• Keine Ergebnisse gefunden

α and SV2A interactome Molecular mechanisms underlying presynaptic plasticity: characterization of the RIM1

N/A
N/A
Protected

Academic year: 2022

Aktie "α and SV2A interactome Molecular mechanisms underlying presynaptic plasticity: characterization of the RIM1"

Copied!
135
0
0

Wird geladen.... (Jetzt Volltext ansehen)

Volltext

(1)

Molecular mechanisms underlying presynaptic plasticity: characterization of the RIM1α and

SV2A interactome

Dissertation

zur

Erlangung des Doktorgrades (Dr. rer. nat.) der

Mathematisch-Naturwissenschaftlichen Fakultät der

Rheinischen Friedrich-Wilhelms-Universität Bonn

vorgelegt von

Ana-Maria Oprişoreanu

aus

Târgovişte, Rumänien

Bonn 2014

(2)

Angefertigt mit Genehmigung der Mathematisch-Naturwissenschaftlichen Fakultät der Rheinischen Friedrich-Wilhelms-Universität Bonn

1. Gutachter Prof. Dr. Susanne Schoch 2. Gutachter Prof. Dr. Albert Haas

Tag der Promotion: 13.01.2015 Erscheinungsjahr: 2015

Diese Dissertation ist auf dem Hochschulschriftenserver der ULB Bonn unter http://hss.ulb.uni-bonn.de/diss_online electronisch publiziert.

(3)

   

Erklärung

Diese Dissertation wurde im Sinne von § 4 der Promotionsordnung vom 17.06.2011 am Institut für Neuropathologie und Klinik für Epileptologie der Universität Bonn unter der Leitung von Frau Prof. Dr. Susanne Schoch angefertigt.

Hiermit versichere ich, dass ich die vorliegende Arbeit selbständig angefertigt habe und keine weiteren als die angegebenen Hilfsmittel und Quelle verwendet habe, die gemäß § 6 der Promotionsordnung kenntlich gemacht sind.

Bonn, den

Ana-Maria Oprişoreanu

   

(4)

 

1.Introduction ... 1

1.1 The synapse ... 1

1.2 Cytometrix at the active zone (CAZ) ... 1

1.2.1 Active Zone Ultrastructure ... 1

1.2.2 Active Zone composition ... 3

1.3 The synaptic vesicle cycle ... 4

1.4 Synaptic plasticity ... 5

1.4.1 Presynaptic dormancy ... 6

1.4.2 Molecular mechanisms involved in presynaptic LTP ... 7

1.5 Two major players in synaptic plasticity ... 7

1.5.1 RIMs ... 8

1.5.1.1 RIM gene structure ... 8

1.5.1.2 RIM protein structure and binding partners... 9

1.5.1.3 RIM function ... 11

1.5.1.3.1 RIM in invertebrates (C.elegans and D.melanogaster) ... 11

1.5.1.3.2 RIM in vertebrates (M.musculus) ... 12

1.5.1.3.2.1 RIM1α knock-out mice ... 12

1.5.1.3.2.2 RIM1αβ double knock-out mice ... 13

1.5.1.3.2.3 RIM2α knock-out mice ... 13

1.5.1.3.2.4 RIM1α/RIM2α double knock-out mice ... 13

1.5.1.3.2.5 RIM conditional knockout mice ... 14

1.5.2 Synaptic vesicle protein 2A (SV2A) ... 15

1.5.2.1 SV2A function ... 15

1.5.2.2 SV2A knock-out mice ... 16

1.6 Aim of the study ... 17

2. Materials ... 18

2.1 Equipment ... 18

2.2 Chemicals ... 19

2.3 Cell culture media ... 20

2.4 Kits ... 20

2.5 Enzymes ... 20

2.6 Inhibitors ... 20

2.7 Diverse materials ... 20

2.8 Cloning primers ... 21

2.9 Sequencing primers ... 22

2.10 Site-directed mutagenesis ... 22

2.11 Oligonucleotides used for HA-tag cloning ... 22

2.12 Oligonucleotides used for shRNA cloning ... 22

2.13 Generated constructs ... 23

2.14 Plasmids obtained from other sources and used in this thesis ... 23

2.15 Primary and secondary antibodies ... 24

3. Methods ... 25

3.1 Molecular Biology ... 25

(5)

 

3.1.1 RNA extraction and cDNA synthesis ... 25

3.1.2 Polymerase chain reaction (PCR) ... 25

3.1.3 Site directed mutagenesis ... 25

3.1.4 Sequencing ... 26

3.1.5 Cloning technique ... 26

3.1.5.1 Oligonucleotides cloning ... 26

3.2 Cell Culture ... 26

3.2.1 HEK (AAV) 293T cell culture ... 26

3.2.2 HEK (AAV) 293T transfection methods ... 27

3.2.2.1 Ca2+ -phosphate method ... 27

3.2.2.2 Lipofectamine method ... 27

3.2.3 Neuronal primary cell culture ... 27

3.2.3.1 Generation of primary cell culture ... 27

3.2.3.2 Transfection of neurons ... 28

3.2.3.3 Infection of neurons ... 28

3.3 Virus Production ... 28

3.3.1 rAAV serotype 1/2 and 8 production (Ca2+-phosphate method) ... 28

3.3.2 rAAV serotype 8 purification ... 29

3.3.3 P0-P3 animal injection ... 29

3.4 Biochemistry ... 30

3.4.1 Preparation of crude synaptosomes ... 30

3.4.2 Protein-protein interaction assays ... 30

3.4.2.1 Protein induction and purification from BL21 bacteria ... 30

3.4.2.2 GST-pull down assay ... 31

3.4.2.3 Co-immunoprecipitation (co-IP) ... 31

3.4.2.4 Immunoprecipitation (IP) ... 31

3.4.3 Protein concentration determination ... 32

3.4.4 Western Blotting (WB) ... 32

3.5 Identification of novel binding partners by tandem-affinity purification (TAP) ... 32

3.5.1 Protein cross-linking ... 32

3.5.2 Strep/FLAG tandem affinity purification ... 33

3.5.3 Protein purification from HEK293T cells ... 34

3.5.4 Binding assays between the different regions of RIM1α and crude synaptosomes ... 34

3.5.5 Sample preparation for mass spectrometer analysis ... 34

3.6 Immunochemical methods ... 36

3.6.1 Pre-treatment of primary neurons with various inhibitors ... 36

3.6.2 Immunofluorescence (IF) ... 36

3.6.3 Immunohistochemistry (IHC) ... 36

3.7 Imaging ... 37

3.8 Quantifications and statistical analysis ... 37

3.8.1 Image quantification ... 37

3.8.2 WB quantification ... 37

3.9 Programmes and URLs ... 37

(6)

 

