Cd k5 Ph o s p h o r y l at i o n o f Mu n c18-1 En h a n c e s Sy n a p t i c

Similar documents
Supplementary Materials for VAMP4 directs synaptic vesicles to a pool that selectively maintains asynchronous neurotransmission

Supplementary Figure 1. SybII and Ceb are sorted to distinct vesicle populations in astrocytes. Nature Neuroscience: doi: /nn.

THE SYNAPTIC VESICLE CYCLE

SUPPLEMENTARY INFORMATION

Presynaptic inhibition upon CB1 or mglu2/3 receptor activation requires ERK/MAPK phosphorylation of Munc18-1

BIPN 140 Problem Set 6

BIPN 140 Problem Set 6

Supplementary Figure 1

Part 11: Mechanisms of Learning

Abstract. 1 Introduction

Supporting Information

Neurons! John A. White Dept. of Bioengineering

SUPPLEMENTARY INFORMATION. Supplementary Figure 1

Synaptic Transmission: Ionic and Metabotropic

1) Drop off in the Bi 150 box outside Baxter 331 or to the head TA (jcolas).

Chapter 3 subtitles Action potentials

MCB MIDTERM EXAM #1 MONDAY MARCH 3, 2008 ANSWER KEY

Autonomous Function of Synaptotagmin 1 in Triggering Synchronous Release Independent of Asynchronous Release

The effects of temperature on vesicular supply and release in autaptic cultures of rat and mouse hippocampal neurons

Synaptotagmin-7-Mediated Asynchronous Release Boosts High-Fidelity Synchronous Transmission at a Central Synapse

Competition between Phasic and Asynchronous Release for Recovered Synaptic Vesicles at Developing Hippocampal Autaptic Synapses

File name: Supplementary Information Description: Supplementary Figures, Supplementary Table and Supplementary References

Common strength and localization of spontaneous and evoked synaptic vesicle release sites

Charlie Taylor, PhD CpTaylor Consulting Chelsea, MI, USA

Reliable and sustainable neurotransmitter release is essential for

Dep. Control Time (min)

TITLE: Altered Astrocyte-Neuron Interactions and Epileptogenesis in Tuberous Sclerosis Complex Disorder

Supplementary Figure 1. mir124 does not change neuron morphology and synaptic

Axon initial segment position changes CA1 pyramidal neuron excitability

Chapter 3 Neurotransmitter release

Is Intrinsic Hyperexcitability in CA3 the Culprit for Seizures in Rett Syndrome?

Enhancement of synaptic transmission by cyclic AMP modulation of presynaptic I h channels. Vahri Beaumont and Robert S. Zucker

Modelling Vesicular Release at Hippocampal Synapses

Cellular Neurobiology / BIPN 140

Supplementary Information

Ultrastructural Contributions to Desensitization at the Cerebellar Mossy Fiber to Granule Cell Synapse

Presynaptic NMDA receptor control of spontaneous and evoked activity By: Sally Si Ying Li Supervisor: Jesper Sjöström

Vesicular Trafficking of Semaphorin 3A is Activity- Dependent and Differs Between Axons and Dendrites

Synaptotagmin-1, -2, and -9: Ca 2+ Sensors for Fast Release that Specify Distinct Presynaptic Properties in Subsets of Neurons

7.06 Spring of PROBLEM SET #6

Readily releasable pool size changes associated with long term depression

Synaptotagmin-1- and Synaptotagmin-7-Dependent Fusion Mechanisms Target Synaptic Vesicles to Kinetically Distinct Endocytic Pathways

The action potential travels down both branches because each branch is a typical axon with voltage dependent Na + and K+ channels.

Authors: K. L. Arendt, M. Royo, M. Fernández-Monreal, S. Knafo, C. N. Petrok, J.

