Research Articles: Behavioral/Cognitive Neuronal Myocyte-specific enhancer factor 2D (MEF2D) is required for normal circadian and sleep behavior in mice https://doi.org/10.1523/JNEUROSCI.0411-19.2019

Cite as: J. Neurosci 2019; 10.1523/JNEUROSCI.0411-19.2019 Received: 20 February 2019 Revised: 8 July 2019 Accepted: 10 August 2019

This Early Release article has been peer-reviewed and accepted, but has not been through the composition and copyediting processes. The final version may differ slightly in style or formatting and will contain links to any extended data.

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

Copyright © 2019 the authors

1 Title: Neuronal Myocyte-specific enhancer factor 2D (MEF2D) is required for 2 normal circadian and sleep behavior in mice 3 4 Abbreviated Title: Neuronal MEF2D and sleep behavior in mice 5 6 Jennifer A. Mohawk1,2, Kimberly H. Cox1, Makito Sato2,3, Seung-Hee Yoo1, Masashi 7 Yanagisawa2,3, Eric N. Olson4, Joseph S. Takahashi1,2* 8 9 Affiliations: 10 1 Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, 11 TX 75390-9111, USA 12 13 2 Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, 14 TX 75390-9111, USA 15 16 3 Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, 17 TX 75390-9111, USA 18 19 4 Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, 20 University of Texas Southwestern Medical Center, Dallas, TX 75390, USA 21 22 23 * To whom correspondence should be addressed: [email protected] 24 25 Number of pages: 25 26 Number of figures: 6 27 Number of tables: 1 28 Number of extended data figures: 0 29 Number of words for abstract: 186 30 for introduction: 616 31 for discussion: 1141 32 33 Conflict of Interest Statement: The authors declare no competing financial interests. 34 35 Acknowledgements: This work was supported by the FIRST program from JSPS and 36 the WPI program from Japan's MEXT (M.Y.); NIH grants AR-067294 and HL-130253 37 and the Robert A. Welch Foundation grant 1-0025 (E.N.O.); and 38 NIH grants MH078024 and AG045795 (J.S.T.). M.Y. was, and J.S.T. is an Investigator 39 in the Howard Hughes Medical Institute. The authors would like to thank Dr. Rhonda 40 Bassel-Duby for contributing the Mef2d knockout and floxed mice, and Dr. Robert 41 Greene for suggesting the analysis in Figure 6g. 42 43 J.A. Mohawk’s current address: 2951 Gentle Creek Trail, Prosper, TX 75078. M. 44 Sato’s and M. Yanagisawa’s current address: International Institute for Integrative 45 Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba 305-8575, Japan. S-H. 46 Yoo’s current address: Department of Biochemistry and Molecular Biology, The 47 University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 48 77030, USA.

49 ABSTRACT 50 51 The , myocyte enhancer factor-2 (), is required for

52 normal circadian behavior in Drosophila; however, its role in the mammalian circadian

53 system has not been established. Of the four mammalian Mef2 , Mef2d is highly

54 expressed in the suprachiasmatic nucleus (SCN) of the hypothalamus, a region critical

55 for coordinating peripheral circadian clocks. Utilizing both conventional and brain-

56 specific Mef2d knockout (Mef2d-/-) mouse lines, we demonstrate that MEF2D is

57 essential for maintaining the length of the circadian free-running period of locomotor

58 activity and normal sleep patterns in male mice. Crossing Mef2d-/- with Per2::luc

59 reporter mice, we show that these behavioral changes are achieved without altering the

60 endogenous period of the master circadian oscillator in the SCN. Together, our data

61 suggest that alterations in behavior in Mef2d-/- mice may be the result of an effect on

62 SCN output, rather than an effect on timekeeping within the SCN itself. These findings

63 add to the growing body of evidence that MEF2 play important roles in the

64 brain.

65 66 67 SIGNIFICANCE STATEMENT 68

69 These studies are the first to show a role for MEF2 proteins in the brain outside

70 of the hippocampus, and our findings suggest that these proteins may play diverse roles

71 in the central nervous system. It is important to continue to build on our understanding

72 of the roles of proteins acting in the SCN, because SCN dysfunction underlies jet lag in

73 humans and influences the response to shift work schedules, which are now known as

1

74 risk factors for the development of cancer. Our work on MEF2D could be the basis for

75 opening new lines of research in the development and regulation of circadian rhythms.

76 77 INTRODUCTION 78 79 The circadian is essential to the proper timing and regulation of behavior

80 and physiology. In mammals, a master pacemaker in the suprachiasmatic nucleus

81 (SCN) of the ventral hypothalamus coordinates circadian clocks throughout the

82 organism (Welsh et al., 2010; Albrecht, 2012; Herzog et al., 2017). The circadian time-

83 keeping mechanism is composed of a transcriptional autoregulatory feedback loop. At

84 the core of this loop, CLOCK and BMAL1 drive transcription of Period (Per1, Per2) and

85 Cryptochrome (Cry1, Cry2), whose products, in turn, dimerize, translocate back

86 into the nucleus, and repress the activity of CLOCK and BMAL1, thereby inhibiting their

87 own expression. In addition, accessory loops interact with the core clock loop, forming a

88 web of modulators contributing to the refinement of the oscillatory network and,

89 ultimately, circadian biological outputs (Shearman et al., 2000; Ueda et al., 2005;

90 Takahashi, 2017).

91 The transcription factor myocyte enhancer factor-2 (MEF2) has been linked to

92 the maintenance of normal circadian behavior in Drosophila (Blanchard et al., 2010;

93 Sivachenko et al., 2013). Overexpression of Mef2 in clock neurons leads to lengthening

94 of the free-running period of locomotor activity and weak behavioral rhythms in flies.