4. Results ... 38

4.1 Impact of phosphorylation status on the properties of RIM1α ... 38

4.1.1 Distribution of RIM1α in synaptic boutons is altered by hyperphosphorylation events ... 38

4.1.2 Identification of novel phosphorylation-dependent RIM1α binding proteins ... 40

4.1.2.1 Identification of protein complexes associated with the C-terminal region of RIM1α ... 41

4.1.2.2 Analysis of protein complexes associated with the N-terminal region of RIM1α ... 44

4.1.2.3 Analysis of the protein complexes co-purified with the overexpressed C- terminal region of RIM1α in primary cultured neurons ... 45

4.1.3 Validation of the newly identified RIM1α binding proteins ... 48

4.1.3.1 Unc-51-like kinase (ULK) ... 48

4.1.3.1.1 ULK proteins bind RIM1α ... 48

4.1.3.1.2 The ULK-kinase domain mediates binding to RIM1α ... 49

4.1.3.1.3 ULK1/2 partially co-localize with endogenous RIM1/2 at synapses ... 50

4.1.3.1.4 Generation of a short-hairpin RNA against ULK2 ... 54

4.1.3.2 Serine-arginine protein kinase 2 (SRPK2) ... 55

4.1.3.2.1 SRPK2 targets RIM1α ... 56

4.1.3.2.2 Non-kinase core regions do not mediate direct binding to RIM1α ... 60

4.1.3.2.3 The effect of SRPIN340 inhibitor on the SRPK2 co-localization with endogenous RIM1α ... 62

4.1.3.3 Vesicle-associated membrane protein (VAMP) associated-protein A/B (VAPA/VAPB) ... 63

4.1.3.3.1 VAPA/VAPB binds RIM1α ... 63

4.1.3.3.2 Kinase inhibition strengthens the VAPA-RIM1α interaction ... 65

4.1.3.3.3 The T812/814A point mutations in the RIM1α C2A-domain impair binding to VAPA ... 66

4.1.3.3.4 VAP proteins bind RIM1α in co-IP assays ... 66

4.1.3.3.5 Co-localisation of VAP proteins with endogenous RIM1/2 in neuronal cell culture ... 67

4.1.3.4 Copine VI ... 71

4.1.3.4.1 Copine VI binds RIM1α ... 71

4.1.3.4.2 The Copine VI-RIM1α interaction is calcium dependent ... 72

4.1.3.4.3 Copine VI and RIM1/2 co-localized at a subset of synapses ... 72

4.2 SV2A ... 73

4.2.1 Generation and characterisation of the TAP-tagged SV2A constructs ... 73

4.2.2 Optimization of SV2A protein purification from primary rat cortical neurons ... 75

4.2.2.1 One-step purification yields good recovery of TAP-tagged SV2A ... 75

4.2.2.2 Two-step purification of fusion proteins leads to a decrease in elusion efficiency ... 76

4.2.3 SV2A overexpression and affinity purification from mouse brain ... 78

(7)

 

4.2.3.1 Analysis of mouse brain transduced with rAAV-SV2A-GFP indicates high

levels of expression of recombinant protein ... 79

4.2.3.2 N- and C-tagged SV2A affinity purification from transduced mouse brain ... 80

4.2.3.2.1 Analysis of single-step purification method ... 80

4.2.3.2.2 Two-step purification procedure ... 82

4.2.4 Analysis of protein complexes co-immunprecipitated with overexpressed SV2A in primary neuronal cell culture ... 83

4.2.4.1 Enrichment of bound protein complexes to SV2A by using cross-linkers and primary neurons from hetero- and homozygous SV2A mice ... 83

4.2.4.2 Identification of novel potential binding partners for SV2A by mass- spectrometry ... 85

5. Discussion ... 86

5.1 Hyperphosphorylation alters the distribution of the presynaptic protein RIM1α at synapses ... 86

5.2 Identification of novel phosphorylation-dependent RIM1α binding proteins ... 89

5.2.1 Two novel potential kinases associate with RIM1α protein ... 90

5.2.1.1 Unc-51-like kinase (ULK) binds the C2-domains of RIM1α ... 91

5.2.1.2 Serine Arginine protein kinase 2 (SRPK2) targets specifically the C2A- domain of RIM1α ... 93

5.2.2 VAPA/B proteins bind specifically the C2A-domain of RIM1α ... 96

5.2.3 Copine VI binds RIM1α in a calcium-dependent manner ... 98

5.3 Identification of novel SV2A binding partners: new experimental approaches ... 99

6. Outlook ... 102

7. Summary ... 103

8. Appendix ... 105

9. Abbreviations ... 113

10. References... 115

11. Acknowledgments ... 128

   

               

(8)

 

1. Introduction 1.1 The synapse

Already in 1897 Foster and Sherrington introduced the term synapse (from Greek synapsis

"conjunction", from synaptein "to clasp", from syn- "together" and haptein "to fasten)1

(WESTFALL et al., 1996). By 1962 the first nervous system, though a simple one, in Phylum Cnidaria (corals, anemones, and jellyfish) was defined by Horridge and Mackay. After Santiago Ramón y Cajal, the founder of modern neuroscience (LLINÁS, 2003), many scientists dedicated themselves in understanding the structure and function of synapses. In 1954 Palade and Palay described for the first time the structure of a vertebrate synapse using electron microscopy (EM). Since that time our understanding of synapse architecture has deepened, facilitated also by enhanced imaging techniques.

The synapse is an asymmetrical structure composed of a presynaptic terminal, a synaptic cleft and a postsynaptic terminal. The presynaptic terminal is important in regulating synaptic vesicle docking, priming, fusion and neurotransmitter release into the cleft, where the neurotransmitter molecules bind to the postsynaptic terminal’s receptors. In the postsynaptic terminal the chemical signal is converted into an electrical one and further propagated within the neuron. Several steps of synaptic vesicle (SV) fusion take place at a specialized structure in the presynaptic terminal, which contains an electron-dense cytoskeletal matrix, known as cytometrix at the active zone (CAZ) (review: SCHOCH and GUNDELFINGER, 2006; review: SÜDHOF, 2012).

1.2 Cytometrix at the active zone (CAZ) 1.2.1 Active Zone Ultrastructure

In a simplistic model the active zone consists of a proximal zone close to the plasma membrane, where the docking of synaptic vesicles (SV) takes place and a more distal zone where vesicles are tethered. Over the decades electron microscopy and tomography (EM) techniques have revealed the existence of an electron-dense structure expanding into the cytoplasma. These observed dense projections differ considerably between species (review:

ZHAI andBELLEN, 2004). At the neuromuscular junction (NMJ) of C.elegans the dense projection has been described as a plaque surrounded within 100nm by a subpopulation of vesicles (Fig.