Anatomy Review. Graphics are used with permission of: Pearson Education Inc., publishing as Benjamin Cummings (

Project report October 2012 March 2013 CRF fellow: Principal Investigator: Project title:

Local Presynaptic Activity Gates Homeostatic Changes in Presynaptic Function Driven by Dendritic BDNF Synthesis

Supplementary figure 1: LII/III GIN-cells show morphological characteristics of MC

1.0. FSL NMDAR-fEPSP 0.8. amplitude (mv) Intensity (µa) 2.0 SD FSL Time (ms)

Chapter 6 subtitles postsynaptic integration

Suppl. Information Supplementary Figure 1. Strategy/latency analysis of individual mice during maze learning. a,

Synaptic Transmission

Impaired GABAergic inhibition in the visual cortex of brain-derived neurotrophic factor heterozygous knockout mice

SUPPLEMENTARY INFORMATION

CDK5ServesasaMajorControlPoint in Neurotransmitter Release

TITLE: Altered Astrocyte-Neuron Interactions and Epileptogenesis in Tuberous Sclerosis Complex Disorder

Supplementary Information

Supplementary Figure 1. Basic properties of compound EPSPs at

Alterations in Synaptic Strength Preceding Axon Withdrawal

Compensatory Contribution of Ca V. 2.3 Channels to Acetylcholine Release at the Neuromuscular Junction of Tottering Mice

DOI: /jphysiol The Physiological Society Rapid Report

Structure of a Neuron:

Elizabeth Biopsychology (PSY 302) The Synapses 08/29/2017. The Synapses

QUIZ YOURSELF COLOSSAL NEURON ACTIVITY

Chapter 45: Synapses Transmission of Nerve Impulses Between Neurons. Chad Smurthwaite & Jordan Shellmire

5-Nervous system II: Physiology of Neurons

The mammalian cochlea possesses two classes of afferent neurons and two classes of efferent neurons.

Structural basis for the role of inhibition in facilitating adult brain plasticity

Ube3a is required for experience-dependent maturation of the neocortex

Supplementary Figure 1. Microglia do not show signs of classical immune activation following MD a-b. Images showing immunoreactivity for MHCII (a)

Supplemental information Acid-sensing ion channel 1a contributes to hippocampal LTP inducibility through multiple mechanisms

NEURONS COMMUNICATE WITH OTHER CELLS AT SYNAPSES 34.3

Chapter 2: Cellular Mechanisms and Cognition

Transient Calcium and Dopamine Increase PKA Activity and DARPP-32 Phosphorylation

The Journal of Neuroscience, December 15, 2002, 22(24):

Synapses and Neurotransmitters

How Nicotinic Signaling Shapes Neural Networks

Formation, stabilisation and fusion of the readily releasable pool of secretory vesicles

Name: Answer Key. Question 1.

PKA-catalyzed phosphorylation of tomosyn and its implication in Ca 2 -dependent exocytosis of neurotransmitter

Synaptic Plasticity and the NMDA Receptor

What Cell Make Up the Brain and Spinal Cord

Action potentials propagate down their axon

Neuronal Differentiation: CNS Synapse Formation

Problem Set 3 - Answers. -70mV TBOA

Activity Dependent Changes At the Developing Neuromuscular Junction

Introduction to Neurobiology

How Synapses Integrate Information and Change

Introduction to the EEG technique

Resonant synchronization of heterogeneous inhibitory networks

1) Drop off in the Bi 150 box outside Baxter 331 or to the head TA (jcolas).

P/Q And N Channels Control Baseline and Spike-Triggered Calcium Levels in Neocortical Axons And Synaptic Boutons

Electrical Properties of Neurons. Steven McLoon Department of Neuroscience University of Minnesota

Properties of Synchronous and Asynchronous Release During Pulse Train Depression in Cultured Hippocampal Neurons

BIPN140 Lecture 8: Synaptic Transmission II

Beyond Vanilla LTP. Spike-timing-dependent-plasticity or STDP

Astrocyte signaling controls spike timing-dependent depression at neocortical synapses

Synaptic Plasticity and Memory

How Synapses Integrate Information and Change

Transcription:

Cd k5 Ph o s p h o r y l at i o n o f Mu n c18-1 En h a n c e s Sy n a p t i c Tr a n s m i s s i o n Sabine K Schmitz 1 Matthijs Verhage 1 Ruud F Toonen 1 1 Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam and VU Medical Center, Amsterdam, The Netherlands in preparation