95 Repressing Mef2 activity has a similar effect, with flies displaying weakly rhythmic or

96 arrhythmic patterns of locomotor activity (Blanchard et al., 2010). However, the role of

97 MEF2 in the mammalian circadian system has not been established.

2

98 In vertebrates, there are four Mef2 genes: Mef2a, Mef2b, Mef2c, and Mef2d.

99 These genes display both differential and overlapping expression patterns throughout

100 the body, but are most abundantly expressed in muscle (both skeletal and cardiac)

101 (Black and Olson, 1998) and brain (Leifer et al., 1993; Ikeshima et al., 1995; Lyons et

102 al., 1995). Their protein products play vital roles in muscle development and stress- and

103 activity-dependent signaling (Shalizi and Bonni, 2005; Potthoff and Olson, 2007;

104 Dietrich, 2013). In neurons, MEF2 targets a diverse set of genes involved in synapse

105 development and remodeling (Flavell et al., 2006; Flavell et al., 2008), while loss of

106 MEF2 has been associated with neuronal apoptosis (Mao et al., 1999; Li et al., 2001;

107 Liu et al., 2003; Akhtar et al., 2012). Both in vitro and in vivo studies have shown that

108 MEF2 activation or overexpression can lead to the reduction of excitatory synapses in

109 hippocampal neurons (Barbosa et al., 2008; Pfeiffer et al., 2010). Interestingly, while

110 prenatal loss of Mef2c in the brain has been shown to impair hippocampal-dependent

111 learning in mice (Barbosa et al., 2008; Akhtar et al., 2012; Rashid et al., 2014),

112 postnatal loss of Mef2c had no such behavioral effect (Adachi et al., 2016), and

113 overexpression of MEF2 in discrete regions within the hippocampus disrupts memory

114 formation (Cole et al., 2012; Rashid et al., 2014). These findings suggest a complex role

115 for MEF2 proteins in the development and function of the hippocampus, but the impact

116 of loss of MEF2 on non-hippocampal dependent behaviors has not been thoroughly

117 investigated.

118 Global deletion of Mef2a or Mef2c is lethal in mice, while Mef2d null mice are

119 viable (Arnold et al., 2007; Kim et al., 2008; Omori et al., 2015) and Mef2d is highly

120 expressed in the SCN(Lein et al., 2007). Therefore, utilizing both a conventional and

3

121 brain-specific Mef2d KO, we began our investigation of the role of MEF2 proteins in the

122 regulation of mammalian circadian behavior. We demonstrate that Mef2D is essential

123 for maintaining the length of the circadian free-running period of locomotor activity. Loss

124 of Mef2D leads to a lengthening of the behavioral period and to disruption and

125 fragmentation of sleep patterns without altering the endogenous period of the master

126 circadian oscillator. Importantly, these effects are specific to Mef2D, as loss of either

127 Mef2A or Mef2C in the brain had no effect on locomotor free-running period.

128 129 MATERIALS AND METHODS 130 131

132 Animals.

133 Mef2d-/- mice (Arnold et al., 2007; Kim et al., 2008) and wild-type littermates on a

134 (129SvEv/C57BL/6J) mixed genetic background were derived from heterozygote

135 crosses. Brain-specific Mef2a, Mef2c, and Mef2d heterozygote and knockout mice (and

136 floxed, cre-negative controls) were derived from crossing CamKIIa-iCre mice (Casanova

137 et al., 2001) with Mef2a/c/dfx/fx triple-floxed mice (Barbosa et al., 2008; Liu et al., 2014;

138 Adachi et al., 2016) and intercrossing to obtain the desired genotypes. For

139 bioluminescence recordings to assess circadian rhythms in the SCN and peripheral

140 tissues, Mef2d-/- mice were crossed with mice carrying the Per2::luc reporter (Yoo et al.,

141 2004). These reporter mice are used to study circadian expression in vivo as

142 the Period2 (mPer2) promoter drives luciferase (luc) expression. In all of our studies,

143 Zeitgeber time (ZT) 0 denotes the time of lights on in light/dark cycles (LD), whereas

144 circadian time (CT) 0, denotes start of subjective day and CT12 denotes start of

145 subjective night in constant darkness (DD).Food and water were provided ad libitum.

4

146 All animal care and procedures were performed in strict accordance with the

147 recommendations of the Guide for the Care and Uses of Laboratory Animals of the

148 National Institutes of Health and approved by the UT Southwestern Medical Center

149 Institutional Animal Care and Use Committee.

150

151 Circadian Behavioral Analyses.

152 Adult male mice (8-12 weeks, n > 12/group) were housed individually in running

153 wheel cages and their activity was recorded continuously as described previously

154 (Shimomura et al., 2001) using ClockLab hardware and software (Actimetrics, Wilmette,

155 IL; RRID:SCR_014309). Phase angle, activity bouts, and free-running period were

156 calculated using ClockLab. Activity onset (psi) was calculated during LD. Bout analyses

157 were calculated after approximately 8 weeks in DD, across 14 days of data, with bouts

158 defined by a 10 counts/min threshold and a 20min maximum gap. Free-running period

159 was calculated after approximately 8 weeks in DD, across 14 days of data using a χ2

160 periodogram. Animals whose data could not be scored for the full time for any measure

161 were excluded from analysis. For behavioral ethograms, mice (n=2/group) were housed

162 in LD and visualized with a Sony DCR-HC62 camcorder, recording 1 frame/minute.

163 Data were recorded over the active (dark) portion of the LD cycle, with recordings

164 extending into the morning hours (~ZT12-ZT3.5).

