This Accepted Manuscript has not been copyedited and formatted. The final version may differ from this version.

Research Articles: Cellular/Molecular

USP8 Deubiquitinates SHANK3 to Control Synapse Density and SHANK3 Activity-dependent Levels

Meghan Kerrisk Campbell1 and Morgan Sheng1

1Department of Neuroscience, Genentech, South San Francisco, CA 94080

DOI: 10.1523/JNEUROSCI.3305-17.2018

Received: 20 November 2017

Revised: 28 February 2018

Accepted: 29 March 2018

Published: 7 May 2018

Author contributions: M.K.C. wrote the first draft of the paper; M.K.C. and M.S. edited the paper; M.K.C. and M.S. designed research; M.K.C. performed research; M.K.C. analyzed data; M.K.C. wrote the paper.

Conflict of Interest: The authors are full time employees of Genentech, a member of the Roche Group.

We thank Eunjoon Kim for providing DNA constructs, Baris Bingol, and Christopher Wall for providing materials and technical advice on biochemical experiments, Christine Kim for proving technical assistance, and Casper Hoogenraad, Borislav Dejanovic, Daniel Fleck, and Annette Vogl for critical discussions on experimental design and results.

Corresponding authors: Morgan Sheng, Department of Neuroscience, Genentech, 340 Point San Bruno Blvd, South San Francisco, CA 94080, USA Tel.: +1 650 467 3437 E-mail: sheng.morgan@.com; Meghan Kerrisk Campbell, current address Alkahest, 75 Shoreway Rd, San Carlos, CA 94070, USA Tel.: +1 650 801 1311 Email: [email protected]

Cite as: J. Neurosci ; 10.1523/JNEUROSCI.3305-17.2018

Alerts: Sign up at www.jneurosci.org/cgi/alerts to receive customized email alerts when the fully formatted version of this article is published.

Accepted manuscripts are peer-reviewed but have not been through the copyediting, formatting, or proofreading process.

Copyright © 2018 the authors

1 USP8 Deubiquitinates SHANK3 to Control Synapse Density and SHANK3 Activity-dependent

2 Protein Levels

3 Abbreviated Title: USP8 Deubiquitinates SHANK3

4

5 Meghan Kerrisk Campbell1, Morgan Sheng1,*

6 1 Department of Neuroscience, Genentech, South San Francisco, CA 94080

7

8 * Corresponding authors: Morgan Sheng, Department of Neuroscience, Genentech, 340 Point

9 San Bruno Blvd, South San Francisco, CA 94080, USA Tel.: +1 650 467 3437 E-mail:

10 [email protected]; Meghan Kerrisk Campbell, current address Alkahest, 75 Shoreway

11 Rd, San Carlos, CA 94070, USA Tel.: +1 650 801 1311 Email: [email protected]

12

13 Number Pages: 44

14 Number Figures, Tables, Multimedia, & 3D Models: 6 Figures, 0 Tables, 0 Multimedia, 0 3D

15 Models

16 Number Words: Abstract (172), Introduction (636), Discussion (876)

17 Conflict of Interest: The authors are full time employees of Genentech, a member of the Roche

18 Group.

19

20 Acknowledgements: We thank Eunjoon Kim for providing DNA constructs, Baris Bingol, and

21 Christopher Wall for providing materials and technical advice on biochemical experiments,

22 Christine Kim for proving technical assistance, and Casper Hoogenraad, Borislav Dejanovic,

23 Daniel Fleck, and Annette Vogl for critical discussions on experimental design and results.

24 Abstract

25 Mutations or altered protein levels of SHANK3 are implicated in neurodevelopmental disorders

26 such as Phelan-McDermid syndrome, autism spectrum disorders, and schizophrenia (Guilmatre

27 et al., 2014). Loss of SHANK3 in mouse models results in decreased synapse density and

28 reduction in the levels of multiple synaptic (Jiang and Ehlers, 2013). The family of

29 SHANK scaffolding molecules are among the most heavily ubiquitinated proteins at the

30 postsynaptic density. The ubiquitin-dependent proteasome degradation of SHANK is regulated

31 by synaptic activity and may contribute to activity-dependent synaptic remodeling (Ehlers, 2003;

32 Shin et al., 2012). However, the identity of the specific deubiquitinating enzymes and E3 ligases

33 that regulate SHANK ubiquitination at synapses are unknown. Here we identify USP8/UBPY as

34 a deubiquitinating enzyme that regulates SHANK3 and SHANK1 ubiquitination and protein

35 levels. In primary rat neurons, USP8 enhances SHANK3 and SHANK1 protein levels via

36 deubiquitination and increases dendritic spine density. Additionally, USP8 is essential for

37 changes in SHANK3 protein levels following synaptic activity modulation. These data identify

38 USP8 as a key modulator of SHANK3 downstream of synaptic activity.

39

40

2

41 Significance Statement

42 Precise regulation of the protein levels of the postsynaptic scaffolding protein SHANK3 is

43 essential for proper neurodevelopment. Mutations of SHANK3 have been identified in Phelan-

44 McDermid syndrome, autism spectrum disorders, and schizophrenia (Guilmatre et al., 2014). In

45 this research, we identify USP8 as a key enzyme that regulates SHANK3 protein levels in

46 neurons. USP8 acts to deubiquitinate SHANK3, which prevents its proteasomal-mediated

47 degradation and enhances overall dendritic spine stability. In the future, modulation of USP8

48 deubiquitinating activity could potentially be utilized to titrate the protein levels of SHANK3 to

49 ameliorate disease.

50

3

51 Introduction

52 The ubiquitin-proteasome system (UPS) is a central mechanism for protein degradation

53 and turnover. Proteins are tagged for degradation through a series of enzymatic reactions

54 culminating with the covalent attachment of K48 polyubiquitin chains to specific lysine residues

55 of target proteins via E3 ligases. This polyubiquitination acts as a molecular tag to direct proteins

56 to the proteasome for proteolysis (Bhattacharyya et al., 2014). Ubiquitin is also used to regulate

57 numerous non-proteolytic cellular functions via conjugation as mono-ubiquitin or non-K48

58 linkages (Swatek and Komander, 2016). Due to its important roles in fundamental cellular

59 processes, the ubiquitination of proteins is counterbalanced by a set of deubiquitinating enzymes

60 (DUBs) that remove ubiquitin from target proteins and thus inhibit their degradation (Nijman et

61 al., 2005).

62 In neurons, the UPS is an important mechanism utilized to regulate activity-dependent

63 protein turnover and structural changes at excitatory synapses (Ehlers, 2003; Bingol and

64 Schuman, 2005; Shin et al., 2012). The morphology of dendritic spines, the postsynaptic actin-

65 based protrusions that house excitatory synapses, changes bidirectionally in response to synaptic

66 activity, and the size and shape of dendritic spines correlates with their synaptic strength

67 (Matsuzaki et al., 2004; Nägerl et al., 2004; Sheng and Hoogenraad, 2007; Araki et al., 2015).

68 Postsynaptic density (PSD) scaffolding molecules, particularly the SHANK and GKAP/SAPAP

69 family of proteins, are some of the most highly ubiquitinated proteins at synapses and their

70 protein levels are controlled bidirectionally by synaptic activity (Ehlers, 2003; Hung et al., 2010;

71 Shin et al., 2012). Blocking synaptic activity results in a reduction of SHANK ubiquitination and

72 enhanced SHANK protein levels, while increasing excitatory synaptic activity leads to

73 heightened ubiquitination and loss of SHANK protein via proteasomal degradation (Ehlers,

4

74 2003). SHANK proteins are abundant and highly concentrated in the PSD, where they interact

75 with GKAP and PSD-95 family members. Together these scaffolds undergird the structure of the

76 PSD and coordinate intracellular responses to extracellular cues by binding to multiple

77 membrane receptors including NMDA and metabotropic glutamate receptors, as well as,

78 intracellular cytoskeletal modulators and signaling enzymes including cortactin and CAMKII

79 (Naisbitt et al., 1999; Tu et al., 1999; Sheng and Hoogenraad, 2007; MacGillavry et al., 2015).

80 Based on its central position and functional effects, the SHANK family has been hypothesized to

81 play a master regulatory role in PSD organization and protein turnover at synapses (Sala et al.,

82 2001; Shin et al., 2012).

83 The SHANK family of proteins consists of three members, SHANK1, SHANK2, and

84 SHANK3. Notably, multiple mutations in SHANK3 (heterozygous gene deletions, gene

85 duplications, and missense mutations) have been identified in neurodevelopmental disorders,

86 including Phelan-McDermid syndrome, autism spectrum disorders (ASD), and schizophrenia

87 (Guilmatre et al., 2014). Recently, SHANK1 and SHANK2 mutations have also been implicated

88 in neurodevelopmental disorders (Berkel et al., 2010; 2012; Sato et al., 2012; Leblond et al.,

89 2014). The human genetics findings, taken together with numerous SHANK3 knockout and

90 transgenic mouse studies (Peça et al., 2011; Han et al., 2013; Lee et al., 2013; Schmeisser, 2015;

91 Mei et al., 2016; Reim et al., 2017), highlight the importance for regulation of SHANK3 protein

92 levels in vivo. However, the enzymes that regulate SHANK3 protein levels through

93 ubiquitination are poorly understood (Hung et al., 2010).

94 Due to SHANK3’s strong link to human disease and the known regulation of SHANK

95 turnover by UPS, we performed an unbiased screen of DUBs that might regulate the protein level

96 of SHANK3. We identified USP8/UBPY as a regulator of SHANK3 ubiquitination and protein

5

97 levels in heterologous cells and neurons. USP8 controls SHANK3 and SHANK1 protein levels

98 (but not GKAP or PSD-95) via deubiquitination, which in turn controls dendritic spine density in

99 neurons. Furthermore, knockdown of USP8 in neurons prevents bidirectional changes in

100 SHANK3 protein levels induced by altered synaptic activity. Taken together, this data identifies

101 USP8 as a critical enzyme that deubiquitinates PSD scaffold proteins SHANK3 and SHANK1,

102 thereby regulating synapse number and size.

103

104

105

6

106 Materials and Methods

107 Animal Use

108 All rodent experiments were approved by the Genentech Animal Care and Use Committee and

109 followed the National Institutes of Health Guide for the Care and Use of Laboratory Animals.