Abstract Cyclin-dependent kinase 5 (Cdk5) functions in many neuronal processes and has recently been implicated in homeostatic plasticity. Cdk5 activation results in reallocation of synaptic strength, including weakening of some connections while strengthening others. The molecular mechanisms underlying these phenomena are thus far not understood. We show that Cdk5 phosphorylation of the presynaptic protein Munc18-1 plays an important role in the regulation of synaptic strength. In the absence of phosphorylation, neurons show reduced spontaneous and evoked release, RRP size and release probability. Hence, Cdk5 phosphorylation of Munc18-1 does not underlie Cdk5-dependent silencing of synapses, but may play a role in neuronal silencing induced increase of presynaptic strength. 96

Introduction Cyclin-dependent kinase 5 (Cdk5), a proline-directed serine/threonine kinase, is highly expressed in postmitotic neurons (Cruz and Tsai, 2; Meyerson et al., 1992; Tsai et al., 1993). Activated by small regulatory proteins p35 and p39, Cdk5 functions in many neuronal processes, ranging from neuronal development and outgrowth (Ko et al., 21; Nikolic et al., 1996; Ohshima et al., 1996), neurogenesis (Lagace et al., 28), protein trafficking (Samuels et al., 27), synaptic transmission (Kim and Ryan, 21), addiction (Meyer et al., 28; Takahashi et al., 25; Taylor et al., 27) and learning (Fischer et al., 25; Fischer et al., 22, 23; Hawasli et al., 27; Sananbenesi et al., 27). In recent studies, Cdk5 has been implicated in homeostatic plasticity (Kim and Ryan, 21; Mitra et al., 211). Homeostatic plasticity commonly describes cellular processes by which a neuron regulates its synaptic strength to stabilize network activity (Davis, 26; Pozo and Goda, 21; Turrigiano, 211). Cdk5 activity increases upon presynaptic neuronal silencing (Mitra et al., 211). Increased Cdk5 activity upon chronic inhibition results in non-uniform homeostatic adaptation of synaptic strength: On the one hand, Cdk5 activity decreases functional connectivity between CA3-CA3 pyramidal cells without obvious loss of synapses. On the other hand, synaptic strength is increased in the remaining recurrent connections. Whereas loss of connectivity could be explained by Cdk5-dependent silencing of synapses and reserve pool modulation (Chergui et al., 2; Kim and Ryan, 21), the mechanisms underlying increased synaptic strength upon Cdk5 activation remain entirely unknown. Although many presynaptic targets of Cdk5, including Synapsin (Matsubara, 1996), Septin 5 (Amin et al., 28), CASK (Samuels et al., 27), CRMP2 (Cole et al., 28), Munc18-1 (Shuang et al., 1998) and the CDK5-like kinase Pctaire (Cheng et al., 22; Fu et al., 211; Liu et al., 26), have been described, the effect of their Cdk5 phosphorylation on neurotransmitter release mostly remains elusive. Hence, it is currently not understood which downstream targets mediate the effect of Cdk5- dependent reallocation of synaptic strength upon presynaptic neuronal silencing. Here, we show that Cdk5 phosphorylation of the presynaptic protein Munc18-1 regulates synaptic strength. Munc18-1, a member of the SM protein family is essential for synaptic vesicle fusion and functions in 97

various steps of the synaptic vesicle cycle (Toonen and Verhage, 27; Verhage et al., 2). Cdk5 phosphorylates Munc18-1 at threonine 57 (T57) (Shuang et al., 1998). This phosphorylation underlies Cdk5 s release enhancing effect in secretory cells (Chergui et al., 2; Fletcher et al., 1999; Lilja et al., 21), but it s role in neurons remains elusive. We investigated the role of Munc18-1 phosphorylation by Cdk5 in munc18-1 null mutant hippocampal neurons infected with either nonphosphorylatable Munc18 or Munc18 as control. Inhibiting Cdk5 phosphorylation of Munc18-1 does not significantly influence neuronal morphology, but results in a strong decrease of both spontaneous and evoked release. Munc18 neurons show paired-pulse facilitation and slower rundown kinetics during action potential trains. Hence, Cdk5 phosphorylation of Munc18-1 enhances synaptic vesicle release and might underlie the Cdk5-dependent strengthening of synapses after presynaptic inhibition. Results To investigate the role of Munc18-1 phosphorylation by Cdk5 in neurons, we infected hippocampal autaptic neurons from munc18-1 null mutant mice with Lentiviruses expressing either the non-phosphorylatable Munc18-1 variant Munc18 (M18 ) or Munc18 (M18 ) as control and studied their electrophysiological properties. Cdk5 phosphorylation of Munc18-1 enhances evoked and spontaneous release. Neurons rescued with M18 showed a significantly smaller evoked postsynaptic current (EPSC) amplitude compared to neurons rescued with M18 (Figure.1A-B). To distinguish between pre- and postsynaptic defects underlying the changes in EPSC amplitude, we investigated the effects of Cdk5 phosphorylation of Munc18-1 on spontaneous release (Figure.1C). Similarly to EPSC size, spontaneous miniature EPSC frequency was lower in M18 (Figure.1D), while mepsc amplitudes and kinetics were unchanged (Figure.1E-G). Hence, Cdk5 phosphorylation of Munc18-1 promotes spontaneous and evoked release by a presynaptic mechanism in hippocampal neurons. 98

