centrosomal
protein RAS association domain family protein 1A (RASSF1A)-binding protein
1 regulates mitotic progression by recruiting RASSF1A to spindle poles. J Biol
Chem 280:3920-3927.
Spain BH, Bowdish KS, Pacal AR, Staub SF, Koo D, Chang CY, Xie W, Colicelli J
(1996) Two human cDNAs, including a homolog of Arabidopsis FUS6
(COP11), suppress G-protein- and mitogen-activated protein kinase-mediated
signal transduction in yeast and mammalian cells. Mol Cell Biol 16:6698-6706.
Standley S, Roche KW, McCallum J, Sans N, Wenthold RJ (2000) PDZ domain
suppression of an ER retention signal in NMDA receptor NR1 splice variants.
Neuron 28:887-898.
Stegenga SL, Kalb RG (2001) Developmental regulation of N-methyl-D-aspartate- and
kainate-type glutamate receptor expression in the rat spinal cord. Neurosci
105:499-507.
Stuppia L, Gatta V, Fogh I, Gaspari AR, Morizio E, Mingarelli R, Di Santo M, Pizzuti
A, Calabrese G, Palka G (2001) Genomic organization, physical mapping, and
involvement in Yq microdeletions of the VCY2 (BPY 2) gene. Genomics
72:153-157.
Stys PK, Lipton SA (2007) White matter NMDA receptors: an unexpected new
therapeutic target? Trends Pharmacol Sci 28:561-566.
Sucher NJ, Akbarian S, Chi CL, Leclerc CL, Awobuluyi M, Deitcher DL, Wu MK,
Yuan JP, Jones EG, Lipton SA (1995) Developmental and regional expression
pattern of a novel NMDA receptor-like subunit (NMDAR-L) in the rodent
brain. J Neurosci 15:6509-6520.
Sucher NJ, Kohler K, Tenneti L, Wong HK, Grunder T, Fauser S, Wheeler-Schilling T,
Nakanishi N, Lipton SA, Guenther E (2003) N-methyl-D-aspartate receptor
subunit NR3A in the retina: developmental expression, cellular localization, and
functional aspects. Invest Ophthalmol Vis Sci 44:4451-4456.
Sun H, Hu XQ, Emerit MB, Schoenebeck JC, Kimmel CE, Peoples RW, Miko A,
Zhang L (2008) Modulation of 5-HT3 receptor desensitization by the light
57
chain of microtubule-associated protein 1B expressed in HEK 293 cells. J
Physiol 586:751-762.
Sun L, Margolis FL, Shipley MT, Lidow MS (1998) Identification of a long variant of
mRNA encoding the NR3 subunit of the NMDA receptor: its regional
distribution and developmental expression in the rat brain. FEBS lett 441:392396.
Sundström E, Whittemore S, Mo L-L, Seiger A (1997) Analysis of NMDA Receptors
in the Human Spinal Cord. Exp Neurol 148:407-413.
Szebenyi G, Bollati F, Bisbal M, Sheridan S, Faas L, Wray R, Haferkamp S, Nguyen S,
Caceres A, Brady ST (2005) Activity-driven dendritic remodeling requires
microtubule-associated protein 1A. Curr Biol 15:1820-1826.
Takei Y, Kondo S, Harada A, Inomata S, Noda T, Hirokawa N (1997) Delayed
development of nervous system in mice homozygous for disrupted microtubuleassociated protein 1B (MAP1B) gene. J Cell Biol 137:1615-1626.
Tong G, Takahashi H, Tu S, Shin Y, Talantova M, Zago W, Xia P, Nie Z, Goetz T,
Zhang D, Lipton SA, Nakanishi N (2008) Modulation of NMDA receptor
properties and synaptic transmission by the NR3A subunit in mouse
hippocampal and cerebrocortical neurons. J Neurophysiol 99:122-132.
Tu JC, Xiao B, Yuan JP, Lanahan AA, Leoffert K, Li M, Linden DJ, Worley PF (1998)
Homer binds a novel proline-rich motif and links group 1 metabotropic
glutamate receptors with IP3 receptors. Neuron 21:717-726.
Tu JC, Xiao B, Naisbitt S, Yuan JP, Petralia RS, Brakeman P, Doan A, Aakalu VK,
Lanahan AA, Sheng M, Worley PF (1999) Coupling of mGluR/Homer and
PSD-95 complexes by the Shank family of postsynaptic density proteins.
Neuron 23:583-592.
Tögel M, Wiche G, Propst F (1998) Novel features of the light chain of microtubuleassociated protein MAP1B: microtubule stabilization, self interaction, actin
filament binding, and regulation by the heavy chain. J Cell Biol 143:695-707.
Tölle TR, Berthele A, Zieglgansberger W, Seeburg PH, Wisden W (1993) The
differential expression of 16 NMDA and non-NMDA receptor subunits in the
rat spinal cord and in periaqueductal gray. J Neurosci 13:5009-5028.
Ulloa L, Avila J, Diaz-Nido J (1993) Heterogeneity in the phosphorylation of
microtubule-associated protein MAP1B during rat brain development. J
Neurochem 61:961-972.
Ulloa L, Diez-Guerra FJ, Avila J, Diaz-Nido J (1994) Localization of differentially
phosphorylated isoforms of microtubule-associated protein 1B in cultured rat
hippocampal neurons. Neurosci 61:211-223.