110

111 DNA Constructs

112 A FLAG-tagged library of 114 human DUB cDNAs was used for the DUB overexpression

113 screen in pRK5 vectors as described previously (Bingol et al., 2014). The USP8 dead vector was

114 generated using QuikChange II XL (Agilent Technologies) to introduce the point mutation

115 C786S. Human SHANK3 cDNA was synthesized at GENEWiz into the pCDNA/FRT/TO

116 (Invitrogen) for stable cell line generation and into pCDNA vector with a TdTomato tag for

117 overexpression and immunofluorescence in cultured neurons. SHANK1 HA-tagged pGW1 was

118 provided by Dr. Eunjoon Kim’s laboratory at the Korea Advanced Institute of Science and

119 Technology. Human SHANK2 Myc-tagged pCMV, human GKAP pCMV, and Myc-tagged rat

120 USP8 cDNAs were purchased (OriGene Technologies). EGAL and PSD-95 cDNA was

121 described previously (Bingol, Nature, 2014). Rat USP8 shRNA constructs were purchased

122 (OriGene Technologies) and targeted the following sequences: USP8_A

123 (ATCTTGACCTGTCACAGTATGTTATTGGC), USP8_B

124 (TTCGGACATCTCCGTGTCTTCTGTGAGGT), USP8_C

125 (ATGAGAACTGGCTCCTGTGCTATCCACAG), USP8_D

126 (TCCGCCTGAGATGGCTCCTTCGTCTGTGC), and non-targeting shRNA (OriGene

127 Technologies, TR30008). For knockdown in HEK293 cells, pools of 4 human siRNAs were

128 purchased (Dharmacon), and targeted the following sequences: USP8

7

129 (TGAAATACGTGACTGTTTA, GGACAGGACAGTATAGATA,

130 AAATAAAGCTCAACGAGAA, GGCAAGCCATTTAAGATTA), SHANK3

131 (GATACAAGCGGCGAGTTTA, GGGCTTCACCTGACTACAA,

132 CGAGATTAGCTCATTGTTT, CGACAAACCAACTGTGATC), non-targeting pool #1 (D-

133 001206-13-05). For knockdown in primary cortical neuron cultures, the following Accell

134 siRNAs were purchased (Dharmacon): non-targeting control pool #1 D-001206, rat SHANK3

135 SMARTpool E-080173, USP3 SMARTpool E-101133, UCHL1 SMARTpool E-089822, USP22

136 SMARTpool E-083656, HDAC6 SMARTpool E-095518, ATXN3 SMARTpool E-096577,

137 JOSD1 SMARTpool E-081141, and USP8 SMARTpool E-092861.

138

139 Antibodies and Reagents

140 SHANK3, Synaptic Systems 162304, guinea pig polyclonal antiserum used for

141 immunofluorescence of cultured neurons 1:1000; SHANK3, Sigma Clone S72-16 SAB5200047

142 monoclonal mouse antibody used for immunoblotting 1:500; SHANK1, Novus NB300-167

143 162013, polyclonal rabbit antibody used for immunoblot and immunofluorescence at 1:1000;

144 SHANK2, NeuroMab UC Davis/NINDS/NIMH Clone N23B/6 75-088, RRID: AB_2254586,

145 monoclonal mouse antibody used for immunoblotting and immunofluorescence at 1:1000; Pan

146 SHANK, NeuroMab UC Davis/NINDS/NIMH Clone N23B/49 75-089, RRID:AB_11213314,

147 monoclonal mouse antibody used for immunoblotting 1:1000; Beta-actin-HRP conjugate, Cell

148 Signaling Clone 13E5 5125, RRID:AB_1903890, monoclonal rabbit antibody used for

149 immunoblotting 1:5000; FLAG, Sigma Clone M2 F1804, RRID:AB_262044, monoclonal mouse

150 antibody used for immunoblotting 1:2000; Myc-tag HRP conjugate, Cell Signaling Clone 9B11,

151 RRID:AB_10707162, mouse monocolonal antibody used for immunoblotting 1:2000; PSD-95,

8

152 NeuroMab UC Davis/NINDS/NIMH Clone K28/43, RRID:AB_2292909, monoclonal mouse

153 antibody used for immunoblotting and immunofluorescence of cultured neurons 1:1000; GKAP,

154 Cell Signaling 13602, polyclonal rabbit antibody used for immunoblotting 1:2000; GKAP,

155 Novus Clone S127-31 NBP1-47612, RRID:AB_10010569, monoclonal mouse antibody used for

156 immunofluorescence of cultured neurons 1:1000; HOMER1A, Cell Signaling 8231, polyclonal

157 rabbit antibody used for immunofluorescence of cultured neurons 1:1000; USP8, Sigma Clone

158 US872 SAB4200527, mouse monoclonal antibody used for immunoblotting 1:100;

159 Ubiquitinated proteins, Millipore Clone FK2 04263, RRID:AB_612093, monoclonal mouse

160 antibody used for immunoblotting 1:1000; HA-Tag, Cell Signaling Clone C29F4,

161 RRID:AB_1549585, rabbit monoclonal antibody used for immunoblotting at 1:2000; MG132,

162 Selleckchem S2619; DMSO, Sigma D8418; TTX, Enzo BML-NA120; BICC, Sigma 14343; PR-

163 619, LifeSensors SI9619; 1,10-phenanthroline, LifeSensors SI9649; N-Ethylmaleimide, Sigma

164 E3876; Papain, Worthington Biological Corps LS003119; Neurobasal Media, Gibco 21103-049;

165 Poly-D-Lysine, Sigma P7280; Mouse Laminin, Invitrogen 23017-015; B27, Gibco 17504-044;

166 Glutamax, Gibco 35050; Pen/Strep, Gibco 15140; Lipofectamine 2000, Invitrogen 11668027;

167 FLAG-magnetic beads, Sigma M8823; Protein A/G Agarose Beads, Pierce 20423; TUBE

168 Magnetic beads, LifeSensors, UM402M; Recombinant USP8, BostonBiochem E-520; TRIzol

169 Reagent, ThermoFischer 15596026; iScript cDNA Synthesis Kit, Biorad 1708890; SHANK3 and

170 HPRT1 TaqMan Probes, Applied Biosystems.

171

172 Generation of Stable Cell Line

173 Stable SHANK3 doxycycline-inducible cell lines were generated using Flp-In T-Rex Core Kit

174 (Invitrogen K6500-01). Briefly, pOG44 Flp-recombinase pCDNA and SHANK3 human

9

175 pCDNA/FRT/TO were transfected into HEK293 Flp-In T-REx cells at a 9:1 ratio. 48HRS post-

176 transfection, cell lines were transferred to selection media containing 50Pg/mL hygromycin and

177 15Pg/mL blasticidin. Media was exchanged every 3-4 days for approximately two weeks before

178 the development cell foci. These foci were picked into independent cultures, expanded, and

179 tested for dox-inducible expression of SHANK3. Positive clones were picked and maintained in

180 selection media. A single stable clone was used for all experiments presented in this manuscript.

181

182 Deubiquitinating Enzyme Overexpression Screen

183 Stable SHANK3 HEK293 cells were plated in 24 wells with doxycycline and each well was

184 transfected with a separate DUB from the library. EGAL was transfected as a baseline control

185 and EGAL transfected cells treated with 1 PM MG132 for 24HRS were used as a positive

186 control. Cell lysates were collected 76HRS post transfection and resolved via SDS-PAGE, then

187 analyzed by immunoblotting for SHANK3, FLAG-DUB, and ACTIN. Band intensity was

188 measured using ImageLab software (BioRad) and SHANK3 intensity was normalized to ACTIN

189 as a loading control. The screen was run twice with 2 technical replicates each time and data was

190 pooled resulting in 11 SHANK3 DUB hits that significantly increased SHANK3 expression over

191 control. A subsequent, more stringent screen was run to further narrow down the list of candidate

192 DUBs. The 11 significant DUBs were transfected into stable SHANK3 cells for 24HRS and then

193 assayed as described above for SHANK3 protein expression. The screen was run 3 times with 2

194 technical replicates each time and data was pooled resulting in 7 statistically significant

195 SHANK3 candidate DUBs. To determine if MG132 and USP8 overexpression had an additive

196 effect, we transfected USP8 or EGAL control in stable SHANK3 HEK293 cells for 24HRS and

10

197 then treated with 1 PM MG132 or vehicle for an additional 24HRS. SHANK3 protein expression

198 levels were then assayed as described above.

199

200 RNA Extraction, RT-PCR, and TaqMan Real-Time PCR Assay

201 Stable SHANK3 HEK293 cells were plated in 6 well plates with doxycycline and each well was

202 transfected with either EGAL control or USP8 for 48HRS. Total RNA was extracted with TRIzol

203 reagent following the manufacturer’s instructions. 2 Pg of RNA was used for cDNA synthesis

204 using iScript kit. Then a TaqMan gene expression assay was run for each cDNA sample in

205 triplicate with a standard curve and SHANK3 mRNA levels were normalized to the

206 housekeeping gene HPRT1.

207

208 Primary Neuronal Culture

209 Embryos from timed pregnant Sprague-Dawley rats were removed at E18 and decapitated into

210 cold HBSS/HEPES buffer. The hippocampus or cortex and hippocampus (as indicated in figure

211 legends) were dissected and placed into 5mL of HBSS/HEPES buffer. Tissue was washed 4

212 times before digestion at 37qC for 20 minutes with papain (0.5mL at 200U/mL into 4.5mL

213 HBSS/HEPES buffer with tissue). Tissue was washed 4 times with warm HBSS/HEPES buffer

214 and once with Neurobasal media. Dissociated tissue was triturated and passed through a 70um

215 filter before plating on poly-D-lysine/laminin coated plates or coverglass with Neurobasal media

216 supplemented with B27, Glutamax, and Pen/Strep. Cells were plated at a density of 120,000

217 cells/well in 24 well plates with coverglass for imagining or 250,000 cells/well in 24 well plates

218 and 8 million cells/6cm plates for biochemical experiments. Media was exchanged 100% 3-

219 5HRS after initial plating and then exchanged 50% once per week.

11

220

221 Knockdown and Analysis of DUBs in Primary Neuronal Cultures

222 Accell siRNAs from Dharmacon were added to primary hippocampal neuronal cultures at DIV10

223 at a final concentration of 1 PM and refreshed at DIV14. Cultures were lysed at DIV18 and

224 resolved on 4-8% Tris-Acetate gels. Gels were analyzed by quantitative immunoblotting using

225 ImageLab software (BioRad) and SHANK3 intensity was normalized to ACTIN as a loading

226 control. The quantifications are from 4 independent cultures with 2-4 replicates from each

227 culture.

228

229 Transfection, Immunofluorescence, and Quantification of Primary Neuronal Cultures

230 Primary neuronal cultures were transfected between DIV12-14 with Lipofectamine 2000.

231 Neurons were transfected by incubating cells with 2PL Lipofectamine, 400-1000ng DNA, and

232 500PL Neurobasal for 45 minutes. In some experiments, neurons were treated with the following

233 drugs prior to fixation: DMSO (3 or 24HRS, vehicle control used at same volume as drug

234 treatment), MG132 (3HRS 1 PM), TTX (24HRS, 1 PM), BICC (24HRS, 40 PM). Cells were

235 fixed 1-3 days post-transfection, as indicated in figure legends, using 4% paraformaldehyde in

236 phosphate-buffered saline for 12 minutes at room temperature or 4% paraformaldehyde in

237 phosphate-buffered saline for 2 minutes at room temperature followed 10 minutes at -20qC with

238 100% methanol. Cells were permeabilized and blocked using GDB buffer (0.2% gelatin, 0.5%

239 Triton-X, 0.8M NaCl in phosphate-buffered saline) and incubated with primary antibodies

240 overnight at 4qC. Primary antibodies were visualized using Alexa dye-conjugated secondary

241 antibodies. For experiments using fluorescent tags (GFP, TdTomato) antibodies were not used to

242 visualize these tags. Images were acquired by a single experimenter blinded to transfection or

12

243 treatment using a Zeiss LSM780 laser scanning confocal microscope with a 100X oil objective

244 (0.5 Pm confocal z-step size). Images were quantified by a single experimenter blinded to

245 transfection or treatment using ImageJ software. Image z-stacks were collapsed into maximum

246 projections and thresholded prior to analysis. In Fig 1A, the fluorescence intensity of the entire

247 field was quantified (81.92Pm x 81.92Pm). For all other figures, a segment of secondary dendrite

248 (20Pm x 5Pm) was selected for analysis. For Fig 1E, F, and Fig 2A, B, E, G, H the entire

249 fluorescence intensity was measured in the SHANK3 channel and normalized to the entire

250 fluorescence intensity in the GFP channel. Spine density was calculated by counting the number

251 of dendrite protrusions less than 2Pm in length using the GFP channel. For Fig 3F, G, Fig 4A,

252 and Fig 6A, B, images were processed using the “Analyze Particles” macro in ImageJ to obtain a

253 fluorescence intensity per puncta, size of individual puncta, and puncta number per 20 Pm. The

254 cumulative frequency of puncta intensity and size was calculated using Prism6 software for Fig

255 3G.