1.1A; WEIMER et al., 2006); while in D.melanogaster, the dense structure takes the shape of a pedestal and a platform (T-bars) enclosed by synaptic vesicles and closely associated with calcium channels (Fig. 1.1B; PROKOP and MEINERTZHAGEN, 2006). In vertebrates (frog), the NMJ has       

1 ONLINE ETYMOLOGY DICTIONARY: www.etymonline.com 

(9)

  a mo (diffe orien macr

(Fig.

fixed with dense ribbo plate

(MER

incre neuro (NM and t

BELLE

ore complex ferent classe

ntated SVs romolecules

1.1E and 1.1F

d and staine synaptic v e structure, on is dictate e-like in bip

RCER and THOR

ease in the a otransmitter MJs) and cen the size of t

EN,2004).

x structure es of active (the lumin s) in order

F; HARLOW e

d samples h vesicles in b in the shap ed by the s polar cells,

RESON, 2011)

active zone rs upon c ntral nervou the nerve ter

formed by e zone mate nal assemb

to facilitate

et al., 2001, 20

highlighted between (rev

pe of a ribbo sensory cell whereas in .

area (T-bars continuous

us system ( rminal is no

y a tight and erial-AZM- bly of mac e the docki

013; SZULE et

the active z

view: SÜDHOF

on, where sy l type; dens

hair cells a

s and ribbon stimulation (CNS) syna ot restricted

d ordered c -macromole cromolecule ing process

t al., 2012). I zone as a he

F, 2012). Sen ynaptic vesi se structure

a spherical

n synapses) n. At vert apses, where d, the active

connection ecules) that

es orientate s with the p n mammali exagonal gri nsory synap icles are pri

in rod and form is dis

Figure 1.

of the ac plaque-lik C.elegans projection (C) ribbon (D) acti mammalia projection 3D arrang undocked and the Pictures w al., 2004;

HARLOW e

T size of t dictated type but the cell projectio in the siz releasabl ), conferring ebrate neu e stimulatio zone may e

of structura connect th ed towards presynaptic ian neurons

id of dense pses exhibi imed. The s d cone phot splayed (den

.1: Electron ctive zone st ke dense p s NMJ; (B) n in D.melano

n synapse in R ive zone an CNS.

n structure in f gement of the

SVs relative presynaptic were taken fr

; SZULE et a et al., 2013.

The differe the dense p not only b t also by the

l. The la ons allow a

ze of the RR le pool) w g a sustaine uromuscular ons are not expand (revi

al elements he correctly the AZM membrane s, EM using projections it a striking shape of the toreceptors, nse bodies)

micrographs tructure. (A) projection in T-bar dense ogaster NMJ;

R.norvegicus;

structure in (E) Dense frog NMJ; (F) e docked and e to the AZM membrane.

rom: ZHAI et al., 2012 and

nce in the rojection is by the cell e activity of arge dense an increase RP (readily without an d release of r junctions continuous

iew: ZHAI and

s y M e g s g e , )

s ) n e

;

; n e ) d M . t d

e s l f e e y n f s s

d

(10)

 

techn the A treatm is los false samp dense betw

FERNA

1.2.2 Desp diver unde adhe comp Picco

CASK

OPRIS

To unco niques have AZ as a re ments samp st due to pr interpretati ple to immo

e projection ween each ot

ANDEZ-BUSNA

2 Active Zon pite the diff

rsity of the erlying the sion, cytos posed of a olo and B

K, calcium/ca

OREANU,2014

over the arc e been appli egular array ples must un rotein aggre

ion of the d obilize the e

ns. Instead ther (connec

ADIEGO et al.,

ne composit ferences be e active zo

plasma m keletal form set of prote Bassoon, an

almodulin-dep 4.

chitecture o ied. EM of f y of thick

ndergo befo egation as a data. New E ntire structu a less orga ctors) and t

2010; review

tion

etween cryo one ultrastr embrane is mation, sca eins highly

nd ELKS

pendent serine

f the active fixed and st projections ore imaging a consequen EM techniq

ure in the na anized arch to the plasm

w: SIKSOU et al

Figu (yell and s from

o-EM and c ructure. Th s very com affolding an

enriched in (CAST/ER

e protein kin

e zone, diff tained samp s surrounde g, it has bee nce of prote ques based o ative state, itecture wit ma membran

l., 2011).

ure 1.2: 3D low), AZ (gr synaptic vesic m FERNANDEZ-

classical EM he nature o mplex. In a nd the fusio n this area RC)) and

(C

SC

SÜ

Fi co th CA BP co co pr SV nase. Adapted

ferent electr ples showed ed by SVs.

en argued th ein cross-lik only on fast

did not reve th fine filam ne (tethers)

D segmentatio ay), synaptic cle tethers (blu

-BUSNADIEGO

M, both app of the intri

addition to on machine

(RIM, RIM proteins a CASK/Veli/

CHOCH and

ÜDHOF, 2012) igure 1.3:

omposition. P e active zone AZ, cytometri P, RIM bindin ommon antige oupled rece rotein; RIM, V2A, synapt d from review

ron tomogr d the dense Due to th hat the nativ king, thus l t freezing o eal the pres ments conn

was observ

on of synap vesicle conn ue). All pictur

O et al., 2010.

pproaches re icate web

proteins i ery, the acti M-BP, Liprin

associated /Mint1) (Fig

GUNDELFIN

).

Active Z Proteins highl e are depicted rix at the activ ng protein; LA en related; GI

eptor kina Rab3 interac tic vesicle w: SCHOCH,

raphy (EM) material of he different ve structure eading to a of the tissue ence of any necting SVs ved (Fig. 1.2;

ptic vesicles nectors (red), res were taken

evealed the of proteins involved in ive zone is n, Munc13, with these

g. 1.3; review:

NGER, 2006;

one protein ly enriched at in light blue.

ve zone; RIM- AR, leukocyte IT, G-protein- ase-interacting cting protein;

protein 2A;

MÜLLER und

) f t e a e y s

;

s , n

e s n s , e

:

;

n t . - e

- g

;

; d

(11)

 

neuro order and plast perfo

2012)

1.3 T The neuro termi The repea neuro

review

sever medi sensi and o

2013)

comp

assis

The acti otransmitter r to facilitat postsynapti ticity. As a ormed with

).

The synapti active zo otransmitter inal, the vo transient C ated cycles otransmitter

ws: SÜDHOF, 2

After the ral changes iated on on itive factor on the othe

). Originall peting with

t the trans

ive zone, d rs release: ( te fast synch ic compartm a result, all

the requisit

ic vesicle cy one is the r release t ltage gated Ca2+ elevatio

of exocytos rs followed

2004, 2013). e synaptic v s until they ne side by th

attachment er side by M

y Munc18- core-compl

sition to an

due to its c (1) SV dock hronous exc ments; (4)

these func te speed an

ycle

e specializ akes place

calcium ch on stimulate

sis-endocyto by docking vesicles are y become f he formatio t protein rec Munc18, Mu