165

166 Bioluminescence Recordings of Circadian Rhythms.

167 To investigate circadian rhythms of clock in the SCN and

168 peripheral tissues of these mice, bioluminescence recordings were performed as

5

169 previously described (Yoo et al., 2004; Liu et al., 2007). Briefly, adult male Per2::luc

170 (WT and Mef2d-/-) mice (n=7/group) were euthanized by rapid cervical dislocation

171 between approximately ZT11-12 for LD experiments or CT11-12 for DD experiments.

172 Tissues were harvested and kept in chilled Hanks’ balanced salt solution. SCN was

173 taken from a 300mm coronal brain section sliced on a vibratome. A heart sample was

174 taken from the exsanguinated atrium. Kidney, liver, and lung were hand sliced into thin

175 samples. Anterior pituitary was cultured whole. Explanted tissues were placed on

176 Millicell culture inserts (MilliporeSigma, Burlington, MA; RRID: SCR_015799) in 35-mm

177 culture dishes in tissue culture medium containing luciferin (Promega, Madison, WI).

178 Cultures were incubated for at least 6 days under constant conditions (constant

179 darkness, constant temperature of 35 °C, and no medium change). The light emitted

180 from each individual culture dish was recorded in real-time with photomultiplier tube

181 detectors using LumiCycle hardware and software (Actimetrics, Wilmette, IL).

182 Bioluminescence data were detrended by subtracting the 24h running average from the

183 raw data. The detrended data sets were smoothed by taking 2h running averages. Free-

184 running period was calculated over 5 cycles with LumiCycle software using Levenberg-

185 Marquardt (LM) sin fit, and mean periods.

186

187 RNA Expression.

188 Tissue was collected from male mice (n=2/3 per group) housed in either LD or

189 DD. Mice were euthanized at the indicated times (2 timepoints for LD and 6 timepoints

190 for DD) by rapid cervical dislocation. RNA isolation and qRT-PCR were performed as

191 previously described (Siepka et al., 2007). Primers for qRT-PCR are shown in Table 1.

6

192 Relative quantification was calculated using the ∆∆ Ct method and Gapdh as

193 housekeeping gene.

194

195 Cell Culture and In Vitro Assays. 196 197 HEK293A cells were cultured in DMEM (#D5671; Sigma-Aldrich, St. Louis, MO)

198 supplemented with 10% fetal bovine serum (#F2442; Sigma-Aldrich, St. Louis, MO).

199 Cells were plated 1 day before transfection and Effectene reagent (#301425; Qiagen,

200 Germantown, MD) was used to transfect DNA according to the manufacturer's protocol.

201 Approximately 36hr after transfection, cells were harvested for downstream

202 applications. For the luminescence assays, cells were lysed, and luminescence was

203 measured from 20 μl of lysate with the Luciferase Assay System (#E1500; Promega,

204 Madison, WI) using a luminometer (AutoLumat Plus; Berthold Technologies, Oak Ridge,

205 TN). Luminescence is reported relative to Renilla (#E2810; Renilla Luciferase Assay,

206 Promega, Madison, WI). For the immunoprecipitation assay, cells were lysed, and

207 immunoprecipitation and Western blotting was performed as previously described

208 (Siepka et al., 2007), using anti-FLAG M2 agarose (#A2220; Sigma-Aldrich, St. Louis,

209 MO). Western blot was performed using anti-HA (Roche, Branchburg, NJ;

210 RRID:AB_2687407), anti-CLOCK (generously given b›Dr. Seung-Hee YooȌǡ anti-Flag

211 (Sigma-Aldrich, St. Louis, MO; RRID:AB_796202), and anti-α-tubulin (Santa Cruz,

212 Dallas, TX). All assays were performed in duplicate and, except for the BMAL::Luc

213 assay, were repeated in 2-3 independent transfection experiments.

214

215

7

216 Sleep Recording and Analyses.

217 Sleep recording was performed as previously described (Funato et al., 2010).

218 Briefly, four electrode pins (for EEG) and two flexible wires (for EMG recording) were

219 implanted under ketamine/xylazine anesthesia. EEG electrodes were positioned over

220 the frontal and occipital cortices [anteroposterior (AP): 0.5mm, mediolateral (ML):

221 1.3mm, dorsoventral (DV): -1.3mm; and AP: -4.5mm, ML: 1.3mm, DV: -1.3mm,

222 respectively]. For recordings, male mice (n=8/group) were housed individually in LD and

223 tethered to a counterbalance arm (#SMCLA; Instech Laboratories, Plymouth Meeting,

224 PA) allowing for free movement. All mice were allowed 14d of recovery prior to

225 recording. EEG/EMG signal was recorded continuously for two, consecutive 24h

226 periods. EEG/EMG signals were amplified using a Grass Model 78 (AstroNova, West

227 Warwick, RI), filtered, digitized at a sampling rate of 250Hz, and displayed using custom

228 polygraph software. The vigilance state in each 20 sec epoch was classified as NREM

229 sleep, REM sleep, or wakefulness by a custom, semi-automated sleep staging software

230 based in MATLAB (Mathworks, Natick, MA). For power density analyses, EEG power

231 density in each frequency bin was expressed as a percentage of the mean total EEG

232 power over all frequency bins and vigilance states.

233 234 Experimental Design and Statistical Analyses. 235 236 All statistical tests were performed using Graphpad Prism version 8.0 for for Mac

237 OS X (GraphPad Software, San Diego, CA; RRID:SCR_002798). For behavioral

238 experiments, sleep studies, and SCN explant experiments, means were calculated for

239 each group for each measure and compared with students t-tests (conventional KO) or

240 one-way ANOVA (brain-specific KO) to compare across genotypes. Realtime PCR data

8

241 were analyzed via two-way ANOVA with genotype and time as between and within

242 variables, respectively. In vitro assays were analyzed with one-way ANOVAs with

243 Bonferroni correction for multiple comparisons or unpaired t-tests with Welch’s

244 correction for differences in variance.