256

257 Tandem Ubiquitin Binding Domain Pulldown

258 Ubiquitination of proteins was determined using a magnetic Tandem Ubiquitin Binding Domain

259 (TUBE) pulldown assay. TUBEs recognize tetra-ubiquitin in the nanomolar range and have been

260 demonstrated to bind to all 7 linkage types. For pulldowns, cells were treated with 1PM MG132

261 for 3-5HRS prior to lysis. Cells were lysed (50mM Tris pH 7.5, 150mM NaCl, 1mM EDTA, 1%

262 NP-40, 10% glycerol) with protease, phosphatase, and DUB inhibitors and lysates were clarified

263 by centrifugation for 10 minutes at 14,000g at 4qC. Input samples were removed prior to adding

264 40PL of magnetic-TUBE slurry to 1mL lysate. Inputs and pulldowns were rotated overnight at

265 4qC. Beads were then washed 3 times with TBST (20mM Tris pH 8.0, 150mM NaCl, 0.1%

13

266 Tween-20) and samples were resolved with SDS-PAGE where input lanes contain 1-5% of

267 protein lysate and analyzed by immunoblotting.

268

269 In Vitro Direct Deubiquitination Assay

270 HA-tagged ubiquitin was contransfected along with SHANK1, SHANK2, or SHANK3 in 6 well

271 plates of HEK293 cells using Lipofectamine 2000 for 48HRS. Cell were treated with 1 PM

272 MG132 for 3-5HRS prior to lysis. Cells were lysed in RIPA buffer with protease, phosphatase,

273 and DUB inhibitors and then heated to 85qC for 10 minutes to kill any remaining DUB activity

274 and disrupt any protein-protein interactions. Lysates were then cooled to 4qC and normalized to

275 total protein concentration. Lysates were precleared with protein A/G beads for 20 minutes

276 rotating at 4qC. Beads were removed and lysates were incubated overnight at 4qC with 2Pg of

277 PAN SHANK antibody. The following day, immune complexes were precipitated with protein

278 A/G beads for 2HRS at 4qC followed by washing with RIPA buffer 3 times and then

279 reconstituting in 100PL DUB buffer (10mM Tris-HCl, pH 8, 1mM dithiothereitol, 1mM EDTA).

280 Next, immunoprecipiated lysates were incubated with vehicle, 5 PM purified recombinant USP8,

281 or 5 PM purified recombinant USP8 plus 40 mM N-Ethylmaleimide DUB inhibitor rotating for

282 37qC for 90 minutes. Beads were again washed three times and then incubated in sample loading

283 buffer for 10 minutes at 100qC before loading on SDS-PAGE and western blotting for SHANK

284 and HA-ubiquitin.

285

286 Experimental Design and Statistical Analysis

287 All statistical analyses were performed using GraphPad Prism 6 software. p < 0.05 was regarded

288 as statistically significant and specific statistical tests along with p values are reported within

14

289 each figure legend. All data are displayed as mean with SEM and significance is represented

290 with asterisks (*) or hashtags (#) within the figures. One-way ANOVA with post-hoc Student’s

291 t-tests were used to analyze the quantification of the DUB screen in Fig 1D-F, I and Fig 2A, B.

292 All imaging data were collected and quantified by a single experimenter blinded to condition or

293 treatment and data were analyzed from independent 3-5 primary cultures with ~10 neurons

294 images per culture (n=neuron). One-way ANOVA with post-hoc Student’s t-tests were used to

295 analyze the quantification of USP8 overexpression or knockdown in Fig 1I, Fig 2C, E, G, H, Fig

296 3A, C, F, Fig 4A, and Fig 6A, B. Cumulative frequency was analyzed with Kolmogorov-

297 Smirnov tests in Fig 3G. A repeated measures ANOVA with post-hoc Student’s t-tests were used

298 to analyze the TUBE pulldown assays in Fig 5B, D.

299

300

15

301 Results

302 Identification of USP8 as a putative SHANK3 DUB using an unbiased overexpression

303 screen

304 To confirm that SHANK3 is regulated by the ubiquitin-proteasome in neurons, we treated

305 cultured dissociated rat cortical neurons at day in vitro (DIV) 21 for 3HRS with the proteasome

306 inhibitor MG132. Compared with vehicle control, MG132 increased the level of endogenous

307 SHANK3 protein by approximately 25%, as measured by immunofluorescence intensity of

308 SHANK3 puncta (Fig1A).

309 By deubiquitinating their substrates, DUBs should protect substrates from proteasome

310 degradation and lead to higher steady state levels of those substrates. To identify specific DUBs

311 that might regulate the ubiquitination of SHANK3, we performed an unbiased screen of DUBs

312 for their ability to boost SHANK3 protein levels in a stable HEK293 cell line engineered to

313 overexpress SHANK3. SHANK3 protein expression increased over time following MG132

314 treatment, confirming it can be degraded by the proteasome in this cell line (Fig1B). From a

315 FLAG-tagged human DUB cDNA library containing all 114 known human DUBs, we found that

316 seven DUBs (USP3, UCHL1, USP22, HDAC6, ATXN3, JOSD1, USP8) consistently and

317 significantly increased SHANK3 protein expression in this stable cell line, compared to EGAL

318 control (Fig1C, D). The degree of elevation of SHANK3 induced by coexpression of these seven

319 DUBs was comparable to MG132 treatment (Fig1C, D).

320 To determine if these seven DUBs regulate SHANK3 protein expression in neurons, we

321 transfected USP3, UCHL1, USP22, HDAC6, ATXN3, JOSD1 or USP8 individually in DIV16

322 rat dissociated hippocampal neurons along with SHANK3-TdTomato and GFP. As expected,

323 SHANK3-TdTomato localized in a punctate synaptic pattern along dendrites (Fig 1G). Only

16

324 cotransfection of USP8 resulted in a significant increase in SHANK3-TdTomato intensity,

325 whereas the other six DUBs tested had no significant effect (Fig1E). Enhancing SHANK3

326 protein levels is known to increase dendritic spine number and promote synapse stability

327 (Roussignol et al., 2005; Durand et al., 2012). Indeed, we found that overexpression of USP8

328 also led to an approximately 60% increase in dendritic spine density in transfected neurons

329 compared to EGAL control, perhaps due to the increase in SHANK3 protein expression (Fig1F).

330 To determine if these seven DUBs can act on endogenous SHANK3, we overexpressed each

331 DUB individually in DIV16 hippocampal neurons and immunostained for SHANK3. We found

332 that only overexpression of USP8 resulted in a significant increase in endogenous SHANK3

333 protein expression (Fig 2A), as well as a 45% increase in dendritic spine density (Fig 2B). These

334 results are consistent with a role for USP8 in deubiquitinating SHANK3 and preventing its

335 degradation via the proteasome.

336 To test whether the effects of USP8 overexpression and proteasome inhibition might

337 occlude each other, we combined USP8 overexpression with proteasome inhibitor treatment

338 (USP8+MG132) in SHANK3 stable HEK293 cells. This combination did not elevate SHANK3

339 protein levels more than either USP8 or MG132 alone (Fig 1H, I). The lack of additivity of the

340 effects of USP8 and MG132 on SHANK3 protein levels is consistent with the idea that USP8

341 overexpression is acting via inhibition of UPS degradation of SHANK3. USP8 overexpression

342 was associated with a slight but non-significant decrease in SHANK3 mRNA (Fig 1J), excluding

343 the possibility that the elevation of SHANK3 protein levels upon USP8 overexpression is due to

344 increased transcription of SHANK3.

345

346

17

347 USP8 regulates SHANK3 and SHANK1 protein expression and dendritic spine density in

348 hippocampal neurons

349 To determine if USP8 DUB activity is important for the increase in SHANK3 protein

350 levels, we made a USP8 “dead” enzyme by mutating the catalytic cysteine residue to a serine

351 (USP8-C786S). This mutation has previously been shown to kill the enzyme activity while

352 preserving the overall structure and folding of USP8 (Wu et al., 2004; Mizuno et al., 2005;

353 Avvakumov et al., 2006). In contrast to wildtype USP8, overexpression of USP8-C786S did not

354 change SHANK3 levels in the SHANK3 stable HEK293 cell line (Fig2C, D) and did not

355 significantly change SHANK3-TdTomato intensity or dendritic spine density in hippocampal

356 neurons (Fig2E, F). Thus DUB activity is necessary for USP8’s positive effects on SHANK3

357 protein levels and dendritic spine density. To investigate if USP8 DUB activity is important for

358 regulating endogenous SHANK3, we transfected USP8 or USP8-C786S in DIV15 hippocampal

359 neurons and quantified endogenous SHANK3 intensity using immunofluorescence 24HRS later.

360 Compared with EGAL control, overexpression of USP8 increased the endogenous protein levels

361 of SHANK3 by ~2-fold as measured by immunofluorescence intensity of SHANK3 and led to an

362 approximately 45% increase in spine density. These changes were not observed with

363 overexpression of the catalytically dead mutant USP8-C786S (Fig 2G, H, I), further supporting

364 the hypothesis that USP8 deubiquitinates endogenous SHANK3 and prevents its proteasome

365 degradation.

366 We next examined the effect of loss-of-function of individual DUBs in primary

367 hippocampal neurons. Loss of DUB function should result in a reduction of substrate levels due

368 to enhanced ubiquitination and targeting of the substrate to the proteasome and subsequent

369 degradation. We used Accell siRNAs to knockdown each DUB for 8 days in mature primary

18

370 hippocampal cultures and quantified total SHANK3 levels by immunoblotting. Among the seven

371 DUBs tested, only siRNA knockdown of the DUB USP8, or knockdown of SHANK3 itself,

372 resulted in a significant loss of SHANK3 protein levels (Fig 3A, B). These findings further

373 support the hypothesis that USP8 plays an important role in deubiquitinating SHANK3.

374 Additionally, in the stable HEK293 SHANK3-expressing cells, 60% knockdown of the

375 endogenous human USP8 with siRNA resulted in an approximately 50% reduction in SHANK3

376 protein levels (Fig3C).

377 To further investigate the impact of loss of USP8 on endogenous SHANK3 in rat

378 hippocampal neurons, we used shRNAs targeting rat USP8. Two of four shRNA-expressing

379 constructs tested (shRNA_A and shRNA_B) were effective in knocking down USP8 protein

380 expression when transiently cotransfected with rat myc-tagged USP8 cDNA in heterologous

381 HEK293 cells (Fig 3D). Relative to a scrambled shRNA control, shRNA_A and shRNA_B

382 resulted in 90% and 65% loss of USP8 protein after 72HRS, respectively (Fig3D). We tested

383 knockdown of USP8 for three days in DIV17 hippocampal neurons using the shRNA_A,

384 shRNA_B, or scrambled shRNA control. shRNA-transfected neurons were identified by GFP

385 that was expressed from the same vector. We used ImageJ software to identify SHANK3 puncta

386 along a segment of transfected dendrite and quantified SHANK3 intensity per puncta.

387 Knockdown of USP8 resulted in ~50% reduction in density of SHANK3 puncta and ~30%

388 decrease in SHANK3 immunofluorescence intensity and size of the puncta that remain (Fig3E,

389 F). The decrease in size and intensity of SHANK3 puncta is also indicated by a significant

390 leftward shift of the cumulative frequency graphs of individual puncta size and density (Fig3G).