-1 was rep lex formatio

n ‘open’ co

complex or king and pr citation/rele mediation ctions guara nd plasticity

zed structu e. When an hannels open es SV exoc

osis. Durin g, priming,

filled with fusion-comp on of the SN

ceptor) con unc13, RIM ported to b

on (DULUBOV

F fil do re m co re fr

S ac do onformation

rganization, riming; (2) ease couplin

of both s antee that p

needed for

ure in the n action po n followed b cytosis. In t ng exocytos fusion and

neurotransm petent (prim

NARE com sisting of sy M and Syna bind syntax

VA et al., 1999

Figure 1.4: Sy ller with neur ocking; 4- pr elease; 6- re mediated endoc

ompartment;

ecycling with rom review: SÜ

Other V priming ctivated by omain (DEN

n of syntax

has four p calcium ch ng; (3) prec

hort- and l presynaptic r the inform

e presynap otential rea by an increa the synaptic is vesicles a neurotransm mitters and ming step).

mplex (solub ynaptobrevi aptotagmin1 in1 in a ‘c

9).

naptic vesicle otransmitters;

riming; 5- fu cycling via cytosis and re 8- clathrin m out endosoma

ÜDHOF, 2004.

r candidate are Munc1

binding to

NG et al., 20

xin1, enabli

principle fu hannels recr cise localiza long-term p c vesicle ex mation transf

ptic termin aches the p ase in Ca2+- c bouton SV

are filled ac mitters relea d docked, th

This prim ble N-ethyl vin-SNAP25

1 (reviews: SÜ

closed’ con

e cycle. 1- SV

; 2- reserve po usion and neu kiss-and-run;

ecycling via t mediated end mal intermedi

.

e proteins t 13 and RIM o the RIM

11), was su ling SNAR

functions in ruitment, in ation of pre- presynaptic xocytosis is fer (SÜDHOF,

nal, where presynaptic - ion influx.

Vs undergo ctively with ase (Fig. 1.4;

hey undergo ming step is lmaleimide- 5-Syntaxin1

ÜDHOF, 2004,

nformation,

Vs are actively ool of SVs; 3- urotransmitter

; 7- clathrin the endosome docytosis and iate. Adapted

to facilitate M. Munc13, Zn2+-finger uggested to RE complex n n - c s

,

e c . o h

;

o s -

,

,

y - r n e d d

e , r o x

(12)

 

assembly and thereby the priming step (BETZ et al., 1997; review: RIZO and SÜDHOF, 2002; STEVENS et al., 2005; review: SÜDHOF, 2013).

After the action potential reaches the presynaptic terminal, voltage gated calcium channels open and the calcium concentration builds up in a microdomain near the priming complex. The calcium sensor synaptotagmin1 (present on the synaptic vesicle) together with the SNARE complex further enables membrane fusion (DAI et al., 2008; CHOI et al., 2010; VRLJIC et al., 2010) with the formation of the pore to release the neurotransmitters into the synaptic cleft.

Following neurotransmitter release SVs are recycled via different routes, like kiss-and-run (vesicles undock and recycle locally), clathrin mediated endocytosis (vesicles are reacidified and refilled directly or by passing via the endosome compartment) (review: SÜDHOF, 2004) or via bulk endocytosis. Activity-dependent bulk endocytosis (ADBE) is the dominant retrieval pathway after an elevated stimulation activity (CHEUNG and COUSIN, 2013).

In accordance with the network’s needs, the amount of SVs ready to release neurotransmitter may very as well. SV recycling is tightly regulated by the action of different proteins, resident at the AZ. Therefore, fluctuations in the activity of synapses could be mediated by the actions of various AZ proteins, as well as by the SVs cycle. These changes represent the fundament of presynaptic plasticity.

1.4 Synaptic plasticity

The concept of synaptic plasticity, which was for the first time formulated by Hebb in 1949, refers to the capacity of synapses to react accordingly to the network’s needs either be weakening (depression) or strengthening (potentiation) its activity. These types of changes may well extend over short periods (short-term plasticity) or long periods of time (long-term plasticity). The Hebbian theory is used to describe these synaptic changes as being associative and rapidly induced, shortly explained as a positive feedback process (HEBB., 1949). For example, upon LTP induction, synapses become more excitable and the entire network activity would increase leading to a runaway potentiation. To prevent such extremes, the homeostatic process, which hinders the network to reach high levels of activity and preserve the stored information, has an important role (review: POZO and GODA, 2010).

In the active state or basal conditions synaptic transmission is mediated by the release of neurotransmitters from presynaptic terminals into the synaptic cleft, followed by the activation of different receptors on the postsynaptic terminal. Under increased network activity presynaptic neurons decrease their release probability (LTD-long-term depression), while the postsynaptic cells decrease the number of their receptors. To offset reduced network

(13)

 

activity, presynaptic neurons enhance the recycling, the number of docked vesicle and the release probability (LTP-long-term potentiation) (review: POZO and GODA, 2010; CASTILLO, 2012).

There are multiple parallel mechanisms responsible for controlling pre- and postsynaptic homeostasis, and consequently affecting synapse activity. The molecular mechanisms that govern the negative feedback (homeostatic plasticity) rely on the efficiency of different intracellular signalling cascades to detect and to respond accordingly to changes in the network. These fine-tuned mechanisms include: gene expression induction, protein synthesis and degradation. Besides the two major mechanisms: transcription and translation, post- translational modifications have emerged as an important factor in controlling plasticity

(review: POZO and GODA., 2010). Several post-translational modifications have been suggested to modulate the function of various pre- and postsynaptic proteins, like: palmitoylation (review:

EL-HUSSEINI and BREDT 2002), myristilation and prenylation (KUTZLEB et al., 1998; O’CALLAGHAN et al., 2003), SUMOylation (Small Ubiquitin-like Modifier) (GIRACH et al., 2013) and phosphorylation

(review: BARRIA, 2001).

1.4.1 Presynaptic dormancy

Presynaptic dormancy is induced as a response to a prolonged strong depolarization or increased action potential firing. Dormant synapses display a decrease in neurotransmitter release. The molecular mechanism is based on the inhibitory action of G proteins on adenylyl cyclase (AC), which causes a decrease in the level of cAMP and thereby directly affects the activity of protein kinase A (PKA) (Fig. 1.5). Therefore, presynaptic proteins are less phosphorylated and become susceptible to degradation through the proteasome (review:

CRAWFORD and MENNERICK, 2012). The protein levels of RIM1α and Munc13-1 were shown to be decreased upon induction of presynaptic dormancy through the action of the ubiquitin- proteasome system, while an overexpression of RIM1α in cultured neurons prevented the induction of silencing (JIANG et al., 2010). Recently two other presynaptic proteins, Piccolo and Bassoon were identified as negative regulators of the E3 ligase Siah1. In the DKO neurons the rate of presynaptic protein degradation was increased, leading to the observation that these two proteins are important regulators of the protein ubiquitination in the presynaptic terminal, therefore maintaining synapse integrity (WAITES et al., 2013).aaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaa

(14)

 

1.4.2 At th in Ca presy an in RIM impo

2002;

signi Addi kinas

CASTI

LTD 1.5 T Even eluci were leadi one r descr funct trans

2 Molecular he mossy fib

a2+ influx v ynaptic term ncrease in cA M1α, a highly

ortant in se

review: CAS

ificant func itionally, P se, PKC wa

ILLO, 2012). D are still no Two major n though t

idated, seve e suggested

ing to neuro resident of ribed to be tion could b smission.