245 246 247 RESULTS 248 249 Mef2d knockout mice have altered circadian behavior. 250 251 Loss of functional MEF2D had a profound impact on behavioral circadian

252 rhythms (Fig. 1). Mef2d-/- (KO) mice housed with running-wheels in 12:12 light/dark

253 cycles with entraining T cycle 24h (T24, LD 12:12; hereafter referred to as LD) exhibited

254 a delayed phase of activity onset (t(29) = 3.762, *p=0.0008; t-test), beginning their

255 wheel-running approximately 25 min later than wild-type (WT) mice (Fig. 1a-c). KO

256 mice also had a substantially lengthened (40 min longer than WT) free-running period

257 (t(38) = 8.340, *p < 0.0001; t-test) in constant darkness (DD) (Fig. 1a-b, d). Notably,

258 running-wheel activity was fragmented in KO mice. KO mice displayed a greater

259 number of activity bouts per day (t(29) = 4.257, *p =0.0002; t-test), with the individual

260 activity bouts being of shorter duration (t(29) = 4.844, *p < 0.0001; t-test) as compared

261 to WT mice (Fig. 1a-b, e). These findings suggested that MEF2D could play a larger

262 role in the general control of behavior. Therefore, we performed an in-depth analysis of

263 the behavior of KO mice throughout the active portion of the 24h day. We compiled

264 ethograms from images of individual mice, which were recorded 1x/min throughout the

265 night. These analyses revealed that KO mice spent less consolidated time on the

266 running-wheel than WT mice, and wheel-running activity of KO mice was frequently

9

267 interrupted by intermittent rest events (Fig. 1f-h). No other gross behavioral alterations

268 were observed.

269 Neuronal Mef2D is Required for Normal Circadian Behavior 270 271 To further investigate whether loss of MEF2D in the brain was responsible for the

272 behavioral phenotype in the global Mef2d KO, we crossed Mef2a/c/d floxed mice with a

273 CamKIIa-iCre line to generate mice with postnatal brain-specific loss of MEF2 function.

274 Intercrossing for subsequent generations resulted in mice with loss of varying

275 combinations of Mef2 genes in the brain. Only postnatal loss of MEF2D in the brain led

276 to a change in circadian free-running period during DD (F(3, 59) = 25.19, *p < 0.0001;

277 one-way ANOVA), as CamKIIa-iCre x Mef2afx/fx or Mef2cfx/fx mice were indistinguishable

278 from WT controls. In contrast, CamKIIa-iCre x Mef2dfx/fx mice had a long circadian

279 period of running-wheel activity similar to what was observed for the conventional KO

280 (Fig. 2a-c). However, these mice did not differ from controls in their phase-response

281 curves to light or dark pulses (data not shown).

282

283 Mef2d knockout in the SCN causes light-dependent effects on free-running 284 period. 285 286 To test the effects of Mef2d on circadian rhythms in the SCN, we crossed Mef2d-

287 /- mice with mice carrying the Per2::luc reporter. When SCNs were harvested from

288 animals housed in LD, the KO SCNs displayed a delayed phase of PER2

289 bioluminescence from the acute culture (Fig. 3a), recapitulating our behavioral findings.

290 Curiously, PER2 rhythms in the SCN of these animals were approximately 50 min

291 shorter in the presence of a T24 entraining cycle (Fig. 3a-b; t(12) = 3.443, *p=0.0049; t-

292 test). PER2::LUC rhythms in other tissues from KO mice were largely unaffected, with

10

293 the exception of a lengthened period in lungs (Fig. 3c; t(17)=3.279, *p=0.0044; t-test).

294 The shorter free-running period in KO SCNs harvested from LD led us to hypothesize

295 that the SCNs of these mice may be exhibiting an after-effect of the LD cycle on the

296 master oscillator. To test this hypothesis, we harvested tissues from animals housed in

297 DD. Interestingly, despite the behavioral differences seen in DD (Fig. 1d), SCNs from

298 mice housed in DD had no observable difference in the rhythm of PER2::LUC (Fig. 3d-

299 e; t(6) = 0.598, p = 0.572; t-test). Likewise, no period difference was found in any of the

300 other tissues tested (Fig. 3f), and SCN amplitude did not differ between genotypes in

301 LD or DD (p > 0.05).

302 Therefore, to explore the impact of LD cycles on the PER2::LUC rhythms of

303 Mef2d-/- SCNs, we housed WT and KO mice in short (T22, LD11:11) and long (T26,

304 LD13:13) LD cycles. SCNs were harvested following at least three weeks of acclimation

305 to the altered cycle length. When analyzed by two-way ANOVA, neither a short (T22)

306 nor a long (T26) LD cycle produced a significant change in free-running period length

307 for either genotype (Fig. 3g-i); however, genotype effects were diminished under T26

308 conditions (Fig. 3i; F(1, 35) = 12.24, * p =0.0013 for T24; two-way ANOVA). Taken

309 together, the data from Mef2d KO SCNs suggests that the behavioral phenotype is not

310 a result of a gross change in Per2 expression in the SCN, but rather is a result of an

311 interaction between MEF2D and the photic environment.