A EPSC amplitude (na) mepsc amplitude (pa) 3 2 1 2 15 1 5 ** B C.5nA 1ms pa 5ms E F G mepsc rise time (ms) 1.5 1..5 D mepsc frequency (Hz) mepsc decay time (ms) 6 2 2. 1.5 1..5 ** Figure.1 Cdk5 phosphorylation of Munc18-1 enhances evoked and spontaneous synaptic vesicle release. Hippocampal autaptic neurons of munc18-1 null mutant mice were rescued with Lentiviruses expressing M18 or M18. (A) Amplitude of excitatory synapse response evoked by an action potential (M18 : 2.9 ±.2nA, n=65; M18 : 1. ±.16nA, n=79; **, Mann-Whitney test, p=.5). (B) Typical example of evoked release. Scale bar represents 1ms and.5na. (C) Typical example of spontaneous vesicle release. Scale bar represents.5s and pa. (D) Spontaneous release frequency (M18 : 3.1 ±.78Hz, n=57; M18 : 11.71 ±.5Hz, n=82; **, Mann-Whitney test, p=.1). (E) Amplitude of spontaneous release events (M18 : 16.36 ±.9pA, n=52; M18 : 15.36 ±.6pA, n=61; ns., Mann-Whitney test, p>.5). (F) Mean rise time of spontaneous release events (M18 : 1.3 ±.8ms, n=52; M18 : 1.1 ±.6ms, n=61; ns., Mann- Whitney test, p=.71). (G) Mean decay time of spontaneous release events (M18 : 1.6 ±.5ms, n=27; M18 : 1.53 ±.6ms, n=1; ns., Mann-Whitney test, p>.5). All data are expressed as mean ± SEM. Changes in neuronal morphology do not account for changes in neurotransmitter release observed in Munc18 neurons. To investigate whether the observed changes in neurotransmitter release are caused by underlying changes in neuronal morphology, autaptic neurons were fixed and immunostained for MAP2 as dendrite marker and VAMP2 as synapse marker (Figure.2A). No significant changes in 99

C 16 1 12 1 8 6 2 D number of dendritic branches B dendrite length (μm) A **.8. synaptic VAMP intensity (a.u.) synapse area (μm2) number of synapses 2 1 2 5 2 15 1 5 15 2 15 1 5 5 1 15 2 distance from soma (μm) H 25 1 ** 2 synaptic VAMP intensity (a.u.) synapse number 3 G 1.6 6 E F 8 distance from soma (μm) 1 3 *** 2 1 5 1 15 2 distance from soma (μm) Figure.2 Neuronal morphology cannot account for the electrophysiological effects of Cdk5 phosphorylation of Munc18. Hippocampal autaptic neurons of munc18-1 null mutant mice were rescued with Lentiviruses expressing M18 or M18 and stained for the dendritic marker MAP2 (red) and synapse marker VAMP2 (green). (A) Typical examples. Scale bar represents 5μm. (B) Mean dendrite length per cell (M18: 1336 ± 6μm, n=38; M18: 197 ± 83μm, n=3; ns., Mann-Whitney test, p>.5). (C) Number of dendritic branches as a function of radial distance from the soma quantified by Sholl analysis. (D) Mean synapse number per cell (M18: 25 ± 15, n=38; M18: 37 ± 27, n=3; ns., Mann-Whitney test, p>.5). (E) Number of synapses as a function of radial distance from the soma. Significance was tested by Mann-Whitney test at the following distances: μm: ***, p<.1; 1μm: *, p=.25; 2μm: ns., p>.5; 3μm: ns., p>.5; μm: ns., p>.5; 5μm: ns., 1