r mechanism bres (MF) p via voltage minal will ac AMP and a y enriched p everal cAM

STILLO, 2012

ction in pr KA may r as reported t

Overall, th ot fully eluci players in he exact m eral cellula

to modulat otransmitter

the AZ (R actively inv be altered,

ms involved presynaptic gated calci ctivate the C activation of

protein in th MP/PKA-dep

) its direct resynaptic

egulate LT to be respon he precise m

idated.

synaptic p mechanisms

r processes e this event rs release, it RIM1α), the

volved in ta via posttran

d in presyna long-term ium channe Ca2+ /calmo f PKA (revie

he active zo pendent form

phosphory LTP (KAES

TP independ nsible for L molecular m

lasticity s governing s, like SV t. Since SV

t’s imperiou e other a co

aking part i nslational m

F In th A is da co ph su pr M M M

aptic LTP potentiation els (VGCG) odulin depen

ew: CASTILLO

one. Althoug ms of pres ylation by P

SER et al.,

dently of R LTP in MF-

mechanism

g the pres priming or priming is us to be un omponent o in the regul modification

Figure 1.5:

ncreased actio he inhibitory Adenylyl cycla

s impaired an ark blue) act onsequence hosphorylated usceptible to

roteasome.

Munc13 protein Model adapte MENNERICK, 20

n (LTP) is i ). The incre ndent-adeny

O, 2012). One gh RIM1α h ynaptic pla PKA could

2008a; YANG

RIM1α pho dentate gyr governing

synaptic pla r posttransl

one of the der a tight of synaptic ation of pri ns, triggerin

Presynaptic on potential fi y G protein

ase (AC, ligh nd protein kin tivity is decr

RIM1α, d (red ma

o degradatio Together w in (green) is a ed after CRA

012.

induced by ease in calc ylyl cyclase e of the PKA has been rep asticity (CAS

not be lin

G and CALA

osphorylatio rus basket c presynapti

asticity are lational mo most impor control. Tw vesicle (SV iming. Mor ng changes

dormancy.

iring activates (Gi, pink), t blue) action nase A (PKA, reased. As a is less arkings) and on, via the with RIM1, also degraded.

AWFORD and

an increase cium in the e, leading to A targets is ported to be

STILLO et al.,

nked to any

AKOS, 2010). on. Another cells (review:

ic LTP and

e not fully odifications, rtant events wo proteins, V2A), were eover, their in synaptic

. s , n , a s d e , . d

e e o s e

,

y . r

:

d

y , s , e r c

(15)

  1.5.1 1.5.1 The RIM distin (α, β RIM

lacks ZF. A α-isof

altern site A C2B and 3 separ block RIM prote (28 i for e and C rich-

1 RIMs 1.1 RIM gen

RIM protei M3 and RIM4

nct promote β, γ) (WANG a

M3γ and RIM

the Rab3 bin All γ-isoforms

forms.

With the native splic A in the N

domains. S 3) (WANG an

rately (FUKU

k, but indep M2. Splice si ein sequenc in RIM1 res

ither the de C, are separ domain (WA

ne structure in family c 4). RIM1 an ers: 2 isofor

and SÜDHOF,

M4γ, respect

nding site, wh are compose

e exception cing (Fig. 1 -terminal Z Splice site A

nd SÜDHOF, 2

UDA, 2003).

pendently, g ite C, splice e, has 3 ex spectively 2 eletion or th rated by onl

ANG and SÜDH

e

onsist of 7 nd RIM2 ge rms for RIM

2003). RIM tively (Fig. 1

ile RIM2β lac d of only the

n of the γ- 1.6B). The α Zn2+-finger d A contains

2003). In br Splice site giving rise t ed as a bloc

ons in RIM 25 in RIM2) he insertion

ly one exon

HOF, 2003). In

members e enes can dir M1 (α, β) (K

M3 and RIM

1.6A; WANG a

cks the entire C2B domain

-transcripts α- isoforms domain and 2 exons, c rain exons B contains to 64 possib ck to preven M1 (26-28) a ) includes a

of a small 9 n, 25 in RIM n compariso

encoded by rect the syn

KAESER et al.,

M4 genes enc

and SÜDHOF, 2

e N-terminal d . (B) Scheme

all other t s have three d splice site

onserved in 2 and 3 ca 6 exons (1 ble variants

nt the disru and 4 exons an alternativ 9 residue se M1 and 21 in

on to rat an

4 separate nthesis of di

, 2008b), and code each f

2003).

domain contai depicting the

ranscripts a e conserved e B and C n both RIM an be splice 19-24) in R

, and only 2 uption of the s in RIM2 ( ve splice ac equence. Th n RIM2, wh d mouse, sp

e genes (RIM ifferent isof d 3 isoform for one tran

Figure 1.6:

representatio RIM protein Domain st different R members.

(RIM1 and R the full set of (zinc finger d (post synap protein Drosophila tumor suppr and zonula protein- zo-1 C2B doma ining the α-he e alternative s

are subject d splicing s

between th M1 and RIM

ed either as RIM1, splic

2 exons (19 e reading fr (22-25). Th cceptor site he two splic hich contain plice site A

M1, RIM2, forms using ms for RIM2 nscript only,

: Schematic on of the n family. (A) tructure of RIM family α-isoforms RIM2) contain f domains: ZF domain), PDZ ptic density -PSD95, disc large ressor- Dlg1, occludens-1 1), C2A and ins. RIM1β elixes and the splicing of the

to intense sites: splice he C2A and M2 (exons 2 a block or ed not as a 9 and 20) in frame in the he last exon responsible cing sites, B ns a proline and exon 3 , g 2 ,

c e ) f y s n F Z y , e , d β e e

e e d 2 r a n e n e B e 3

(16)

  are m brain may

2003)

respo

MÜLL

1.5.1 RIM doma

1997, (KAES

isofo

(Fig.1

doma termi for o bind comp Mun Mun prim Mun

missing from n specific. A be respons

). In invert onsible for t

LER et al., 201

1.2 RIM pro M1α and RIM

ain, a centr

2000). RIM

SER et al., 20

orms contain

1. 6A; WANG e

The N-te ain, is impo inal α1 heli other Rab m

RIM2α) (F

posed of th nc13-1 and u nc13 exists ming. The in nc13 homod

m the human Another inte ible for the tebrates (C the synthesi

2).

otein structu M2α protein

al PDZ dom M1β lacks th

008b), RIM2 n only the l

et al., 2000; W

erminal dom ortant for th ix of RIM1α molecules (R

Fig.1.7; SUN

he zinc dom ubMunc13-

in an inac nteraction o dimerization

n transcript, eresting fea generation C.elegans, D

is of the RI

ure and bin ns contain t main, and tw

e α1-helix i 2β lacks th last C-termi

WANG and SÜD

main, contai he formation

α is sufficien Rab10, Rab

N et al., 2001

main, α2 he 2 (BETZ et al

ctive homo of RIM1α n (DENG et al.,

, while exon ature is the n of two oth D.melanoga IM protein (

nding partn the complet wo C- term in the N-ter he entire N inal C2B do

DHOF, 2003). ining two α n of the trip

nt to media b26 and Rab

1; WANG et

elix and SG

., 2001; DULU

odimerizatio with Munc

, 2011).