312 313 Clock gene expression is normal in Mef2d knockout tissues. 314 315 To begin to elucidate the actions of MEF2D on core clock genes, we next

316 measured messenger RNA (mRNA) expression in cerebellum and heart samples from

317 WT and Mef2d KO mice housed in LD. There was no obvious effect of genotype on

11

318 clock gene expression in the cerebellum at two different time-points (Fig. 4a). However,

319 for Per1 mRNA expression there was an interaction between genotype and time-of-day,

320 with a less dramatic increase observed at ZT12 in the cerebellum of KO mice compared

321 to WT mice (interaction (F(1,7)= 8.502, p=0.0225) and main effect of time

322 (F(1,7)=21.19, p=0.0025); * p =0.0089; two-way ANOVA). In heart samples, only Rev-

323 erba expression differed between WT and KO mice, with KO mice having higher levels

324 of Rev-erba at ZT12 (Fig. 4b; interaction (F(1,7)=8.076, p=0.0250, main effect of time

325 (F(1,7)=43.10, p=0.0003, and main effect of genotype (F(1,7)=6.001, p=0.0441); *p =

326 0.0218; two-way ANOVA. Mef2d expression differed significantly with time-of-day in the

327 cerebellum where it was slightly higher at ZT12 than ZT0 (Fig. 4a; t(3)=3.296,

328 *p=0.0459; t-test). No differences were observed in clock gene expression or Mef2d

329 expression in either cerebellum or heart tissues from mice housed in constant darkness

330 (DD; data not shown).

331 Because such small differences in clock gene expression were observed, we

332 also tested for the ability of MEF2D to transcriptionally regulate clock genes in

333 fibroblasts using a luciferase reporter assay. MEF2D did not activate Per1, Per2, or

334 Bmal1 in our experiments (Fig. 5a-c), but did trans-activate a positive control (desMef2,

335 Fig. 5d; t(3.127)=8.472; *p=0.029; t-test). Further, we observed no physical interactions

336 between MEF2D and BMAL1 in co-immunoprecipitation experiments (Fig. 5e). Along

337 with our gene expression results, these findings suggest that the robust period

338 difference in the circadian behavior of Mef2d KO mice is not the result of a substantial,

339 direct impact on clock gene or protein expression.

340

12

341 Mef2d KO mice have disrupted sleep patterns. 342 343 Our findings of disturbed circadian running-wheel behavior and increased rest

344 events in Mef2d-/- mice, without corresponding broad changes in the molecular circadian

345 clock, led us to hypothesize these behavioral changes were reflective of abnormal

346 sleep. Thus, as a final behavioral analysis, we recorded sleep patterns from WT and KO

347 mice. Wake and sleep episodes were phase delayed in KO mice (Fig. 6a-c), mirroring

348 what was observed for running-wheel behavior (Fig. 1a-c). There were no differences in

349 the EEG power density across sleep stages or the total time spent in any sleep stage

350 (data not shown); however, KO mice tended to have more transitions (Fig. 6d) from

351 wakefulness to NREM sleep (t(14)= 2.077; p = 0.0567; t-test) and from NREM sleep to

352 wakefulness (t(14)= 2.015; p = 0.0635; t-test) or REM sleep (t(14)= 2.002; p = 0.0651; t-

353 test). More in-depth analysis revealed that KO mice had extremely fragmented sleep

354 with significantly shorter REM latency (Fig. 6e; (Full day: t(14)=3.147, *p=0.0071),

355 (Light: t(14)=2.848, **p=0.0129), (Dark: t(14)=3.108, ***p=0.0077); t-tests) as well as

356 shorter NREM episode duration (Fig. 6f; Full day: t(14)=2.829, *p=0.0134), (Light:

357 t(14)=2.557, **p=0.0228), (Dark: t(14)=2.910, ***p=0.0114); t-tests). Plotting the number

358 of NREM episodes as a function of episode duration (Fig. 6g) revealed that the majority

359 of NREM episodes were shorter in KO mice as compared to WT mice. Taken together,

360 these results suggest that the circadian alterations in activity in Mef2d-/- mice are due, at

361 least in part, to disrupted sleep patterns.

362

13

363 DISCUSSION 364 365 366 It is rare for a single-gene mutation/loss to cause a large change in the circadian

367 free-running period. When such changes do occur, they are typically the result of loss of

368 function for a gene directly involved in the core clock feedback loop or a gene that

369 strongly influences the expression or stability of core clock genes (Takahashi et al.,

370 2008). Here we demonstrate that loss of Mef2d results in a 40 min lengthening of the

371 free-running period of locomotor activity. This change in free-running period is not

372 associated with concomitant changes in the levels of core clock components, and thus

373 appears to result from a role of Mef2d downstream of the molecular clock.

374 In this study, we observed a lengthening of period both in conventional Mef2d-/-

375 and CamKIIa-iCre x Mef2dfx/fx mice, therefore it is likely that the loss of Mef2d impacts

376 the circadian system through its actions in the brain. The CamKIIa-iCre line used

377 expresses at postnatal day 3, with maximal expression occurring by postnatal day 15

378 (Casanova et al., 2001), thus our results suggest that the effects of Mef2d on the

379 circadian system are not due to developmental loss of Mef2d. The floxed Mef2d allele

380 does not cause a complete knockout, but rather it produces a non-functional truncated

381 protein product (Kim et al., 2008). However; these same mice have been crossed with

382 a different Cre-driver line shown to have reduced Mef2d gene expression in the

383 hippocampus (Akhtar et al., 2012). Importantly, the long free-running period observed in

384 Mef2d-/- mice is specific, as we did not observe a change in any circadian parameter in

385 brain-specific Mef2a or Mef2c floxed mice.

386 MEF2D has been shown to influence synapse elimination, dendritic spine

387 formation, and the expression of neurotrophic factors in hippocampal neurons (Lyons et

14

388 al., 1995; Akhtar et al., 2012; Dietrich, 2013; Rashid et al., 2014; Chang et al., 2017), so

389 it is possible that the postnatal loss of MEF2D causes reorganization of SCN circuitry.