p>.5; 1μm: ns., p>.5; 15μm: ns., p>.5; 2μm: ns., p>.5. (F) Mean synapse area (M18 : 9 ±.μm2, n=38; M18 : 1.8 ±.5μm2, n=3; **, Mann-Whitney test, p=.5). (G) Mean synaptic VAMP2 intensity (M18 : 1997 ± 97, n=38; M18 : 2259 ± 99, n=3; ns., Mann-Whitney test, p=.6). (H) Mean synaptic VAMP2 intensity as function of radial distance from the soma. Significance was tested by Mann-Whitney test at the following distances: μm: *, p=.2; 1μm: *, p=.1; 2μm: *, p=.2; 3μm: ns., p>.5; μm: ns., p>.5; 5μm: ns., p>.5; 1μm: ns., p>.5; 15μm: ns., p>.5; 2μm: ns., p>.5. All data are expressed as mean ± SEM. dendritic length or branching were observed between mutant and wild-type Munc18-1 rescued neurons (Figure.2B-C). Although not significantly different we observed a trend towards higher synapse numbers in M18 neurons compared to M18 rescued neurons (Figure.2D), resulting from an increase in synapses onto proximal dendrites (Figure.2E). Moreover, synapse area was increased in M18 cells (Figure.2F-H) and synaptic VAMP2 intensities tended to be higher, arguing for a larger total synaptic vesicle pool in these groups. Like the trend on synapse number, the effect on VAMP2 levels is mediated by a preferential increase at proximal synapses (Figure.2H). To conclude, changes in the Cdk5 phosphorylation site of Munc18-1 lead to a relatively small increase in synapse number, area and VAMP2 levels close to the soma in M18 rescued neurons. Nonetheless, synaptic vesicle release is decreased in M18 neurons. Cdk5 phosphorylation of Munc18-1 influences RRP size The amount of neurotransmitter release is dependent on the size of the readily releasable pool (RRP) and the release probability of an individual vesicle within that pool (P ves ). To distinguish between the two, we probed the RRP by application of 5mM hyperosmotic sucrose solution (Rosenmund and Stevens, 1996). RRP sucrose size in M18 neurons was smaller compared to M18 neurons (Figure.3A-B). Vesicular release probability calculated as quotient of EPSC and RRP sucrose charge was not significantly changed even though a trend towards lower release probability in M18 neurons was observed (Figure.3C). Alternatively, one can deplete the RRP by a high frequency train of action potentials (1 APs at Hz). We plotted cumulative EPSC charge, fitted the last 2 APs (steady state) with linear regression and back-extrapolated to the time point. 11

A.2nA B RRP size (nc) 1..8.6..2 * C P ves (%) 3 2 1 D 1s E F cumulative total charge (pc) 1 8 6 2.5 1. 1.5 2. 2.5 time (s) RRP estimate (pc) 3 2 1 slope (pc/s) 3 2 1 Figure.3 Cdk5 phosphorylation of Munc18-1 affects sucrose pool. Hippocampal autaptic neurons of munc18-1 null mutant mice were rescued with Lentiviruses expressing M18 or M18. (A) Typical response to a single hyperosmotic sucrose application (5mM, 3.5s). Scale bar represents.2na and 1s. (B) Mean RRP size as assessed with 5mM sucrose application (M18 :.9 ±.9nC, n=21; M18 :.65 ±.8nC, n=21; *, Mann-Whitney test, p=.39). (C) Vesicular release probability P ves, calculated as quotient of EPSC charge by sucrose triggered charge (M18 : 2.1 ±.3%, n=21; M18 : 1.6 ±.%, n=21; ns., Mann-Whitney test, p>.5). (D) Cumulative plot of the total EPSC charge during a Hz train (1APs). Yellow lines indicate back-extrapolation from the steady-state phase (AP 8-1) to y-axis intercept, which can be used as a RRP estimate. (E) RRP estimate as measured with back-extrapolation of cumulative total charge from Hz train (M18 : 138. ± 17.15pC, n=32; M18 : 192.2 ± 33.33pC, n=35; ns., Mann-Whitney test, p>.5). (F) Slope of Hz cumulative charge (M18 : 17.53 ± 28.9pC/s, n=32; M18 : 238.33 ± 52.91pC/s, n=35; ns., Mann-Whitney test, p=.2). All data are expressed as mean ± SEM. The intercept on the Y-axis can be used to estimate the initial RRP APs size (Schneggenburger et al., 1999). RRP APs size was not significantly different between M18 and M18 neurons (Figure.3D-E). Furthermore, RRP refilling, as measured by the slope of the cumulative plot, was similar in both genotypes (Figure.3F). Thus, phosphorylation of Munc18-1 is needed to set RRP size but Ca 2+ -dependent processes can rescue the initial reduction of RRP size in M18 neurons. 12