391 Beyond SHANK3, we sought to determine if USP8 knockdown affected other PSD

392 proteins using immunofluorescence to quantify the endogenous levels of SHANK1, SHANK2,

19

393 PSD-95, GKAP, and HOMER1A. As predicted from the reduction of spine density (and of

394 SHANK3 puncta density), the density of puncta of each of these PSD proteins along dendrites

395 was reduced (though this did not reach significance with HOMER1A), presumably correlating

396 with the loss of synapses in USP8 knockdown neurons (Fig4A, B). However, when quantifying

397 the intensity and size of individual immunofluorescence puncta, only SHANK1 puncta intensity

398 and puncta size were impacted by the knockdown of USP8 (Fig4A, B). The size and

399 fluorescence intensity of SHANK2, PSD-95, GKAP, and HOMER1A puncta were unchanged

400 with USP8 knockdown. Taken together, these results suggest that USP8 controls the protein

401 levels of SHANK1, as well as SHANK3, at excitatory synapses. Depletion of SHANK1 and

402 SHANK3 proteins by USP8 knockdown could account for an overall loss of synapse density.

403

404 USP8 is a DUB for SHANK3 and SHANK1

405 To determine more directly if USP8 can act as a DUB on SHANK, we utilized a Tandem

406 Ubiquitin Binding Entities (TUBE) pulldown assay to quantify ubiquitination of proteins.

407 TUBEs have a nanomolar affinity for polyubiquitinated proteins and can be conjugated to

408 magnetic beads to pulldown polyubiquitinated species from total cell lysates. In SHANK3-

409 expressing stable cell lines transfected with USP8, steady-state SHANK3 protein levels were

410 increased in input lysates, as compared to the same cell line transfected with EGAL control or

411 the USP8 catalytically dead mutant USP8-C786S (Fig5A; also see Fig2C, D). However, the level

412 of polyubiquitinated SHANK3 in USP8 overexpressing cells was significantly reduced, as

413 measured by TUBE pulldown from the same lysates (Fig5A, B). These results are consistent

414 with a role for USP8 in deubiquitinating SHANK3 to enhance protein stability. The total levels

415 of USP8 protein were similar for USP8 WT and USP8-C786S, as observed in the input lanes

20

416 (Fig5A); however, we noted that a larger fraction of USP8-C786S is ubiquitinated than for WT

417 USP8 (Fig5A), perhaps explicable by the self-deubiquitination activity of WT USP8 (Mizuno et

418 al., 2005).

419 Since knockdown of USP8 also decreased SHANK1 expression in neurons (Fig4A, B),

420 we tested if USP8 could deubiquitinate SHANK1 in the same TUBE assay. Similar to SHANK3,

421 SHANK1 protein expression increased with coexpression of USP8 (Fig5C, input lanes). As

422 measured by TUBE pulldown assay, SHANK1 was less polyubiquitinated with coexpression of

423 USP8, as compared to EGAL control or USP8-C786S (Fig5C, D, “TUBE” lanes), suggesting

424 USP8 can deubiquitinate SHANK1 as well as SHANK3. In similar experiments, WT USP8

425 coexpression had no effect on steady state levels or polyubiquitination of SHANK2 (Fig5C, D),

426 which is consistent with our finding in neurons that knockdown of USP8 reduced the synaptic

427 levels of SHANK1 and SHANK3, but not SHANK2 (Fig3E, F, Fig4A, B). By TUBE pulldown,

428 we found that PSD-95 was weakly ubiquitinated, consistent with previous studies (Colledge et

429 al., 2003; Ehlers, 2003; Na et al., 2012) and the small amount that was ubiquitinated was not

430 influenced by USP8 coexpression (Fig5C, D). Finally, we probed if USP8 could deubiquitinate

431 GKAP, an important SHANK binding protein whose ubiquitination is known to play an

432 important role at synapses (Hung et al., 2010; Shin et al., 2012). We found that the levels of

433 GKAP were unchanged between USP8, USP8-C786S, and EGAL control in both input lanes and

434 TUBE pulldowns (Fig5C, D). Thus USP8 shows selective DUB activity towards SHANK1 and

435 SHANK3, among the PSD proteins tested.

436 To determine if USP8 can directly deubiquitinate SHANK3 and SHANK1, we performed

437 an in vitro deubiquitination assay using purified recombinant USP8 enzyme and immunopurified

438 ubiquitinated SHANK proteins. We generated ubiquitinated SHANK proteins by overexpressing

21

439 SHANK1, SHANK2, or SHANK3 in HEK293 cells along with HA-ubiquitin. Cells were treated

440 with MG132 to enhance substrate ubiquitination and lysates were heated to 85qC to kill any

441 enzymatic activity and disrupt protein-protein interactions. SHANK1, SHANK2, or SHANK3

442 were immunoprecipitated and then incubated with recombinant USP8. As measured by HA-

443 ubiquitin signal, we found that SHANK3 and SHANK1 were deubiquitinated in the presence of

444 recombinant USP8 in vitro, but SHANK2 was unaffected (Fig 5E). Furthermore, the

445 deubiquitination of SHANK3 and 1 by USP8 was blocked when DUB inhibitor NEM was

446 included in the assay. These biochemical data indicate that USP8 can act directly as a DUB for

447 SHANK proteins, with apparent specificity for SHANK3 and SHANK1.

448

449 Loss of USP8 blocks the regulation of SHANK3 by synaptic activity

450 Prolonged inhibition of synaptic activity with tetrodotoxin (TTX) increases the protein

451 levels of SHANK, while stimulation of synaptic activity with bicuculline (BICC) leads to

452 reduction in SHANK (Ehlers, 2003; Hung et al., 2010; Tao-Cheng et al., 2010; Shin et al., 2012).

453 To test if deubiquitination of SHANK3 via USP8 is important for these changes, we knocked

454 down USP8 in mature hippocampal neuron cultures and then treated them with TTX, BICC, or

455 DMSO vehicle control for 24HRS. In control neurons transfected with a scrambled shRNA

456 construct, we found that TTX treatment led to an approximately 50% increase in SHANK3

457 puncta immunofluorescence intensity and 40% increase in SHANK3 puncta size, whereas BICC

458 caused an approximately 20-25% reduction in SHANK3 puncta immunofluorescence intensity

459 and puncta size (Fig6A, C). Knockdown of USP8 with shRNA_A or shRNA_B reduced the

460 baseline intensity and size of SHANK3 puncta (Fig6A, C), as seen before (Fig 3E, F).

461 Furthermore, knockdown of USP8 prevented the upregulation of SHANK3 puncta size and

22

462 puncta intensity induced by TTX, and occluded the downregulation of SHANK3 puncta size and

463 puncta intensity induced by BICC (Fig6A, C). Unlike SHANK3, the postsynaptic levels of PSD-

464 95, measured by puncta size and immunofluorescence intensity, were unaffected by shRNA_A

465 or shRNA_B in basal conditions; nor was the up- and down-regulation of PSD-95 in response to

466 TTX and BICC impacted by USP8 knockdown (Fig6B, C). Thus, USP8 plays a specific role in

467 modulation of SHANK3 ubiquitination and protein levels in response to synaptic activity.

468

469

23

470 Discussion

471 SHANK3 is a member of the SHANK family of postsynaptic scaffolding proteins that

472 coordinates the recruitment of receptors and cytoskeletal regulators to influence structure and

473 signaling at the excitatory synapse (Sheng and Kim, 2000). In humans, haploinsufficiency of

474 SHANK3 due to small deletions or balanced translocations have been identified in patients with

475 neurodevelopmental and intellectual disability (Wong et al., 1997; Anderlid et al., 2002;

476 Bonaglia et al., 2006) and loss of SHANK3 is described as the genetic cause of Phelan-

477 McDermid syndrome, which is characterized by developmental delay and autistic behaviors

478 among other symptoms (Wilson et al., 2003). Additionally, mutations in the SHANK3 gene are

479 estimated to contribute to approximately 1% of ASD (Durand et al., 2007; Gauthier et al., 2009;

480 Boccuto et al., 2013; Leblond et al., 2014) and may also play a role in schizophrenia (Failla et

481 al., 2007; Choi et al., 2015; de Sena Cortabitarte et al., 2017). In mice, multiple groups have

482 found that deletion or mutation of Shank3 causes significant changes to the composition of

483 synaptic proteins, alters synapse number and morphology, and impairs animal behavior (Bozdagi

484 et al., 2010; Peça et al., 2011; Wang et al., 2011; Yang et al., 2012; Mei et al., 2016).

485 Furthermore, overexpression of Shank3 causes manic-like behaviors and abnormal excitatory-

486 inhibitory balance in mice (Roussignol et al., 2005; Han et al., 2013). These data highlight the

487 importance of precisely controlled levels of SHANK3 protein for proper synapse development

488 and brain function.

489 In neurons, we and others have shown that SHANK3 and its family members are highly

490 ubiquitinated and this ubiquitination is an important mechanism to control activity-dependent

491 protein turnover and morphological changes at the synapse (Fig1, Fig5, Fig6) (Ehlers, 2003;

492 Bingol and Schuman, 2006; Shin et al., 2012). Identifying the DUB that deubiquitinates

24

493 SHANK3 is an important step, as manipulation of this enzyme could enhance protein stability of

494 SHANK3, potentially increasing steady state SHANK3 levels and perhaps ameliorating the

495 symptoms in patients with haploinsufficiency of SHANK3 or ASD (Mei et al., 2016;

496 Selimbeyoglu et al., 2017; Siegel et al., 2017). From screening a library >100 DUBs, we

497 identified USP8 as a DUB that can remove ubiquitin from SHANK3 and enhance SHANK3

498 protein expression in cultured neurons. Previous publications have found that loss of SHANK3

499 in mice or cultured neurons leads to an overall loss of synapses and reductions in the levels of

500 multiple PSD proteins (Bozdagi et al., 2010; Peça et al., 2011; Wang et al., 2011; Yang et al.,

501 2012; Reim et al., 2017). Here we show that loss of USP8 leads to decreased dendritic spine

502 density, while overexpression of USP8 causes an increase in spine density. We hypothesize that

503 by regulating the ubiquitination and proteasomal degradation of SHANK3 and its family member

504 SHANK1, USP8 controls SHANK1/3 stability and dendritic spine density.

505 In non-neuronal cells, USP8 is reported to act at multiple stages in the ESCRT pathway.

506 USP8 promotes the recycling of cell surface receptors, such as EGFR, back to the plasma

507 membrane or enhances their degradation depending on when and where it deubiquitinates its

508 substrate along the recycling pathway (Mizuno et al., 2005; Niendorf et al., 2007; Wright et al.,

509 2011). Furthermore, USP8 interacts with the proline-rich SH3 domain of the ESCRT0 protein

510 STAM2 via two non-canonical SH3-binding domains (Kato et al., 2000; Berry et al., 2002).

511 SHANK3 also contains an SH3-domain, which may act as potential site of interaction with USP8

512 (Naisbitt et al., 1999; Tu et al., 1999). In fact, USP8 has previously been identified as a putative

513 SHANK3 binder via immunoprecipitation from mouse brain overexpressing Shank3-GFP (Han

514 et al., 2013) and is enriched in dendritic spines of primary cultured neurons (Scudder et al.,

515 2014).