390 While there is some evidence that alterations in synaptic connectivity lead to changes in

391 circadian outputs in the fly (Fernandez et al., 2008; Gilestro et al., 2009; Gorostiza et al.,

392 2014; Krzeptowski et al., 2018), it remains to be established whether this is the case in

393 mammals. Previous studies have concluded that MEF2 in the fly may be necessary for

394 a daily pattern of dendritic remodeling via inhibition of the cell-adhesion molecule

395 Fasciclin2 (Fas2) (Sivachenko et al., 2013), so it is possible that MEF2 may exert its

396 actions through similar mechanisms in mice.

397 Interestingly, while the behavioral free-running period was lengthened in Mef2d-/-

398 mice, SCNs from animals previously entrained to LD cycles had shorter free-running

399 periods than WT mice. This effect was not observed when SCNs were harvested from

400 mice held in constant DD conditions, suggesting that the short period SCN in Mef2d-/-

401 mice is an after-effect of the LD cycle and not due to an intrinsic change in the period of

402 the SCN itself. After-effects of LD cycles on SCN rhythms have been reported

403 previously, although they are not well-understood (Aton et al., 2004; Mickman et al.,

404 2008; Molyneux et al., 2008). When SCNs were taken from mice housed in LD cycles

405 shorter than 24h, (T22; LD11:11), the periods of KO mice remained shorter than those

406 of WT mice, although this difference did not reach statistical significance. It should be

407 noted, however, that in this short LD cycle, the phase of locomotor activity onset was

408 very variable for both genotypes, suggesting that animals may not have been fully

409 entrained in this condition. It has been proposed that normal MEF2 levels may be

410 necessary to maintain synchrony between clock neurons in the fly brain (Blanchard et

15

411 al., 2010; Sivachenko et al., 2013). In mice, however, we did not observe any evidence

412 via gene expression data that the SCN is strongly affected by the loss of MEF2D. Based

413 on our data, we suggest that the long period of locomotor activity we observe in Mef2d-/-

414 mice may be due to their being pushed to conform to a 24-hour LD cycle, and could be

415 the result of an effect on SCN output, rather than an effect on timekeeping within the

416 SCN itself. While we did not directly assess the SCN anatomy or neuronal activity in

417 Mef2d-/- mice, nor did we measure core SCN-driven physiological outputs, these would

418 be important to measure in future studies to test this hypothesis.

419 Alternatively, it is possible that rather than affecting SCN outputs, the loss of

420 MEF2D alters SCN inputs from the retina. MEF2D binds to retinal-specific promoters to

421 drive photoreceptor development, and Mef2d-/- mice have progressive retinal

422 degeneration beginning at P30 (Andzelm et al., 2015; Omori et al., 2015). While these

423 studies examined gross changes to rods and cones, intrinsically photosensitive retinal

424 ganglion cells could also be disrupted. Changes in behavioral circadian rhythms have

425 been reported with retinal degeneration or bilateral enucleation (Lupi et al., 1999;

426 Yamazaki et al., 2002; Tosini et al., 2007), therefore it is possible that the Mef2d-/- mice

427 have altered behavioral rhythms due to decreased retinal signaling. Future studies

428 could examine light-induction in the SCN and light-induced phase shifts in behavior in

429 these mice. In addition, while we did not examine aging mice, it would be interesting for

430 future studies to examine the circadian phenotype over extended periods of time to

431 determine whether the progressive retinal degeneration seen in these mice correlates

432 with behavioral changes.

16

433 In addition to the circadian effects, loss of MEF2D had a profound effect on

434 sleep/wake patterning. Both activity and sleep were extremely fragmented in Mef2d-/-

435 mice. NREM sleep in particular was largely restricted to multiple, very short duration

436 bouts in KOs. This difference in the patterning of NREM sleep occurred without a

437 corresponding change in either total time spent in NREM sleep across the day or power

438 density during any sleep stage. While we did not measure melatonin levels or other

439 physiological markers of sleep patterning, future studies should examine the larger

440 implications of this disrupted sleep pattern on behavioral and physiological function in

441 these animals. In particular, it would be fascinating to determine whether loss of

442 MEF2D influences memory consolidation during sleep. The MEF2 family of

443 transcription factors have been implicated in memory formation and synaptic plasticity

444 (Barbosa et al., 2008; Akhtar et al., 2012; Dietrich, 2013; Rashid et al., 2014; Adachi et

445 al., 2016). Further, a growing body of literature suggests that MEF2 proteins exert

446 neuroprotective effects in response to neuronal inflammation and oxidative stress (Gong

447 et al., 2003; Tang et al., 2005; Smith et al., 2006; She et al., 2011; Ryan et al., 2013;

448 Yang et al., 2015). The present study provides additional evidence for a specific role of

449 MEF2D in maintaining normal function of the mammalian circadian clock and sleep

450 homeostasis. Future studies should focus on understanding the influence of MEF2 on

451 sleep and circadian function and its impacts on neuronal health.