A PP ratio 1. 1..8 * * * B.5nA 1ms.6 C normalized EPSC amplitude E normalized EPSC amplitude.8.6..2 Hz 1..8.6..2.2.5.1.2.5 1 PP interval (s) 5Hz 1. normalized EPSC amplitude 5 1 15 2 time (s) normalized EPSC amplitude 1..8.6..2 1..8.6..2.5 1. 1.5 2. time (s).5.1.15.2 time (s).5 1. 1.5 2. 2.5 time (s) D normalized EPSC amplitude F synchronous/total charge (%) 1Hz 1..8.6..2 1 8 6 2 normalized EPSC amplitude 1..8.6..2.2..6.8 time (s) 2 6 8 1 time (s).5 1. 1.5 2. 2.5 time (s) Figure. Cdk5 phosphorylation of Munc18-1 sets release probability. Hippocampal autaptic neurons of munc18-1 null mutant mice were rescued with Lentiviruses expressing M18 or M18. (A) Paired pulse ratio (ratio of the second to the first synaptic response plotted as a function of interstimulus interval) (n=33 (M18 ), n=37 (M18 ), Mann-Whitney tests: PP(2ms): *, p=.3; PP(5ms): *, p=.2; PP(1ms): ns., p=.56; PP(2ms): ns., p>.5; PP(5ms): ns., p>.5; PP(1ms): *, p=.11). (B) Typical examples of paired pulse responses of a M18 and M18 neuron. Scale bars represent.5na and 1ms. (C) EPSC amplitudes during a 5Hz train normalized to first EPSC (1 APs). Insert shows zoom of the first 1 APs (n=25 (M18 ), n=3 (M18 ). (D) EPSC amplitudes during a 1Hz train normalized to first EPSC (1 13

APs). Insert shows zoom of the first 1 APs (n=25 (M18 ), n=32 (M18 ). (E) EPSC amplitudes during a Hz train normalized to first EPSC (1 APs). Insert shows zoom of the first 1 APs (n=32 (M18 ), n=35 (M18 ). (F) Percentage of synchronous charge out of the total EPSC charge during a Hz train (1 APs) (n=32 (M18 ), n=35 (M18 ). All data are expressed as mean ± SEM. Cdk5 phosphorylation of Munc18-1 sets release probability. To investigate the role of Cdk5 phosphorylation of Munc18-1 on release probability, we stimulated cells with paired pulses (PP) of varying interstimulus intervals ranging form 2 to 1ms. M18 neurons showed paired-pulse facilitation compared to M18 neurons, arguing for a lower initial synaptic release probability in those neurons (Figure.A-B). In line with the observed paired-pulse facilitation, rundown kinetics during action potential trains at 5Hz were slower in M18 neurons (Figure.C). However, no significant differences were observed in rundown kinetics after higher frequency (1Hz or Hz) stimulation (Figure.D-E). Furthermore, both synchronous and asynchronous release components scaled similarly with the effect on EPSC size (Figure.F). Hence, Cdk5 phosphorylation is a determinant of synaptic release probability and short-term plasticity, but does not affect the ratio of synchronous and asynchronous release components. Cdk5 phosphorylation of Munc18-1 increases synaptic Munc18-1 and Syntaxin-1 protein levels Synaptic strength strongly correlates with Munc18-1 protein levels (Toonen et al., 26). To investigate whether the observed electrophysiological changes in the Cdk5 phosphorylation deficient variant of Munc18-1 could be explained by changes in Munc18-1 protein levels, we immunostained neurons for Munc18-1 together with the dendritic marker MAP2 and the synapse marker VAMP2. No significant changes in synaptic Munc18-1 levels were observed between M18 and M18 neurons (Figure.5A-B). Protein levels of syntaxin are known to strongly depend on the presence of its binding partner Munc18-1 (Verhage et al., 2). In addition, Cdk5 phosphorylation of Munc18-1 decreases the affinity for monomeric Syntaxin in secretory cells (Fletcher et al., 1999; Shuang et al., 1998). To investigate the effect of a lack of Cdk5 phosphorylation of Munc18-1 on the transport of syntaxin, we immunostained neurons for 1