Verkhratsky A, Kirchhoff F (2007) NMDA Receptors in glia. Neuroscientist 13:28-37.
Walikonis RS, Jensen ON, Mann M, Provance DW, Jr., Mercer JA, Kennedy MB
(2000) Identification of proteins in the postsynaptic density fraction by mass
spectrometry. J Neurosci 20:4069-4080.
Walikonis RS, Oguni A, Khorosheva EM, Jeng CJ, Asuncion FJ, Kennedy MB (2001)
Densin-180 forms a ternary complex with the (alpha)-subunit of
Ca2+/calmodulin-dependent protein kinase II and (alpha)-actinin. J Neurosci
21:423-433.
Washbourne P, Bennett JE, McAllister AK (2002) Rapid recruitment of NMDA
receptor transport packets to nascent synapses. Nat Neurosci 5:751-759.
Washbourne P, Liu X-B, Jones EG, McAllister AK (2004) Cycling of NMDA
Receptors during Trafficking in Neurons before Synapse Formation. J Neurosci
24:8253-8264.
Wenthold RJ, Prybylowski K, Standley S, Sans N, Petralia RS (2003) Trafficking of
NMDA receptors. Annu Rev Pharmacol Toxicol 43:335-358.
Wenzel A, Scheurer L, Kunzi R, Fritschy JM, Mohler H, Benke D (1995) Distribution
of NMDA receptor subunit proteins NR2A, 2B, 2C and 2D in rat brain.
Neuroreport 7:45-48.
58
Wenzel A, Fritschy JM, Mohler H, Benke D (1997) NMDA receptor heterogeneity
during postnatal development of the rat brain: differential expression of the
NR2A, NR2B, and NR2C subunit proteins. J Neurochem 68:469-478.
Westphal RS, Tavalin SJ, Lin JW, Alto NM, Fraser ID, Langeberg LK, Sheng M, Scott
JD (1999) Regulation of NMDA receptors by an associated phosphatase-kinase
signaling complex. Science NY 285:93-96.
Wiche G, Herrmann H, Leichtfried F, Pytela R (1982) Plectin: a high-molecular-weight
cytoskeletal polypeptide component that copurifies with intermediate filaments
of the vimentin type. Cold Spring Harbor symposia on quantitative biology 46
Pt 1:475-482.
Wolosker H (2007) NMDA receptor regulation by D-serine: new findings and
perspectives. Mol Neurobiol 36:152-164.
Wong EY, Tse JY, Yao KM, Lui VC, Tam PC, Yeung WS (2004) Identification and
characterization of human VCY2-interacting protein: VCY2IP-1, a
microtubule-associated protein-like protein. Biol Reprod 70:775-784.
Wong HK, Liu XB, Matos MF, Chan SF, Pérez-Otano I, Boysen M, Cui J, Nakanishi
N, Trimmer JS, Jones EG, Lipton SA, Sucher NJ (2002) Temporal and regional
expression of NMDA receptor subunit NR3A in the mammalian brain. J Comp
Neurol 450:303-317.
Wong RW, Setou M, Teng J, Takei Y, Hirokawa N (2002) Overexpression of motor
protein KIF17 enhances spatial and working memory in transgenic mice. Proc
Natl Acad Sci USA 99:14500-14505.
Wyszynski M, Lin J, Rao A, Nigh E, Beggs AH, Craig AM, Sheng M (1997)
Competitive binding of alpha-actinin and calmodulin to the NMDA receptor.
Nature 385:439-442.
Xiao B, Tu JC, Petralia RS, Yuan JP, Doan A, Breder CD, Ruggiero A, Lanahan AA,
Wenthold RJ, Worley PF (1998) Homer regulates the association of group 1
metabotropic glutamate receptors with multivalent complexes of homer-related,
synaptic proteins. Neuron 21:707-716.
Yamazaki M, Mori H, Araki K, Mori KJ, Mishina M (1992) Cloning, expression and
modulation of a mouse NMDA receptor subunit. FEBS lett 300:39-45.
Yao Y, Mayer ML (2006) Characterization of a soluble ligand binding domain of the
NMDA receptor regulatory subunit NR3A. J Neurosci 26:4559-4566.
Yeaman C, Grindstaff KK, Wright JR, Nelson WJ (2001) Sec6/8 complexes on transGolgi network and plasma membrane regulate late stages of exocytosis in
mammalian cells. J Cell Biol 155:593-604.
Yuen EY, Jiang Q, Feng J, Yan Z (2005a) Microtubule Regulation of N-Methyl-Daspartate Receptor Channels in Neurons. J Biol Chem 280:29420-29427.
Yuen EY, Jiang Q, Chen P, Gu Z, Feng J, Yan Z (2005b) Serotonin 5-HT1A Receptors
Regulate NMDA Receptor Channels through a Microtubule-Dependent
Mechanism. J Neurosci 25:5488-5501.
Zerangue N, Schwappach B, Jan YN, Jan LY (1999) A new ER trafficking signal
regulates the subunit stoichiometry of plasma membrane K(ATP) channels.
Neuron 22:537-548.
Zhang S, Ehlers MD, Bernhardt JP, Su CT, Huganir RL (1998) Calmodulin mediates
calcium-dependent inactivation of N-methyl-D-aspartate receptors. Neuron
21:443-453.
Zukin RS, Bennett MV (1995) Alternatively spliced isoforms of the NMDARI receptor
subunit. Trends Neurosci 18:306-313.
59