25

516 UPS driven protein degradation contributes to PSD remodeling in response to synaptic

517 activity (Bingol and Sheng, 2011). Therefore, we wanted to determine if modulating the

518 deubiquitination status of SHANK3 via USP8 changes its response to synaptic activity. We

519 found that loss of USP8 leads to decreased SHANK3 protein levels and occludes the

520 bidirectional changes in SHANK3 in response to synaptic activity. However, loss of USP8 does

521 not change the response of PSD-95 to synaptic activity, suggesting the specificity of USP8 for

522 SHANK3. Previous publications have found that overexpression of USP8 increases synaptic

523 strength and reduces AMPAR ubiquitination, while loss of USP8 decreases synaptic strength and

524 increases AMPAR ubiquitination (Scudder et al., 2014). This study additionally reported that

525 overexpression of USP8 prevents the loss of surface AMPAR following heightened synaptic

526 activity and contributes to the downscaling of synaptic strength (Scudder et al., 2014).

527 Our results suggest a model where USP8 deubiquitinates SHANK3 following chronic

528 blockage of synaptic activity, preventing degradation via the proteasome, and increasing synaptic

529 SHANK3 protein levels. In contrast, following chronic synaptic activity, SHANK3 is no longer

530 deubiquitinated by USP8 and subsequently degraded. Previous publications have found that total

531 USP8 protein levels are down regulated following chronic synaptic activity, suggesting a

532 mechanism for decreased deubiquitination of USP8 substrates, such as SHANK3 (Scudder et al.,

533 2014). Together with our results, these studies highlight the critical role for USP8 in controlling

534 the levels of synaptic proteins, such as SHANK3, SHANK1, and AMPAR, in response to

535 synaptic activity modulation. Future work should determine functionally how USP8 is modulated

536 in response to synaptic activity and how it interacts with its substrates.

537

538

26

539 References

540 Anderlid B-M, Schoumans J, Annerén G, Tapia-Paez I, Dumanski J, Blennow E, Nordenskjöld

541 M (2002) FISH-mapping of a 100-kb terminal 22q13 deletion. Hum Genet 110:439–443.

542 Araki Y, Zeng M, Zhang M, Huganir RL (2015) Rapid dispersion of SynGAP from synaptic

543 spines triggers AMPA receptor insertion and spine enlargement during LTP. Neuron

544 85:173–189.

545 Avvakumov GV, Walker JR, Xue S, Finerty PJ, Mackenzie F, Newman EM, Dhe-Paganon S

546 (2006) Amino-terminal dimerization, NRDP1-rhodanese interaction, and inhibited catalytic

547 domain conformation of the ubiquitin-specific protease 8 (USP8). J Biol Chem 281:38061–

548 38070.

549 Berkel S, Marshall CR, Weiss B, Howe J, Roeth R, Moog U, Endris V, Roberts W, Szatmari P,

550 Pinto D, Bonin M, Riess A, Engels H, Sprengel R, Scherer SW, Rappold GA (2010)

551 Mutations in the SHANK2 synaptic scaffolding gene in autism spectrum disorder and mental

552 retardation. Nat Genet 42:489–491.

553 Berkel S, Tang W, Treviño M, Vogt M, Obenhaus HA, Gass P, Scherer SW, Sprengel R, Schratt

554 G, Rappold GA (2012) Inherited and de novo SHANK2 variants associated with autism

555 spectrum disorder impair neuronal morphogenesis and physiology. Hum Mol Genet 21:344–

556 357.

557 Berry DM, Nash P, Liu SK-W, Pawson T, McGlade CJ (2002) A high-affinity Arg-X-X-Lys

558 SH3 binding motif confers specificity for the interaction between Gads and SLP-76 in T cell

559 signaling. Curr Biol 12:1336–1341.

27

560 Bhattacharyya S, Yu H, Mim C, Matouschek A (2014) Regulated protein turnover: snapshots of

561 the proteasome in action. Nat Rev Mol Cell Biol 15:122–133.

562 Bingol B, Schuman EM (2005) Synaptic protein degradation by the ubiquitin proteasome

563 system. Curr Opin Neurobiol 15:536–541.

564 Bingol B, Schuman EM (2006) Activity-dependent dynamics and sequestration of proteasomes

565 in dendritic spines. Nature 441:1144–1148.

566 Bingol B, Sheng M (2011) Deconstruction for reconstruction: the role of proteolysis in neural

567 plasticity and disease. Neuron 69:22–32.

568 Bingol B, Tea JS, Phu L, Reichelt M, Bakalarski CE, Song Q, Foreman O, Kirkpatrick DS,

569 Sheng M (2014) The mitochondrial deubiquitinase USP30 opposes parkin-mediated

570 mitophagy. Nature 510:370–375.

571 Boccuto L, Lauri M, Sarasua SM, Skinner CD, Buccella D, Dwivedi A, Orteschi D, Collins JS,

572 Zollino M, Visconti P, Dupont B, Tiziano D, Schroer RJ, Neri G, Stevenson RE, Gurrieri F,

573 Schwartz CE (2013) Prevalence of SHANK3 variants in patients with different subtypes of

574 autism spectrum disorders. Eur J Hum Genet 21:310–316.

575 Bonaglia MC, Giorda R, Mani E, Aceti G, Anderlid B-M, Baroncini A, Pramparo T, Zuffardi O

576 (2006) Identification of a recurrent breakpoint within the SHANK3 gene in the 22q13.3

577 deletion syndrome. J Med Genet 43:822–828.

578 Bozdagi O, Sakurai T, Papapetrou D, Wang X, Dickstein DL, Takahashi N, Kajiwara Y, Yang

579 M, Katz AM, Scattoni ML, Harris MJ, Saxena R, Silverman JL, Crawley JN, Zhou Q, Hof

28

580 PR, Buxbaum JD (2010) Haploinsufficiency of the autism-associated Shank3 gene leads to

581 deficits in synaptic function, social interaction, and social communication. Mol Autism 1:15.

582 Choi S-Y, Pang K, Kim JY, Ryu JR, Kang H, Liu Z, Kim W-K, Sun W, Kim H, Han K (2015)

583 Post-transcriptional regulation of SHANK3 expression by microRNAs related to multiple

584 neuropsychiatric disorders. Mol Brain 8:74.

585 Colledge M, Snyder EM, Crozier RA, Soderling JA, Jin Y, Langeberg LK, Lu H, Bear MF, Scott

586 JD (2003) Ubiquitination regulates PSD-95 degradation and AMPA receptor surface

587 expression. Neuron 40:595–607.

588 de Sena Cortabitarte A, Degenhardt F, Strohmaier J, Lang M, Weiss B, Roeth R, Giegling I,

589 Heilmann-Heimbach S, Hofmann A, Rujescu D, Fischer C, Rietschel M, Nöthen MM,

590 Rappold GA, Berkel S (2017) Investigation of SHANK3 in schizophrenia. Am J Med Genet

591 B Neuropsychiatr Genet 174:390–398.

592 Durand CM et al. (2007) Mutations in the gene encoding the synaptic scaffolding protein

593 SHANK3 are associated with autism spectrum disorders. Nat Genet 39:25–27.

594 Durand CM, Perroy J, Loll F, Perrais D, Fagni L, Bourgeron T, Montcouquiol M, Sans N (2012)

595 SHANK3 mutations identified in autism lead to modification of dendritic spine morphology

596 via an actin-dependent mechanism. Mol Psychiatry 17:71–84.

597 Ehlers MD (2003) Activity level controls postsynaptic composition and signaling via the

598 ubiquitin-proteasome system. Nat Neurosci 6:231–242.

599 Failla P, Romano C, Alberti A, Vasta A, Buono S, Castiglia L, Luciano D, Di Benedetto D,

29

600 Fichera M, Galesi O (2007) Schizophrenia in a patient with subtelomeric duplication of

601 22q. Clin Genet 71:599–601.

602 Gauthier J, Spiegelman D, Piton A, Lafrenière RG, Laurent S, St-Onge J, Lapointe L, Hamdan

603 FF, Cossette P, Mottron L, Fombonne E, Joober R, Marineau C, Drapeau P, Rouleau GA

604 (2009) Novel de novo SHANK3 mutation in autistic patients. Am J Med Genet B

605 Neuropsychiatr Genet 150B:421–424.

606 Guilmatre A, Huguet G, Delorme R, Bourgeron T (2014) The emerging role of SHANK in

607 neuropsychiatric disorders. Carbonetto S, Bourgeron T, eds. Dev Neurobiol 74:113–122.

608 Han K, Holder JL, Schaaf CP, Lu H, Chen H, Kang H, Tang J, Wu Z, Hao S, Cheung SW, Yu P,

609 Sun H, Breman AM, Patel A, Lu H-C, Zoghbi HY (2013) SHANK3 overexpression causes

610 manic-like behaviour with unique pharmacogenetic properties. Nature 503:72–77.

611 Hung AY, Sung CC, Brito IL, Sheng M (2010) Degradation of postsynaptic scaffold GKAP and

612 regulation of dendritic spine morphology by the TRIM3 ubiquitin ligase in rat hippocampal

613 neurons. Tell F, ed. PLoS ONE 5:e9842.

614 Jiang Y-H, Ehlers MD (2013) Modeling autism by SHANK gene mutations in mice. Neuron

615 78:8–27.

616 Kato M, Miyazawa K, Kitamura N (2000) A deubiquitinating enzyme UBPY interacts with the

617 Src homology 3 domain of Hrs-binding protein via a novel binding motif

618 PX(V/I)(D/N)RXXKP. J Biol Chem 275:37481–37487.

619 Leblond CS et al. (2014) Meta-analysis of SHANK Mutations in Autism Spectrum Disorders: A

30

620 Gradient of Severity in Cognitive Impairments. Barsh GS, ed. PLoS Genet 10:e1004580.

621 Lee PCW, Dodart J-C, Aron L, Finley LW, Bronson RT, Haigis MC, Yankner BA, Harper JW

622 (2013) Altered social behavior and neuronal development in mice lacking the Uba6-Use1

623 ubiquitin transfer system. Mol Cell 50:172–184.

624 MacGillavry HD, Kerr JM, Kassner J, Frost NA, Blanpied TA (2015) Shank-cortactin

625 interactions control actin dynamics to maintain flexibility of neuronal spines and synapses.

626 Eur J Neurosci 43:179–193.

627 Matsuzaki M, Honkura N, Ellis-Davies GCR, Kasai H (2004) Structural basis of long-term

628 potentiation in single dendritic spines. Nature 429:761–766.

629 Mei Y, Monteiro P, Zhou Y, Kim J-A, Gao X, Fu Z, Feng G (2016) Adult restoration of Shank3

630 expression rescues selective autistic-like phenotypes. Nature 530:481–484.

631 Mizuno E, Iura T, Mukai A, Yoshimori T, Kitamura N, Komada M (2005) Regulation of

632 epidermal growth factor receptor down-regulation by UBPY-mediated deubiquitination at

633 endosomes. Mol Biol Cell 16:5163–5174.

634 Na CH, Jones DR, Yang Y, Wang X, Xu Y, Peng J (2012) Synaptic protein ubiquitination in rat

635 brain revealed by antibody-based ubiquitome analysis. J Proteome Res 11:4722–4732.

636 Naisbitt S, Kim E, Tu JC, Xiao B, Sala C, Valtschanoff J, Weinberg RJ, Worley PF, Sheng M

637 (1999) Shank, a novel family of postsynaptic density proteins that binds to the NMDA

638 receptor/PSD-95/GKAP complex and cortactin. Neuron 23:569–582.

639 Nägerl UV, Eberhorn N, Cambridge SB, Bonhoeffer T (2004) Bidirectional activity-dependent

31

640 morphological plasticity in hippocampal neurons. Neuron 44:759–767.