A synaptic Munc18 intensity (a.u.) C synaptic Munc18 intensity (a.u.) D synaptic syx1 intensity (a.u.) F synaptic syx intensity (a.u.) 15 1 5 2 15 1 5 1 8 6 2 1 B cumuulative probability cumulative probability 1 8 6 2 1 2 3 synaptic Munc18 intensity (a.u.) 5 1 15 distance from soma (μm) 8 6 2 E 1 8 6 2 1 2 3 synaptic syx1 intensity (a.u.) 5 1 15 distance from soma (μm) Figure.5 Cdk5 phosphorylation of Munc18-1 influences synaptic Munc18-1 and syntaxin protein levels. Hippocampal neurons of munc18-1 null mutant mice were rescued with Lentiviruses expressing M18 or M18 and were immunostained for MAP2, VAMP2 and Munc18-1 or Syntaxin-1. (A) Mean synaptic Munc18-1 intensity (M18 : 1232 ± 127, n=38; M18 : 118 ± 622, n=37; ns., Mann-Whitney test, p>.5). (B) Cumulative probability plot of synaptic Munc18-1 intensities. (C) Mean synaptic Munc18-1 intensity as a function of radial distance from the soma. (D) Mean synaptic syntaxin intensity (Munc18 : 582 ± 12, n=7; Munc18 : 588 ± 65, n=11; ns., Mann-Whitney test, p>.5). (E) Cumulative probability plot of synaptic syntaxin intensities. (F) Mean synaptic syntaxin intensity as a function of radial distance from the soma. All data are expressed as mean ± SEM. 15

syntaxin and analyzed its amount and localization. In line with Munc18-1 levels, synaptic Syntaxin-1 levels were similar in M18 and M18 neurons (Figure.5D-E). Furthermore, we did not observe any distance dependence of Munc18-1 and Syntaxin-1 localization (Figure.5C,F). Thus, Syntaxin-1 levels highly correlate with Munc18-1 levels and both proteins are trafficked independently of Cdk5 phosphorylation of Munc18-1. Discussion This study shows that in hippocampal neurons Cdk5 phosphorylation of Munc18-1 is important for synaptic transmission. Neurons in which Cdk5 cannot phosphorylate Munc18-1 have normal neuronal morphology, but show a strong reduction in evoked and spontaneous release. Lack of phosphorylation decreases RRP size, release probability and influences short-term plasticity. Levels and localization of Munc18-1or its binding partner Syntaxin-1 are independent of Cdk5 phosphorylation. Cdk5 phosphorylation of Munc18-1 enhances release in neurons and secretory cells Our data in hippocampal neurons are in line with the phenotype observed in secretory cells where Cdk5 promotes vesicle fusion via a Munc18- dependent mechanism (Chergui et al., 2; Fletcher et al., 1999; Lilja et al., 21). In pancreatic beta cells, the non-phosphorylatable variant Munc18 cannot support Cdk5-dependent enhancement of secretion (Lilja et al., 2). In addition, overexpression of Munc18 in bovine adrenal chromaffin cells significantly slows fusion kinetics. In secretory cells, Cdk5 phosphorylation of Munc18 lowers its affinity to monomeric Syntaxin-1 (Fletcher et al., 1999; Shuang et al., 1998). Although not affecting Syntaxin-1 levels and localization in neurons, Cdk5 phosphorylation of Munc18-1 may dissociate Munc18-1 from closed Syntaxin-1, allowing Syntaxin-1 to participate in SNARE complex formation and therewith enhance vesicle fusion. Cdk5 phosphorylation thus changes intrinsic molecular properties of Munc18-1 that enhance vesicle fusion independent of the cell-type used. 16