n 22 is retin alternative her splice va aster) only (unc-10) (KO

ers

te set of do minal C2 dom

rminal posit terminal Z omain and -helices (α1 partite comp ate binding t

b37 bind R

al., 2001; F

GAWFY m

UBOVA et al., 2

on state un c13 activate

na specific a splice site i ariants in th one gene

OUSHIKA et al

mains: an N mains: C2A tion, respon Zn2+- finger

a short isof 1 and α2) fl plex: Rab3/

to GTP- bou IM1α; Rab3

FUKUDA, 200

motif displa

2005; ANDREW

nable to pe es vesicle p

and exon 24 involving ex he retina (JO

was identi

l., 2001; GRA

N-terminal A and C2B (

nsible for Ra domain, w form specifi lanking the /RIM/Munc und Rab3A 3A/B/C/D

03), while t ays a high

WS-ZWILLING

erform its f priming by

Figure 1.7 representa RIM1α partners. A MITTELSTA

4 and 26 are xon 17 that

OHNSON et al.,

ified to be

F et al., 2012;

Zn2+-finger

(WANG et al.,

ab3 binding while the γ- ic sequence Zn2+-finger c13. The N- /B/C/D and and Rab8A the module affinity for

G et al., 2006). function in y impairing

7: Schematic ation of binding Adapted after

AEDt, 2010. e

t

,

e

;

r

,

g - e

r - d A e r . n g

c f g r

(17)

 

The sequence between the zinc and PDZ domains contains several amino acid residues that have been suggested to be important in modulating RIM’s function (Fig. 1.7). Serine 413 was identified as a phospho-switch that triggers presynaptic LTP in cultured cerebellar granular and Purkinje cell neurons, upon phosphorylation by PKA (LONART et al., 2003). These findings however were not confirmed by studies in knockin mice, bearing the S413A mutation. The phosphorylation of serine 413, although important in binding 14-3-3 proteins, displayed no significant role in presynaptic plasticity or in learning and memory (KAESER et al., 2008a; YANG and CALAKOS, 2010). Other phosphoserines (Ser241 and Ser287 in RIM1α, and Ser335 in RIM2α) were also associated with binding to 14-3-3 proteins, when phosphorylated by the Ca2+/calmodulin dependent kinase II (CaMKII). The ability of RIM to bind 14-3-3 proteins does apparently not impair the binding between RIM-Munc13 and RIM-Rab3A (SUN

et al., 2003). The same linker region between zinc finger and PDZ domain may also act as a substrate for ERK2 kinase, which phosphorylates Ser447, a residue linked to the enhancement of glutamatergic transmission in hippocampal CA1 after stimulation with BDNF (SIMSEK- DURAN and LONART, 2008).

The central PDZ domain that interacts with the ELKS2/CAST protein (OHTSUKA et al., 2002; WANG et al., 2002), plays an important role in RIM1’s distribution in cultured neurons; the truncated form lacking this domain being diffusely localized (Fig. 1.7; OHTSUKA et al., 2002). CAST binds directly not only RIM1, but also Bassoon and Piccolo, and the entire ternary complex RIM1-CAST-Bassoon is involved in controlling neurotransmitter release (TAKAO- RIKITSU et al., 2004). Two reports from 2011 attribute to RIM1/2 a key role in controlling not only the number of docked vesicles but also the distribution and/or density of calcium channels at the active zone (HAN et al., 2011; KAESER et al., 2011). By generating RIM1/2 floxed mouse lines, in which all RIM isoforms containing a PDZ domain can be deleted by cre- recombinase in vitro, it was shown that the PDZ domain alone was required for the proper localization of N- and P/Q type calcium channels (KAESER et al., 2011).

The α- and β-RIMs contain two C-terminal domains: C2A and C2B that are separated by a proline-rich domain and two splice sites (B and C) (Fig. 1.7; WANG and SÜDHOF, 2003). Both domains do not contain the consensus calcium binding sites present in synaptotagmin’s C2- domains (WANG et al., 2000; DAI et al., 2005). The C2A domain was shown to have affinity in a calcium dependent manner for SNAP25 and Synaptotagmin1 (COPPOLA et al., 2001), even though NMR studies suggested that there was little binding between these proteins (DAI et al., 2005). Very intriguing is a point mutation in human RIM1 (R844H) that was identified in a patient with autosomal dominant cone-rod dystrophy-CORD7, characterized by impaired vision due

(18)

 

to the reduction in the cone and rod sensitivity (JOHNSON et al., 2003; MICHAELIDES et al., 2005). The C2B domain has been shown to interact with several proteins that may have an impact on RIM1α function at the active zone, among them Synaptotagmin1, identified to bind with high affinity to the C2B domain in biochemical assays (COPPOLA et al., 2001; SCHOCH et al., 2002), results not reproduced by NMR studies (GUAN et al., 2007). Other proteins that bind the C2B domain are: liprins-α (SCHOCH et al., 2002); the E3 ubiquitin ligase SCRAPPER (YAO et al., 2007)

that controls RIM1 turn-over, facilitating ubiquitination and degradation; SAD kinase (INOUE et al., 2006); and the β4 subunit of voltage gated calcium channels (COPPOLA et al., 2001; KIYONAKA et al., 2007). In addition the interaction between RIM1 and the α1 subunit of the N-type calcium channel is regulated by cyclin-dependent kinase 5 (Cdk5), which enhances channel opening and facilitates neurotransmitters release (SU et al., 2012).

SUMOylation was recently reported by the group of Hanley to act as a molecular switch for RIM1α. SUMOylated RIM1α confers affinity for Cav2.1, therefore promoting calcium channel clustering and synchronous synaptic vesicle release, while non-SUMOylated form is responsible only for vesicle priming and docking (GIRACH et al., 2013).

Other proteins that couple RIM1/2 to calcium channels are RIM-BPs. On one hand RIM-BP binds the proline-rich domain of RIM1/2 (WANG et al., 2000) and on the other hand calcium channels, bringing these proteins in close proximity at the active zone (HIBINO et al., 2002).

1.5.1.3 RIM function

1.5.1.3.1 RIM in invertebrates (C.elegans and D.melanogaster)

Analysis of RIM protein function in C.elegans demonstrated that UNC-10 has a major role in coordinating vesicle docking and priming by regulating UNC-13 activity. It has been hypothesised that UNC-10/RIM may signal syntaxin, via UNC-13, to change its conformation from a closed to an open state. UNC-10 mutants exhibit a decrease in vesicle fusion at release sites, an effect suppressed by the expression of the open form of syntaxin (KOUSHIKA et al., 2001). Furthermore, disruption of the unc-10 gene triggers a depletion of docked synaptic vesicles since the normal connections between SVs and dense projection filaments are impaired (STIGLOHER et al., 2011).