641 Niendorf S, Oksche A, Kisser A, Löhler J, Prinz M, Schorle H, Feller S, Lewitzky M, Horak I,

642 Knobeloch K-P (2007) Essential role of ubiquitin-specific protease 8 for receptor tyrosine

643 kinase stability and endocytic trafficking in vivo. Mol Cell Biol 27:5029–5039.

644 Nijman SMB, Luna-Vargas MPA, Velds A, Brummelkamp TR, Dirac AMG, Sixma TK,

645 Bernards R (2005) A genomic and functional inventory of deubiquitinating enzymes. Cell

646 123:773–786.

647 Peça J, Feliciano C, Ting JT, Wang W, Wells MF, Venkatraman TN, Lascola CD, Fu Z, Feng G

648 (2011) Shank3 mutant mice display autistic-like behaviours and striatal dysfunction. Nature

649 472:437–442.

650 Reim D, Distler U, Halbedl S, Verpelli C, Sala C, Bockmann J, Tenzer S, Boeckers TM,

651 Schmeisser MJ (2017) Proteomic Analysis of Post-synaptic Density Fractions from Shank3

652 Mutant Mice Reveals Brain Region Specific Changes Relevant to Autism Spectrum

653 Disorder. Front Mol Neurosci 10:26.

654 Roussignol G, Ango F, Romorini S, Tu JC, Sala C, Worley PF, Bockaert J, Fagni L (2005)

655 Shank expression is sufficient to induce functional dendritic spine synapses in aspiny

656 neurons. J Neurosci 25:3560–3570.

657 Sala C, Piëch V, Wilson NR, Passafaro M, Liu G, Sheng M (2001) Regulation of dendritic spine

658 morphology and synaptic function by Shank and Homer. Neuron 31:115–130.

659 Sato D et al. (2012) SHANK1 Deletions in Males with Autism Spectrum Disorder. Am J Hum

32

660 Genet 90:879–887.

661 Schmeisser MJ (2015) Translational neurobiology in Shank mutant mice - Model systems for

662 neuropsychiatric disorders. Ann Anat 200:115–117.

663 Scudder SL, Goo MS, Cartier AE, Molteni A, Schwarz LA, Wright R, Patrick GN (2014)

664 Synaptic strength is bidirectionally controlled by opposing activity-dependent regulation of

665 Nedd4-1 and USP8. J Neurosci 34:16637–16649.

666 Selimbeyoglu A, Kim CK, Inoue M, Lee SY, Hong ASO, Kauvar I, Ramakrishnan C, Fenno LE,

667 Davidson TJ, Wright M, Deisseroth K (2017) Modulation of prefrontal cortex

668 excitation/inhibition balance rescues social behavior in CNTNAP2-deficient mice. Science

669 Translational Medicine 9:eaah6733.

670 Sheng M, Hoogenraad CC (2007) The postsynaptic architecture of excitatory synapses: a more

671 quantitative view. Annu Rev Biochem 76:823–847.

672 Sheng M, Kim E (2000) The Shank family of scaffold proteins. J Cell Sci 113 ( Pt 11):1851–

673 1856.

674 Shin SM, Zhang N, Hansen J, Gerges NZ, Pak DTS, Sheng M, Lee SH (2012) GKAP

675 orchestrates activity-dependent postsynaptic protein remodeling and homeostatic scaling.

676 Nat Neurosci 15:1655–1666.

677 Siegel JJ, Chitwood RA, Ding JM, Payne C, Taylor W, Gray R, Zemelman BV, Johnston D

678 (2017) Prefrontal cortex dysfunction in Fragile X mice depends on the continued absence of

679 Fragile X Mental Retardation Protein in the adult brain. J Neurosci 37:7305–7317.

33

680 Swatek KN, Komander D (2016) Ubiquitin modifications. Cell Res 26:399–422.

681 Tao-Cheng JH, Dosemeci A, Gallant PE, Smith C, Reese T (2010) Activity induced changes in

682 the distribution of Shanks at hippocampal synapses. Neuroscience 168:11–17.

683 Tu JC, Xiao B, Naisbitt S, Yuan JP, Petralia RS, Brakeman P, Doan A, Aakalu VK, Lanahan

684 AA, Sheng M, Worley PF (1999) Coupling of mGluR/Homer and PSD-95 complexes by the

685 Shank family of postsynaptic density proteins. Neuron 23:583–592.

686 Wang X, McCoy PA, Rodriguiz RM, Pan Y, Je HS, Roberts AC, Kim CJ, Berrios J, Colvin JS,

687 Bousquet-Moore D, Lorenzo I, Wu G, Weinberg RJ, Ehlers MD, Philpot BD, Beaudet AL,

688 Wetsel WC, Jiang Y-H (2011) Synaptic dysfunction and abnormal behaviors in mice lacking

689 major isoforms of Shank3. Hum Mol Genet 20:3093–3108.

690 Wilson HL, Wong ACC, Shaw SR, Tse W-Y, Stapleton GA, Phelan MC, Hu S, Marshall J,

691 McDermid HE (2003) Molecular characterisation of the 22q13 deletion syndrome supports

692 the role of haploinsufficiency of SHANK3/PROSAP2 in the major neurological symptoms. J

693 Med Genet 40:575–584.

694 Wong AC, Ning Y, Flint J, Clark K, Dumanski JP, Ledbetter DH, McDermid HE (1997)

695 Molecular characterization of a 130-kb terminal microdeletion at 22q in a child with mild

696 mental retardation. Am J Hum Genet 60:113–120.

697 Wright MH, Berlin I, Nash PD (2011) Regulation of endocytic sorting by ESCRT-DUB-

698 mediated deubiquitination. Cell Biochem Biophys 60:39–46.

699 Wu X, Yen L, Irwin L, Sweeney C, Carraway KL (2004) Stabilization of the E3 ubiquitin ligase

34

700 Nrdp1 by the deubiquitinating enzyme USP8. Mol Cell Biol 24:7748–7757.

701 Yang M, Bozdagi O, Scattoni ML, Wöhr M, Roullet FI, Katz AM, Abrams DN, Kalikhman D,

702 Simon H, Woldeyohannes L, Zhang JY, Harris MJ, Saxena R, Silverman JL, Buxbaum JD,

703 Crawley JN (2012) Reduced excitatory neurotransmission and mild autism-relevant

704 phenotypes in adolescent Shank3 null mutant mice. J Neurosci 32:6525–6541.

705

706

707

35

708 Figure Legends

709 Figure 1: Screen for Deubiquitinating Enzymes that Modulate SHANK3

710 A, DIV21 dissociated rat cortical neurons were treated for 3HRS with DMSO (vehicle) or 1 μM

711 MG132 (proteasome inhibitor) and were stained for endogenous SHANK3. SHANK3

712 immunofluorescence intensity increases with MG132 treatment. Scale bar 5 μm. n=10 images, 1

713 culture, Student’s t-test p=0.0248. B, Stable human SHANK3-expressing cell line shows

714 increase in SHANK3 protein levels upon proteasome inhibition with 0.5 μM MG132 over time.

715 C, Representative western blots of SHANK3 and ACTIN after overexpression of individual

716 FLAG-tagged DUBs from a human cDNA library for 76HRS in stable SHANK3-expressing

717 cells. Blots show ßGAL control, ßGAL with MG132 treatment as a positive control, the 7 DUBs

718 that increase SHANK3 protein levels, and 3 control DUBs that do not show a difference from

719 control, USPL1, USP1, USP30. D, Quantification of SHANK3/ACTIN intensity reveals 7 DUBs

720 with statistically significant increases compared to ßGAL control. ßGAL transfected cells treated

721 with MG132 was used as a positive control. n=5 biological replicates with 2-3 technical

722 replicates per experiment. One-way ANOVA F=2.316, p<0.0001, post-hoc Student’s t-test

723 EGAL vs. MG132 p=0.0001, EGAL vs. USP3 p=0.0033, EGAL vs. USP22 p=0.0279, EGAL vs.

724 HDAC6 p=0.0112, EGAL vs. ATXN3 p=0.0332, EGAL vs. UCHL1 p=0.0249, EGAL vs.

725 JOSD1 p=0.0427, EGAL vs. USP8 p=0.021. E-G, DIV15 rat hippocampal neurons were

726 transfected with GFP, SHANK3-TdTomato, and individual FLAG-tagged DUBs (as indicated)

727 for 24HRS. Confocal microscope images were obtained by observers blinded to transfection

728 conditions, and collapsed z-stack images were quantified by a single blinded experimenter.

729 Overexpression of USP8 results in a significant increase in SHANK3-TdTomato/GFP intensity

730 (E) and dendritic spine density (F). n=3-5 independent cultures, ~10 neurons quantified for each

36

731 condition per culture. Intensity: One-way ANOVA F=3.093, p=0.0146, post-hoc Student’s t-test

732 EGAL vs. USP8 p=0.0002; Spine Density: One-way ANOVA F=4.595, p=0.0011, post-hoc

733 Student’s t-test EGAL vs. USP8 p=0.0068. G, Representative images: scale bar 20 μm. H, I,

734 Overexpression of USP8 in combination with 1 μM MG132 treatment (USP8+MG132) for

735 24HRS does not have an additive effect compared with USP8 or MG132 alone. One-way

736 ANOVA F=12.18, p=0.0002, post-hoc Student’s t-test EGAL vs. EGAL + MG132 p=0.004,

737 EGAL vs. USP8 p=0.0061, EGAL vs. USP8 + MG132 p=0.0007. J, Quantification by TaqMan

738 qPCR of total SHANK3 mRNA relative to HPRT1 mRNA in stable human-SHANK3 expressing

739 HEK293 cells following overexpression of either EGAL or USP8. Student’s t-test, p=0.077.

740

741

742 Figure 2: Overexpression of Deubiquitinating Enzymes in Dissociated Neurons

743 A, B, DIV15 rat hippocampal neurons were transfected with GFP and individual FLAG-tagged

744 DUBs (as indicated) for 24HRS. Confocal microscope images were obtained by observers

745 blinded to transfection conditions, and collapsed z-stack images were quantified by a single

746 blinded experimenter. Overexpression of USP8 results in a significant increase in endogenous

747 SHANK3/GFP intensity (A) and dendritic spine density (B). n=3-5 independent cultures, ~10

748 neurons quantified for each condition per culture. Intensity: One-way ANOVA F=8.605,

749 p<0.0001, post-hoc Student’s t-test EGAL vs. USP8 p<0.0001; Spine Density: One-way

750 ANOVA F=2.478, p=0.018, post-hoc Student’s t-test EGAL vs. USP8 p=0.0008. C,D,

751 Overexpression of catalytically dead mutant USP8-C786S in the human SHANK3-expressing

752 stable cells does not increase SHANK3 protein levels like WT USP8. n=9 independent

753 transfections per construct, One-way ANOVA F=16.28, p<0.0001, post hoc Student’s t-test

37

754 EGAL vs. USP8 p<0.0001, EGAL vs. C786S p=0.1062, USP8 vs. C786S p=0.0022. E, F,

755 Overexpression of WT USP8 in DIV15 rat hippocampal cultures results in an increase in

756 SHANK3-TdTomato intensity and dendritic spine density compared to ßGAL control or USP8-

757 C786S (enzyme dead). n=3 independent cultures ~10 neurons/culture. Intensity: One-way

758 ANOVA F=9.338, p=0.0002, post-hoc Student’s t-test EGAL vs. USP8 p=0.0002, EGAL vs.

759 C786S p=0.6652, USP8 vs. C786S p=0.0176; Spine Density: One-way ANOVA F=7.175,

760 p=0.0015, post-hoc Student’s t-test EGAL vs. USP8 p=0.0079, EGAL vs. C786S p=0.3168,

761 USP8 vs. C786S p=0.0007. (F) Representative images: scale bar 5 μm. G-I, DIV15 rat

762 hippocampal neurons were transfected with GFP, and USP8 or USP8-C786S (enzyme dead) for

763 24HRS and stained for endogenous SHANK3. Confocal microscope images were obtained by

764 observers blinded to transfection conditions and collapsed z-stack images were quantified by a

765 single blinded experimenter. Overexpression of USP8, but not USP8-C786S results in an

766 increase in immunofluorescence intensity of endogenous SHANK3 clusters (G) and in dendritic

767 spine density (H) compared to control. n=3 independent cultures, ~10 neurons quantified for

768 each condition per culture. Intensity: One-way ANOVA F=11.46, p<0.0001, post-hoc Student’s

769 t-test EGAL vs. USP8 p=0.0001, EGAL vs. C786S p=0.6596, USP8 vs. C786S p=0.0027; Spine

770 Density: One-way ANOVA F=9.888, p=0.0001, post-hoc Student’s t-test EGAL vs. USP8

771 p=0.0026, EGAL vs. C786S p=0.2037, USP8 vs. C786S p=0.0007. (I) Representative images:

772 scale bar 5 μm.