Munc18-1 as presynaptic target in Cdk5-dependent regulation of synaptic strength Cdk5 is activated upon presynaptic neuronal silencing resulting in differential regulation of synaptic strength in recurrent networks. Whereas some CA3-CA3 neuron pairs loose their functional connectivity upon TTX treatment, other synapses are strengthened (Mitra et al., 211). Loss of connectivity upon chronic activity deprivation might be explained by Cdk5 mediated synaptic silencing and restriction of recycling pool size (Kim and Ryan, 21) or phosphorylation of P/Q-type calcium channels (Tomizawa et al., 22). In the remaining active synapses, the enhanced synaptic strength is due to a marked increase in release probability (Mitra et al., 211). Though in different synapses, both loss of connectivity and increase in synaptic strength are mediated by Cdk5 activation. So far, the exact molecular mechanisms and presynaptic Cdk5 targets involved are missing. Here, we propose a role for Cdk5 phosphorylation of Munc18-1 in the regulation of synaptic strength and exclude Munc18-1 as a major target involved in presynaptic silencing. Neurons expressing nonphosphorylatable Munc18 show smaller RRP sucrose and reduced release probability, leading to decreased spontaneous and evoked release. In line with the effect on release probability, action potential trains at 5Hz have slower rundown kinetics in the absence of Cdk5 phosphorylation. In contrast, rundown kinetics at higher frequency trains (1Hz or Hz) or RRP APs were more similar in the two genotypes. This might be due to the fact that our measurements were performed in autaptic neurons. These neurons are grown in complete isolation and are thus devoid of network activity, arguing for initial high Cdk5 activity. Ca 2+ -dependent activation of phosphatases might lead to dephosphorylation of Munc18-1 upon high frequency stimulation. The balance between Cdk5 and the phosphatase calcineurin has already shown to be important for the regulation of recycling pool size (Kim and Ryan, 21). Likewise, Cdk5-dependent regulation of synaptic strength might also be influenced by the equilibrium between kinase and phosphatase activities. Heterogeneous effect of Cdk5 mediated homeostatic adaptation Cdk5 activation upon chronic activity deprivation triggers loss of connectivity or increased synaptic strength. The differential effects in different synapse might be explained by independent mechanisms depending on expression 17

and relative amounts of Cdk5 targets. We identify Munc18-1 as one important target in Cdk5-mediated increase in synaptic strength, while other presynaptic Cdk5 targets must exist to mediate Cdk5 s effects on synaptic silencing and/or recycling pool size. Presynaptic protein levels, and Munc18-1 is no exception, are highly variable between synapses (SK Schmitz, unpublished observation), as has been observed for release probability (Branco and Staras, 29). The amounts of Munc18-1 relative to one or multiple other Cdk5 targets might therefore determine the combinatorial outcome of Cdk5 activity on functional connectivity. Munc18 s involvement in epilepsy Recently, Munc18-1 has been linked to early-infantile epilepsy syndromes, such as Ohtahara syndrome and West syndrome (Saitsu and Matsumoto, 211). One patient carries a mutation in STXBP1 gene (172A>C) that leads to an amino acid change in the Cdk5 phosphorylation site on Munc18-1 (57T>P). The patient suffered from seizures from the first day of life until the age of 1, displayed tremor, jerks stereotypic movements and ataxic walk. Furthermore, the patient showed autistic features and was unable to speak (Milh et al., 211). Due to the mutation, Cdk5 cannot phosphorylate Munc18 at T57. We identified Munc18-1 as major Cdk5 target that contributes to the regulation of synaptic strength. It is tempting to speculate that dysfunctional homeostatic adaptation due to the lack to Munc18-1 phosphorylation by Cdk5 underlies the disease etiology. Acknowledgements We thank Hans Lodder and Jurjen Broeke for their invaluable technical assistance, Robbert Zalm for cloning of Munc18-1 variants and Lentivirus production and Desiree Schut, Ingrid Saarloos and Boukje Beuger for autaptic cultures. 18