452 453

17

454 REFERENCES 455 456 †ƒ Š‹ǡ‹ǡ”ƒƒ˜ ǡ‘–‡‰‰‹ƒȋʹͲͳ͸Ȍ‘•–ƒ–ƒŽ‘••‘ˆ‡ˆʹ ‡•—Ž–•‹ 457 ‹••‘ ‹ƒ–‹‘‘ˆˆˆ‡ –•‘›ƒ’•‡—„‡”ƒ†‡ƒ”‹‰ƒ†‡‘”›Ǥ‹‘Ž 458 •› Š‹ƒ–”›ͺͲǣͳͶͲǦͳͶͺǤ 459 Š–ƒ”ǡ‹ǡ†ƒ Š‹ǡ‘””‹• ǡ‹ǡ‹ Šƒ”†•‘ ǡƒ••‡ŽǦ—„›ǡŽ•‘ǡ 460 ƒ˜ƒŽƒŽ‹ǡ‘–‡‰‰‹ƒȋʹͲͳʹȌ ˜‹˜‘ƒƒŽ›•‹•‘ˆ ʹ–”ƒ• ”‹’–‹‘ˆƒ –‘”•‹ 461 •›ƒ’•‡”‡‰—Žƒ–‹‘ƒ†‡—”‘ƒŽ•—”˜‹˜ƒŽǤ‘‡͹ǣ‡͵Ͷͺ͸͵Ǥ 462 Ž„”‡ Š–ȋʹͲͳʹȌ‹‹‰–‘’‡”ˆ‡ –‹‘ǣ–Š‡„‹‘Ž‘‰›‘ˆ ‡–”ƒŽƒ†’‡”‹’Š‡”ƒŽ ‹” ƒ†‹ƒ 463 Ž‘ •Ǥ‡—”‘͹ͶǣʹͶ͸Ǧʹ͸ͲǤ 464 †œ‡ŽǡŠ‡””› ǡ ƒ”‹ǡ‘‡‡ǡ‡‡ǡ ‡„‡”‰ǡƒ™Ž›ǡƒŽ‹ǡ 465 Žƒ˜‡ŽŽǡƒ†„‡”‰ǡƒ˜‹‘Žƒǡ ”‡‡„‡”‰ȋʹͲͳͷȌ ʹ†”‹˜‡• 466 ’Š‘–‘”‡ ‡’–‘”†‡˜‡Ž‘’‡––Š”‘—‰Šƒ‰‡‘‡Ǧ™‹†‡ ‘’‡–‹–‹‘ˆ‘”–‹••—‡Ǧ•’‡ ‹ˆ‹  467 ‡Šƒ ‡”•Ǥ‡—”‘ͺ͸ǣʹͶ͹Ǧʹ͸͵Ǥ 468 ”‘Ž†ǡ‹ǡœ—„”›–ǡŠƒǡ ƒŽŽ› ǡ‹ǡŠ‡Ž–‘ ǡ‹ Šƒ”†•‘ ǡƒ••‡ŽǦ 469 —„›ǡŽ•‘ȋʹͲͲ͹Ȍ ʹ–”ƒ• ”‹’–‹‘ˆƒ –‘” ‘–”‘Ž• Š‘†”‘ ›–‡ 470 Š›’‡”–”‘’Š›ƒ†„‘‡†‡˜‡Ž‘’‡–Ǥ‡˜‡ŽŽͳʹǣ͵͹͹Ǧ͵ͺͻǤ 471 –‘ ǡŽ‘  ǡ‡‹ ǡƒƒœƒ‹ǡ ‡”œ‘‰ȋʹͲͲͶȌŽƒ•–‹ ‹–›‘ˆ ‹” ƒ†‹ƒ„‡Šƒ˜‹‘”ƒ† 472 –Š‡•—’”ƒ Š‹ƒ•ƒ–‹ — Ž‡—•ˆ‘ŽŽ‘™‹‰‡š’‘•—”‡–‘‘ǦʹͶǦŠ‘—”Ž‹‰Š– › Ž‡•Ǥ ‹‘Ž 473 Š›–Š•ͳͻǣͳͻͺǦʹͲ͹Ǥ 474 ƒ”„‘•ƒǡ‹ǡ”–— ǡ†ƒ Š‹ǡ‡Ž•‘ǡ ƒŽŽ› ǡ‹ Šƒ”†•‘ ǡƒ˜ƒŽƒŽ‹ǡ 475 ‘–‡‰‰‹ƒǡƒ••‡ŽǦ—„›ǡŽ•‘ȋʹͲͲͺȌ ʹǡƒ–”ƒ• ”‹’–‹‘ˆƒ –‘”–Šƒ– 476 ˆƒ ‹Ž‹–ƒ–‡•Ž‡ƒ”‹‰ƒ†‡‘”›„›‡‰ƒ–‹˜‡”‡‰—Žƒ–‹‘‘ˆ•›ƒ’•‡—„‡”•ƒ† 477 ˆ— –‹‘Ǥ”‘ ƒ–Ž ƒ† ‹ͳͲͷǣͻ͵ͻͳǦͻ͵ͻ͸Ǥ 478 Žƒ ǡŽ•‘ȋͳͻͻͺȌ”ƒ• ”‹’–‹‘ƒŽ ‘–”‘Ž‘ˆ—• Ž‡†‡˜‡Ž‘’‡–„››‘ ›–‡ 479 ‡Šƒ ‡”ˆƒ –‘”Ǧʹȋ ʹȌ’”‘–‡‹•Ǥ—‡˜‡ŽŽ‡˜‹‘ŽͳͶǣͳ͸͹Ǧͳͻ͸Ǥ 480 Žƒ Šƒ”† ǡ‘ŽŽ‹•ǡ›”ƒǡ ƒ ‘  ǡƒ›Ž‘”ǡŽƒ— ȋʹͲͳͲȌŠ‡–”ƒ• ”‹’–‹‘ 481 ˆƒ –‘”‡ˆʹ‹•”‡“—‹”‡†ˆ‘”‘”ƒŽ ‹” ƒ†‹ƒ„‡Šƒ˜‹‘”‹”‘•‘’Š‹ŽƒǤ ‡—”‘• ‹ 482 ͵ͲǣͷͺͷͷǦͷͺ͸ͷǤ 483 ƒ•ƒ‘˜ƒǡ ‡Š•‡ˆ‡Ž†ǡƒ–ƒƒ†‹‘–‹•ǡ‡„‡”‰‡”ǡ ”‡‹‡”ǡ–‡™ƒ”– ǡ Š—–œ  484 ȋʹͲͲͳȌƒ ƒŽ’Šƒ‹”‡ƒŽŽ‘™•„”ƒ‹Ǧ•’‡ ‹ˆ‹ ‰‡‡‹ƒ –‹˜ƒ–‹‘Ǥ ‡‡•‹• 485 ͵ͳǣ͵͹ǦͶʹǤ 486 Šƒ‰ǡ‹Ž‡”•‘ ǡ ƒŽ‡ ǡ ‹„•‘ ǡ —„‡”ȋʹͲͳ͹Ȍ‹•–‹ –•–ƒ‰‡•‘ˆ•›ƒ’•‡ 487 ‡Ž‹‹ƒ–‹‘ƒ”‡‹†— ‡†„›„—”•–ˆ‹”‹‰‘ˆͳ‡—”‘•ƒ††‹ˆˆ‡”‡–‹ƒŽŽ›”‡“—‹”‡ 488  ʹȀǤŽ‹ˆ‡͸Ǥ 489 ‘Ž‡ ǡ‡” ƒŽ†‘ǡ‡•–‹˜‘ǡ‹—ǡ‡‡”‡• ǡ ƒ ǡ‡–‡”‡ ǡ‡ƒ”ǡ‘•• ǡ‡˜‡ 490 ǡ ”ƒŽƒ†ǡ ‘••‡Ž›ȋʹͲͳʹȌ ʹ‡‰ƒ–‹˜‡Ž›”‡‰—Žƒ–‡•Ž‡ƒ”‹‰Ǧ‹†— ‡† 491 •–”— –—”ƒŽ’Žƒ•–‹ ‹–›ƒ†‡‘”›ˆ‘”ƒ–‹‘Ǥƒ–‡—”‘• ‹ͳͷǣͳʹͷͷǦͳʹ͸ͶǤ 492 ‹‡–”‹ Š ȋʹͲͳ͵ȌŠ‡ ʹˆƒ‹Ž›ƒ†–Š‡„”ƒ‹ǣˆ”‘‘Ž‡ —Ž‡•–‘‡‘”›Ǥ‡ŽŽ‹••—‡ 493 ‡•͵ͷʹǣͳ͹ͻǦͳͻͲǤ 494 ‡”ƒ†‡œǡ‡”‹ ǡ‡”‹ƒ‹ ȋʹͲͲͺȌ‹” ƒ†‹ƒ”‡‘†‡Ž‹‰‘ˆ‡—”‘ƒŽ ‹” —‹–• 495 ‹˜‘Ž˜‡†‹”Š›–Š‹ „‡Šƒ˜‹‘”Ǥ‘‹‘Ž͸ǣ‡͸ͻǤ 496 Žƒ˜‡ŽŽǡ‹ǡ ”ƒ› ǡ ƒ”‹ǡ ‡„‡”‰ǡ ‘‰ ǡƒ”‡• ‘ˆˆǦƒ’ƒ†‹‹–”‹‘— 497 ǡ‡ƒ”ǡ ”‡‡„‡”‰ȋʹͲͲͺȌ ‡‘‡Ǧ™‹†‡ƒƒŽ›•‹•‘ˆ ʹ–”ƒ• ”‹’–‹‘ƒŽ 498 ’”‘‰”ƒ”‡˜‡ƒŽ••›ƒ’–‹ –ƒ”‰‡–‰‡‡•ƒ†‡—”‘ƒŽƒ –‹˜‹–›Ǧ†‡’‡†‡– 499 ’‘Ž›ƒ†‡›Žƒ–‹‘•‹–‡•‡Ž‡ –‹‘Ǥ‡—”‘͸ͲǣͳͲʹʹǦͳͲ͵ͺǤ