773

774 Figure 3: USP8 Regulates SHANK3 Protein Levels and Dendritic Spine Density in Neurons

775 A, Primary hippocampal neurons were treated from DIV10-18 with Accell siRNA to knockdown

776 individual DUBs (as indicated) or SHANK3. SHANK3 protein levels were measured by

38

777 immunoblot and normalized to ACTIN. Knockdown of SHANK3 or USP8 resulted in a

778 significant loss of SHANK3 protein levels relative to control. One-way ANOVA F=2.899,

779 p=0.0051, post-hoc Student’s t-test Control vs. SHANK3 p=0.0007, Control vs. USP8 p=0.0046.

780 B, Representative western blots from knockdown with control (CTRL) or USP8 siRNA. C,

781 Knockdown of USP8 or SHANK3 with Dharmacon ON-TARGET siRNA pools in stable

782 SHANK3-expressing cells for 72HRS significantly reduces SHANK3 protein levels. n=4

783 independent cultures, One-way ANOVA F=137.1, p<0.0001, post-hoc Student’s t-test CTRL vs.

784 USP8 p=0.0006, CTRL vs. SHANK3 p<0.0001, USP8 vs. SHANK3 p<0.0001. D, HEK293 cells

785 were transfected with MYC-tagged rat USP8 and 4 shRNA constructs targeting rat USP8 or a

786 scrambled shRNA control for 72HRS. Cell lysates were blotted for MYC and ACTIN loading

787 control. shRNA constructs A and B efficiently knocked down USP8 65-90% and were used in all

788 following experiments. E, Representative images: scale bar 5 μm. F, Rat hippocampal

789 dissociated neurons were transfected at DIV14 with a scrambled shRNA or one of two shRNAs

790 that target rat USP8 (USP8_A, USP8_B) for 72HRS. At DIV17 neurons were fixed and stained

791 for endogenous SHANK3. Collapsed z-stack images were thresholded and a mask was

792 developed to identify SHANK3 puncta by a single blinded experimenter in a 20 Pm x 5 Pm

793 region of interest. Knockdown of USP8 results in decreased SHANK3 puncta intensity, reduced

794 SHANK3 puncta size, and less SHANK3 puncta/20 Pm of dendrite. n=3 independent cultures,

795 ~10 neurons quantified for each condition per culture. Puncta intensity: One-way ANOVA

796 F=15.89, p<0.0001, post-hoc Student’s t-test scrambled vs. USP8_A p<0.0001, scrambled vs.

797 USP8_B p<0.0001, USP8_A vs. USP8_B p=0.9341; Puncta Size: One-way ANOVA F=19.68,

798 p<0.0001, post-hoc Student’s t-test scrambled vs. USP8_A p<0.0001, scrambled vs. USP8_B

799 p<0.0001, USP8_A vs. USP8_B p=0.9352; Puncta Count: One-way ANOVA F=18.50,

39

800 p<0.0001, post-hoc Student’s t-test scrambled vs. USP8_A p<0.0001, scrambled vs. USP8_B

801 p<0.0001, USP8_A vs. USP8_B p=0.9943. G, Cumulative frequency of SHANK3 puncta

802 intensity and puncta size was significantly reduced in USP8 knockdown neurons relative to

803 control. Cumulative Frequency Puncta Intensity: scrambled n=374, USP8_A n=184, USP8_B

804 n=196, Kolmogorov-Smirnov test scrambled vs. USP8_A p=0.0007, scrambled vs. USP8_B

805 p=0.0002, USP8_A vs. USP8_B p=0.9473; Cumulative Frequency Puncta Size: scrambled

806 n=375, USP8_A n=184, USP8_B n=196, scrambled vs. USP8_A p=0.0012, scrambled vs.

807 USP8_B p<0.0001, USP8_A vs. USP8_B p=0.9410.

808

809 Figure 4: Knockdown of USP8 Leads to a Loss of Synaptic Puncta and Regulates SHANK1

810 Puncta Size and Intensity

811 A, Rat hippocampal dissociated neurons were transfected at DIV14 with a scrambled shRNA or

812 one of two shRNAs that target rat USP8 (USP8_A, USP8_B) for 72HRS. At DIV17 neurons

813 were fixed and stained for endogenous synaptic proteins with antibodies to SHANK1, SHANK2,

814 PSD-95, GKAP, and HOMER1A. Collapsed z-stack images were thresholded and a mask was

815 developed to identify synaptic protein puncta by a single blinded experimenter in a 20 Pm x 5

816 Pm region of interest. Knockdown of USP8 causes a reduction in SHANK1 synaptic puncta

817 intensity and size, while SHANK2, PSD-95, GKAP, and HOMER1A puncta are unaffected. The

818 number of synaptic protein puncta per 20 μm are reduced for all, with the exception of

819 HOMER1A not reaching significance. n=3-5 independent cultures, ~10 neurons quantified for

820 each condition per culture. Puncta intensity SHANK1: One-way ANOVA F=12.37, p<0.0001,

821 post-hoc Student’s t-test scrambled vs. USP8_A p<0.0001, scrambled vs. USP8_B p=0.0014,

822 USP8_A vs. USP8_B p=0.5416; Puncta intensity SHANK2: One-way ANOVA F=0.5540,

40

823 p=0.5772; Puncta intensity PSD-95: One-way ANOVA F=2.583, p=0.0837; Puncta intensity

824 GKAP: One-way ANOVA F=0.4559, p=0.6356; Puncta intensity HOMER1A: One-way

825 ANOVA F=1.899, p=0.1566; Puncta size SHANK1: One-way ANOVA F=4.553, p=0.0147,

826 post-hoc Student’s t-test scrambled vs. USP8_A p=0.0371, scrambled vs. USP8_B p=0.0304,

827 USP8_A vs. USP8_B p=0.9995; Puncta size SHANK2: One-way ANOVA F=0.5615, p=0.5728;

828 Puncta size PSD-95: One-way ANOVA F=1.804, p=0.1732; Puncta size GKAP: One-way

829 ANOVA F=0.7936, p=0.4559; Puncta size HOMER1A: One-way ANOVA F=1.222, p=0.3002;

830 Puncta number SHANK1: One-way ANOVA F=9.892, p=0.0002, post-hoc Student’s t-test

831 scrambled vs. USP8_A p=0.0002, scrambled vs. USP8_B p=0.0054, USP8_A vs. USP8_B

832 p=0.4991; Puncta number SHANK2: One-way ANOVA F=6.297, p=0.0030, post-hoc Student’s

833 t-test scrambled vs. USP8_A p=0.0031, scrambled vs. USP8_B p=0.0245, USP8_A vs. USP8_B

834 p=0.7621; Puncta number PSD-95: One-way ANOVA F=13.88, p<0.0001, post-hoc Student’s t-

835 test scrambled vs. USP8_A p=0.0006, scrambled vs. USP8_B p<0.0001, USP8_A vs. USP8_B

836 p=0.5508; Puncta number GKAP: One-way ANOVA F=5.567, p=0.0054, post-hoc Student’s t-

837 test scrambled vs. USP8_A p=0.0066, scrambled vs. USP8_B p=0.0302, USP8_A vs. USP8_B

838 p=0.8321; Puncta number HOMER1A: One-way ANOVA F=1.621, p=0.2040. B,

839 Representative images: scale bar 5 μm.

840

841 Figure 5: USP8 Regulates the Ubiquitination of SHANK3 and SHANK1

842 A, Representative immunoblots from assays that used Tandem Ubiquitin Binding Entities

843 (TUBE) coupled to magnetic beads to pulldown polyubiquitinated proteins. Stable SHANK3-

844 expressing cells were transfected with EGAL control, USP8, or USP8-C786S (enzymatic dead)

845 and treated with the proteasome inhibitor MG132 for 3HRS prior to lysis and pulldown. B,

41

846 Intensity of SHANK3 immunoblot bands from pulldown lanes were quantified. Overexpression

847 of USP8 leads to a decrease in the level of polyubiquitinated SHANK3 relative to EGAL control

848 or USP8-C786S. n=4 assays from independent cultures. One-way ANOVA F=12.63, p=0.0024,

849 EGAL vs. USP8 p=0.0264, EGAL vs. C786S p=0.2388, USP8 vs. C786S p=0.0020. C,

850 Representative immunoblots of TUBE pulldown assays from HEK293 lysates transiently

851 transfected with SHANK1, SHANK2, PSD-95, or GKAP along with EGAL, USP8, or USP8-

852 C786S. D, Quantification of TUBE pulldown assays from panel C. SHANK1 was less

853 ubiquitinated when USP8 was overexpressed relative to EGAL control or USP8-C786S.

854 SHANK2, PSD-95, and GKAP polyubiquitination were unaffected by USP8 overexpression.

855 n=3-5 assays from independent cultures. SHANK1: One-way ANOVA F=14.53, p=0.0015,

856 EGAL vs. USP8 p=0.0085, EGAL vs. C786S p=0.4814, USP8 vs. C786S p=0.0016; SHANK2:

857 One-way ANOVA F=0.9666, p=0.4326; PSD-95: One-way ANOVA F=3.235, p=0.5189;

858 GKAP: One-way ANOVA F=0.03123, p=0.9693. E, Ubiquitinated SHANK proteins were

859 immunoprecipitated from HEK293 cells overexpressing SHANK1, 2, or 3 together with HA-

860 ubiquitin and treated with MG132. Immunoprecipitated SHANK1, 2, or 3 was incubated with

861 vehicle, recombinant USP8, or recombinant USP8 plus the DUB inhibitor N-Ethylmaleimide

862 (NEM) in an in vitro deubiquitination assay. HA-ubiquitin signal on SHANK3 and SHANK1

863 was reduced by recombinant USP8.