D.melanogaster RIM mutants show decreased evoked synaptic transmission as a consequence of the reduction in the size of the RRP of SVs and altered Ca2+-channels clustering together with a decreased calcium influx. Mutants present a normal cellular morphology with no major changes in active zone architecture (GRAF et al., 2012; MÜLLER et al., 2012).

(19)

 

1.5.1.3.2 RIM in vertebrates (M.musculus)

In the recent years several reports have been published, providing new data about the possible role of RIMs at the active zone. Different mouse models have been generated, knocking out either one or more isoforms, in order to gain new insights into how different variants of RIMs influence neurotransmitter release and presynaptic plasticity as well as to understand ability of the various isoforms to compensate for each other.

1.5.1.3.2.1 RIM1α knock-out mice

The first model generated targeted the most abundant isoform in the brain, RIM1α (SCHOCH et al., 2002). Homozygous mice were viable and fertile, with no evident structural abnormalities or changes in brain architecture. Overall, active zone architecture was comparable to WT littermates. Among the AZ proteins, Munc13-1 showed a major decrease of 60% in KOs, while several postsynaptic density proteins (SynGAP, PSD95, SHANK) exhibited a moderate increase, suggesting a role for RIM1α in synaptic remodelling (SCHOCH et al., 2002). Electrophysiological recordings revealed that RIM1α knockout caused a decrease in the size of the RRP, with no effect on synaptic vesicle recycling. These data together with findings from D.melanogaster and C.elegans suggest a role for RIM1α in vesicle maturation, from priming to calcium triggered fusion (KOUSHIKA et al., 2001; SCHOCH et al., 2002; CALAKOS et al., 2004;

MÜLLER et al., 2012). Additionally, the RIM1α protein seems to be involved both in short-term plasticity as well as in presynaptic long-term potentiation (LTP) (review: MITTELSTAEDT et al., 2010).

Cryo-electron tomography revealed a series of changes in the AZ with regard to vesicle tethering and vesicle concentration in synaptosomes from RIM1α KO mice (40%

reduction in proximal vesicles compared to control) that may account for the decrease in the size of the RRP. Blocking proteasome activity with MG132, the KO phenotype was rescued and the treated KO synaptosomes became indistinguishable from WT synaptosomes, displaying an increase in the number of vesicles at the AZ. This recent study highlights the importance of the ubiquitin-proteasome system (UPS) in the turn-over of RIM proteins, emerging as a key factor in controlling presynaptic plasticity (FERNANDEZ-BUSNADIEGO et al., 2013).

Besides deficits in synaptic transmission, KO mice display impaired learning and memory (POWELL et al., 2004), schizophrenia-like behaviour (BLUNDELL et al., 2010), and a higher susceptibility to develop spontaneous seizures after status epilepticus (PITSCH et al., 2012).

(20)

 

1.5.1.3.2.2 RIM1αβ double knock-out mice

Mutant mice lacking both RIM1 isoforms, α and β, display a more severe impairment in synaptic transmission and significant changes in the solubility of different active zone proteins. Both isoforms are expressed in a similar pattern in the brain, with a slight increase of RIM1β levels in the brainstem. During development RIM1β is highly expressed in the early postnatal phase in this region, which may account for the lethality of the DKO mice.

Interestingly, in RIM1α KO mice the level of RIM1β is increased 2 fold, indicating a compensatory effect. Among the presynaptic proteins, ELKS1/2, RIM-BP2 and the remaining Munc13-1 (reduced to 30% in these mutant mice), showed a higher dissociation rate from the insoluble protein matrix, supporting the notion of RIMs acting as scaffolding proteins for various AZ proteins. Synaptic transmission is severely impaired in the DKO mice with the observation that presynaptic long-term plasticity is not aggravated by this double deletion compared to RIM1 KO. Therefore, it has been suggested that RIM1α mediates both long-term plasticity via Rab3 as well as short-term plasticity via Munc13, while RIM1β (since it lacks the binding motif for Rab3) is involved only in short-term plasticity (KAESER et al., 2008b). 1.5.1.3.2.3 RIM2α knock-out mice

Since RIM1α and RIM2α, which is much less abundant, display high homology, it was expected that the knockout of RIM2α might partially resemble the phenotype of the RIM1α KO. However, deletion of the RIM2α gene did not trigger any change in release probability compared to the impairment in synaptic transmission and facilitation observed in the RIM1α KO mice (CASTILLO et al., 2002; SCHOCH et al., 2002, 2006). RIM2α KO mice were viable and fertile, and displayed normal brain morphology (SCHOCH et al., 2006).

1.5.1.3.2.4 RIM1α/RIM2α double knock-out mice

Deletion of both α isoforms (RIM1 and RIM2) turned out to be lethal, RIM1α/2α DKO mice die immediately at birth, not due to changes in brain development but due to breathing problems. No obvious alterations in brain morphology were detected by conventional EM.

Protein composition analysis revealed no additional decrease in the level of Munc13-1 compared to RIM1α KO mice. Nonetheless, immunostaining analysis of the whole-mount diaphragm muscle at E18.5 revealed an increased innervation or expansion of innervation with no major changes in the ultrastructure of the NMJ in the DKO mice. These changes were accompanied by impairment in synaptic transmission. Spontaneous or Ca2+-dependent exocytosis was not abolished, only evoked synaptic transmission (Ca2+- triggering exocytosis) was strongly impaired in these mutants (SCHOCH et al., 2006).

(21)

 

1.5.1.3.2.5 RIM conditional knockout mice

As both RIM1α/RIM1β (KAESER et al., 2008b) and RIM1α/RIM2α (SCHOCH et al., 2006) DKO mice were lethal, conditional knockouts (floxed mouse lines) were generated to further study the consequences of a deficiency of all RIMs isoforms. Deletion of both RIM genes in vitro supported the role of RIMs in controlling vesicle priming and neurotransmitter release (KAESER et al., 2012). Furthermore, RIMs were shown to be responsible for proper tethering of the Ca2+

channels via the PDZ domain (HAN et al., 2011; KAESER et al., 2011).

Single deletions (RIM1αβ or RIM2αβγ) altered SV priming, while double deletion (RIM1αβ/RIM2αβγ) impaired not only the priming but also the calcium responsiveness and synchronization of release. In HEK293T cells and in RIM1/2 double deficient neurons, RIM2γ wasn’t able to rescue the phenotype, suggesting that the C2 domain alone neither contributes to calcium channel activity modulation nor plays an important role in the synaptic function of RIM proteins (KAESER et al., 2012).

Taken together, RIM1α plays an important role in synaptic vesicle priming, and in both presynaptic short-term and long-term plasticity. Moreover, the level of RIM1α seems to be correlated with the synaptic activity.