864

865 Figure 6: Loss of USP8 Occludes the Bidirectional Changes in SHANK3 Puncta Intensity

866 and Size in Response to Chronic Synaptic Activity Modulation

867 A,B, Dissociated rat cortical neurons were transfected with GFP, and a scrambled shRNA or one

868 of 2 shRNAs that target rat USP8 (USP8_A, USP8_B) for 72HRS. Cultures were treated with

42

869 DMSO, TTX (1 PM), or BICC (40 PM) for the final 24HRS prior to fixation. Neurons were

870 stained for endogenous synaptic proteins with antibodies to SHANK3 or PSD-95. Collapsed z-

871 stack images were thresholded and a mask was created to identify synaptic puncta. All

872 quantifications were done by a single blinded experimenter. (A) Knockdown of USP8 causes a

873 significant reduction (#) in SHANK3 puncta fluorescence intensity and size, as shown previously

874 (Fig 3F). Additionally, knockdown of USP8 blocks the significant bidirectional changes (*)

875 observed upon treatment with TTX or BICC in control neurons. n=3 independent cultures, ~8

876 neurons quantified for each condition per culture. Puncta intensity within drug treatment p=*:

877 One-way ANOVA F=8.273, p<0.0001, post-hoc Student’s t-test scrambled DMSO vs. scrambled

878 TTX p=0.0174, scrambled DMSO vs. scrambled BICC p=0.0463, scrambled TTX vs. scrambled

879 BICC p=0.0004, USP8_A DMSO vs. USP8_A TTX p=0.8546, USP8_A DMSO vs. USP8_A

880 BICC p=0.9759, USP8_A TTX vs. USP8_A BICC p=0.8644, USP8_B DMSO vs. USP8_B TTX

881 p=0.5462, USP8_B DMSO vs. USP8_B BICC p=0.8134, USP8_B TTX vs. USP8_B BICC

882 p=0.4643, Puncta intensity between shRNA treatment p=#: scrambled DMSO vs. USP8_A

883 DMSO p=0.0161, scrambled DMSO vs. USP8_A TTX p=0.0023, scrambled DMSO vs.

884 USP8_A BICC p=0.0094, scrambled DMSO vs. USP8_B DMSO p=0.0249, scrambled DMSO

885 vs. USP8_B TTX p=0.0902, scrambled DMSO vs. USP8_B BICC p=0.0304; Puncta size within

886 drug treatment p=*: One-way ANOVA F=7.330, p<0.0001, post-hoc Student’s t-test scrambled

887 DMSO vs. scrambled TTX p=0.0197, scrambled DMSO vs. scrambled BICC p=0.0293,

888 scrambled TTX vs. scrambled BICC p=0.0004, USP8_A DMSO vs. USP8_A TTX p=0.6818,

889 USP8_A DMSO vs. USP8_A BICC p=0.4774, USP8_A TTX vs. USP8_A BICC p=0.6699,

890 USP8_B DMSO vs. USP8_B TTX p=0.3103, USP8_B DMSO vs. USP8_B BICC p=0.9049,

891 USP8_B TTX vs. USP8_B BICC p=0.3346, Puncta size between shRNA treatment p=#:

43

892 scrambled DMSO vs. USP8_A DMSO p=0.0123, scrambled DMSO vs. USP8_A TTX

893 p=0.0119, scrambled DMSO vs. USP8_A BICC p=0.0488, scrambled DMSO vs. USP8_B

894 DMSO p=0.0288, scrambled DMSO vs. USP8_B TTX p=0.2267, scrambled DMSO vs. USP8_B

895 BICC p=0.0406. (B) Knockdown of USP8 has no change on overall PSD-95 puncta fluorescence

896 intensity or puncta size, as shown previously (Fig 4A) and PSD-95 in USP8 knockdown neurons

897 still responds synaptic activity modulation (*). n=3 independent cultures, ~8 neurons quantified

898 for each condition per culture. Puncta intensity within drug treatment: One-way ANOVA

899 F=15.45, p<0.0001, post-hoc Student’s t-test scrambled DMSO vs. scrambled TTX p=0.0026,

900 scrambled DMSO vs. scrambled BICC p=0.0057, scrambled TTX vs. scrambled BICC

901 p<0.0001, USP8_A DMSO vs. USP8_A TTX p<0.0001, USP8_A DMSO vs. USP8_A BICC

902 p=0.0338, USP8_A TTX vs. USP8_A BICC p<0.0001, USP8_B DMSO vs. USP8_B TTX

903 p=0.0013, USP8_B DMSO vs. USP8_B BICC p=0.0032, USP8_B TTX vs. USP8_B BICC

904 p<0.0001; Puncta size within drug treatment: One-way ANOVA F=4.753, p<0.0001, post-hoc

905 Student’s t-test scrambled DMSO vs. scrambled TTX p=0.0231, scrambled DMSO vs.

906 scrambled BICC p=0.2250, scrambled TTX vs. scrambled BICC p=0.0005, USP8_A DMSO vs.

907 USP8_A TTX p=0.0091, USP8_A DMSO vs. USP8_A BICC p=0.6029, USP8_A TTX vs.

908 USP8_A BICC p=0.0002, USP8_B DMSO vs. USP8_B TTX p=0.0162, USP8_B DMSO vs.

909 USP8_B BICC p=0.7856, USP8_B TTX vs. USP8_B BICC p=0.0092. C, Representative

910 images: scale bar 5 μm.

911

912

44 A B 0.5 μM MG132

DMSO MG132 HRS: 0 4 6 10 24 2.0 * SHANK3 1.5 C

1.0 MG132

0.5 USP3 ßGAL UCHL1ATXN3ßGAL+HDAC6JOSD1 USP8 USPL1USP1 USP30USP22

fluorescence intensity fluorescence 0.0 SHANK3 Normalized SHANK3 puncta ACTIN DMSO MG132

D DUB OVEREXPRESSION IN 293 CELLS E DUB OVEREXPRESSION IN NEURONS 5 *** 6 *** ** ****** 4

4 3

2 2 /GFP intensity /GFP 1 Normalized SHANK3/ACTIN Normalized SHANK3-tomato 0 0

ßGAL USP3 USP8 USP22 ßGAL USP3 USP8 UCHL1 HDAC6ATXN3 JOSD1 F UCHL1 USP22HDAC6 ATXN3 JOSD1 1.0 ** ßGAL+MG132 H ßGAL USP8 0.8 ßGAL +MG132 USP8 +MG132

SHANK3 0.6 USP8- FLAG 0.4 Spines/μm ACTIN I 0.2 2.5 ** J ** 0.0 *** 2.0 1.0 ßGAL USP3 USP8 UCHL1 USP22HDAC6 ATXN3 JOSD1 1.5 G ßGAL USP8

1.0 0.5

0.5 SHANK3 mRNA Normalized SHANK/ACTIN Normalized 0.0 0.0 SHANK3-tomato GAL GAL GAL ß ß USP8 USP8 ß USP8 +MG132 +MG132 AB**** 6 1.5 ***

4 1.0

2 Spines/μm 0.5 SHANK3/GFP intensity SHANK3/GFP Normalized endogenous

0 0.0

ßGAL USP3 USP8 ßGAL USP3 USP8 UCHL1 USP22HDAC6 ATXN3 JOSD1 UCHL1 USP22HDAC6 ATXN3 JOSD1 C 2.0 E ** *** *** ** *** * 0.8 3 1.5 0.6

2 1.0 0.4 Spines/μm 0.5 1 0.2 /GFP intensity /GFP

0.0 0 0.0 Normalized SHANK/ACTIN Normalized Normalized SHANK3-tomato GAL ß USP8 USP8- ßGAL USP8 USP8- ßGAL USP8 USP8- C786S C786S C786S

D F ßGAL USP8 USP8-C786S USP8- ßGAL USP8 C786S SHANK3-tomato

SHANK3 GFP USP8- FLAG MERGE

ACTIN GHI 1.0 ** *** 6 *** ** ßGAL USP8 USP8-C786S 0.8 SHANK3 4 0.6 GFP

0.4 2 USP8 Spines/μm 0.2 MERGE SHANK3/GFP intensity SHANK3/GFP Normalized endogenous 0 0.0

ßGAL USP8 USP8- ßGAL USP8 USP8- C786S C786S AB 4 *** ** siRNA: 3 CTRL USP8

SHANK3 2

USP8 1 ACTIN Normalized SHANK3/ACTIN 0 1

CTRL USP3 USP8 UCHL USP22HDAC6ATXN3JOSD1 SHANK3 CEshRNA: Scrambled USP8_A USP8_B siRNA: CTRL USP8 SHANK3 SHANK3 SHANK3 GFP USP8 MERGE ACTIN F *** *** 2 0.6 25 1.5 *** *** 400 *** *** *** *** *** 20 300 0.4 1.0 15 200 10 0.5 0.2 100 5 SHANK3 puncta fluorescence intensity fluorescence 0.0 0 0.0 0 SHANK3 puncta size (μm ) Normalized SHANK/ACTIN puncta/20μm of SHANK3 #

CTRL USP8 USP8_AUSP8_B USP8_AUSP8_B USP8_AUSP8_B SHANK3 Scrambled Scrambled Scrambled D shRNA to rat USP8 G 100 100 Sc D C B A

MYC-USP8 50 50 ACTIN Scrambled Scrambled USP8_A *** USP8_A ** USP8_B *** USP8_B *** 0 0 0 frequency (%) Cumulative 01234 Cumulative frequency (%) Cumulative 500 1000 1500 2000 2500 2 SHANK3 Spine Intensity Spine Size μm AB Scrambled 400 ** *** USP8_A Scrambled USP8_A USP8_B USP8_B SHANK1 300 GFP

200 MERGE

100 SHANK2 GFP

Puncta fluorescence intensity 0 0.5 MERGE * *

2 0.4 PSD-95

GFP 0.3 MERGE 0.2

Puncta size (μm ) GKAP 0.1 GFP 0 20 MERGE ** * * * *** ** ** ** 15 HOMER1A GFP 10 MERGE

5 # of of puncta/20μm #

0

GKAP SHANK1 SHANK2 PSD-95 HOMER1A A INPUT TUBE CTRL B

2.0 *** 1.5 ßGALUSP8USP8-C786SßGALUSP8USP8-C786SßGALUSP8USP8-C786S SHANK3 1.0

0.5

UBIQUITIN 0.0 Normalized SHANK3 intensity SHANK3 Normalized ßGAL USP8

USP8-C786S

USP8-FLAG

CDINPUT TUBE

2.0 ßGAL ** ** USP8 ßGALUSP8USP8-C786SßGALUSP8USP8-C786S 1.5 SHANK1 USP8-C786S

SHANK2 1.0

PSD-95 0.5 Normalized intensity GKAP 0.0 SHANK1 SHANK2 PSD-95 GKAP E

VEHICLEUSP8USP8 + NEM VEHICLEUSP8USP8 + NEM VEHICLEUSP8USP8 + NEM

SHANK3 SHANK1 SHANK2

HA-UBIQUITIN HA-UBIQUITIN HA-UBIQUITIN A * 0.8

200 2 * * *** ****

0.6 150

0.4 100 ###### # ### # #

50 0.2 SHANK3 puncta fluroscence intensity fluroscence 0 SHANK3 puncta size μm 0.0

TTX BICC TTX BICC TTX BICC TTX BICC TTX BICC TTX BICC DMSO DMSO DMSO DMSO DMSO DMSO Scrambled shRNA A shRNA B Scrambled shRNA A shRNA B B * *** * *** * ** 0.5 300 ** * ** 2 ** *** *** *** ** *** 0.4

200 0.3

0.2 100

PSD-95 puncta 0.1 fluroscence intensity fluroscence 0 PSD-95 puncta size μm 0.0 X X TTX BICC TT BICC TTX BICC TTX BICC TT BICC TTX BICC DMSO DMSO DMSO DMSO DMSO DMSO Scrambled shRNA A shRNA B Scrambled shRNA A shRNA B C DMSO DMSO

Scrambled USP8_A USP8_B Scrambled USP8_A USP8_B PSD95 SHANK3 GFP GFP MERGE MERGE TTX TTX PSD95 SHANK3 GFP GFP MERGE MERGE BICC BICC PSD95 SHANK3 GFP GFP MERGE MERGE