(22)

  1.5.2 1.5.2 Syna integ (SV2 have distri the s SV2A C2B facili presu

(PYLE

chan

was i the t vesic regio howe (TMR local foldi NAD level

2 Synaptic v 2.1 SV2A fu aptic vesicl grated in th

2A, SV2B a a more ub ibution. Ho same neuro A, but not S

domain o itated by p umably the

E et al., 2000), nges in synap

identified to trafficking o cles (YAO et

on appears n ever. Additi R1) and in lization or

ng (CHANG a

Interestin D), pointing

ls (YAO and B

vesicle prote unction

le proteins he synaptic and SV2C), biquitous ex owever, both ons and eve

SV2B, was of synaptota phosphoryla affinity of , and inhibit

ptotagmin1

o be require of Synaptot

al., 2010). C not to play, ionally, mu n the glyco

abolish com

and SÜDHOF, 2

ngly, SV2 p g to the fact

BAJJALIEH, 20

ein 2A (SV2

(SV2s) ar c vesicles m

expressed xpression t h isoforms, en on the s

reported to agmin1 (SC

ation event the N-term ted by high , determinin

ed for bindin tagmin1 an Chang and S if any, a m tations of d osylation si mpletely S

2009; NOWAC

proteins bin that these p

008).aaaaaaaaa

V2A)

are a class membrane

only in neu throughout

SV2A and ame synapt o interact di

CHIVELL et a

ts of differ minal domain h concentrat

ng its dissoc

ng to differe nd affecting Südhof (2009

major role in different am ite were re V2A funct

CK et al., 2010

nd with hig proteins’ ac

aaaaaaaaaaaaa

of transm (Fig.1.8), w urons and en

the brain, d B, can be tic vesicles rectly throu

l., 1996). T rent serine n toward th ion of calci ciation from

Figu SV2 SV2 glyc pink to Syn clath

N-t ent clathrin g in this ma

9), whose re SV2A func mino acid res eported to tion, by alt

0).

gh specificit ctivity may

aaaaaaaaaaaaa

membrane g with three c

ndocrine ce while SV2B found at va

(BAJJALEH e

ugh its N-ter his binding /threonine he calcium s

um, which m SV2A (SC

ure 1.8: Sche 2A structure 2A is a 12-pa coprotein (gl k). The N-term

bind to aptotagmin 1 hrin adaptor p

The en terminal reg

adaptor pro anner its am esults show ction, contra sidues in tra either imp tering its n ty adenine be modulat

aaaaaaaaaaaaa

glycosylate characterise ells. SV2A i B has a m arying ratio

et al., 1992;

erminal regi g was repo

residues, p sensor syna

induce conf

CHIVELL et al.,

ematic repre and its bindi ass transmem lycosylation minal region

the C2B 1(light blue) proteins (grey)

ndocytosis m gion of SV2 oteins, cont mount on th wed that the

adicted this ansmembra pair SV2A normal proc nucleotides ted by diffe

aaaaaaaaaaaaa

d proteins, ed isoforms is known to more limited s though in

1993; 1994). on with the orted to be potentiating aptotagmin1 formational

, 1996).

esentation of ing partners.

mbrane (TMR) depicted in was reported domain of and different ).

motif in the 2A (YSRF) rolling thus he synaptic N-terminal s hypothesis ane region 1 subcellular cessing and s (ATP and rent energy

aaaaaaaaaaaaa

, s o d n . e e g

l

f . ) n d f t

e ) s c l s r d d y

a

(23)

 

1.5.2.2 SV2A knock-out mice

In spite of all the data collected until now the exact function of SV2A still remains enigmatic.

To gain further insights into SV2A function, SV2A deficient mice were generated (CROWDER et al., 1999; JANZ et al., 1999). Albeit SV2A KO littermates appeared normal at birth, mice experienced severe seizures and died about three weeks after birth. No obvious alterations of synaptic density or morphology in the brain of SV2A KO mice were observed (CROWDER et al., 1999; JANZ et al., 1999). Therefore, SV2A seems not to be required in embryonic development but rather its presence is essential for survival afterwards. Electrophysiological studies further revealed that inhibitory (CROWDER et al., 1999; CHANG and SÜDHOF, 2009) as well as excitatory

(CUSTER et al., 2006) neurotransmission in these mice were impaired. A similar impairment was also detected in adrenal chromaffin cells from SV2A KO mice, where the exocytotic burst defining the size of the readily releasable pool (RRP) was observed to be decreased with no evident alterations in the calcium level (XU and BAJJALIEH, 2001). A role in priming after vesicle tethering was suggested by Custer et al. (2006), who observed a similar decrease in RRP in the SV2A deficient mice’s brain, with no oscillation in calcium level.

However, earlier studies using SV2A/SV2B double knockout mice with a phenotype resembling SV2A KO, proposed a role in regulating the calcium level during repetitive stimulation trains rather than priming (JANZ et al., 1999). The described decrease in the RRP size

(CUSTER et al., 2006) was not reproduced by Chang (CHANG and SÜDHOF, 2009). A further observation that the protein components of SNARE complexe were reduced in SV2A KO mice supported the hypotheses that SV2A may have a role in the fusion mechanism (XU and BAJJALIEH, 2001).

Taken together, the collected data suggest a role of SV2A in SV priming. Moreover, SV2A act as a receptor for the anti-epileptic drug Keppra. It has been suggested that Keppra may inhibit inappropriate interactions to occur when SV2A is overexpressed in neuronal cell cultures. Neurons with elevated amount of overexpressed SV2A display similar impairments in synaptic transmission as neurons from SV2A KO mice (NOWACK et al., 2011). It seems that the protein amount plays an important role in maintaining the neuronal function as well. The molecular mechanism of action of Keppra on SV2A is not fully elucidated.

Referenzen

ÄHNLICHE DOKUMENTE

(2002): The role of iron in Actinobacillus pleuropneumoniae infection: Identification and in vivo characterization of virulence-associated genes.. Tierärztiche Hochschule

Replacement of the benzothiazolyl group of GK181 by a benzoxazolyl one (GK491, entry 5) led led to a slight decrease of the activity (77%) in comparison to GK181, while the

While there was no difference in the overall spine density between wild-type and Cpne 6D167N mutant mice (Figure 6C), morphometric analysis of dendritic spines from

Cdk5rap2 mutant mice have small brains with abnormal orientation and morphology of mitotic spindles in neural progenitors, premature neuronal differentiation, reduced proliferation,

In the middle of the sarcomere lies the darker A-band (“Anisotropic” in polarized light). Based on fine-structure in electron.. The main components of the

Because protein kinases require the interaction of the P+1 loop with the C-loop for substrate binding and since such an interaction is fostered intermolecularly in the present

Furthermore, when assessing the CNP knockout we found that only 0.4 ± 0.06 percent of the assessed myelin sheaths had non-compacted layers (fig 3.32). Taken all together,

Integrin Activation Contributes to Lower Cisplatin Sensitivity in MV3 Melanoma Cells by Inducing the Wnt Signalling Pathway. Fuereder T, Wanek T, Pflegerl P, Jaeger-Lansky