18

500 Žƒ˜‡ŽŽǡ‘™ƒǡ‹ǡ ”‡‡”ǡ‹ǡƒ”ƒ†‹•ǡ ”‹ˆˆ‹–Šǡ —ǡŠ‡ǡ 501 ”‡‡„‡”‰ȋʹͲͲ͸Ȍ –‹˜‹–›Ǧ†‡’‡†‡–”‡‰—Žƒ–‹‘‘ˆ ʹ–”ƒ• ”‹’–‹‘ˆƒ –‘”• 502 •—’’”‡••‡•‡š ‹–ƒ–‘”›•›ƒ’•‡—„‡”Ǥ ‹‡ ‡͵ͳͳǣͳͲͲͺǦͳͲͳʹǤ 503 —ƒ–‘ ǡƒ–‘ǡ‹–‘ǡ ƒ—–”‘ǡ‹ŽŽ‹ƒ•ǡƒ ŠǡŽ“—‹•– ǡ‘—Ž– Š‹ ǡ 504 ƒƒ‰‹•ƒ™ƒȋʹͲͳͲȌ‘••‘ˆ ‘‘•‡ ‘‹†ǦŽ‹‡ƒ†‹ ‡‘”‰‡•›†”‘‡ ”‹–‹ ƒŽ”‡‰‹‘ 505 ͳͶ‹‹–‡”’‡†— —Žƒ”— Ž‡—•”‡•—Ž–•‹ƒŽ–‡”‡†”‡‰—Žƒ–‹‘‘ˆ”ƒ’‹†‡›‡‘˜‡‡– 506 •Ž‡‡’Ǥ”‘ ƒ–Ž ƒ† ‹ͳͲ͹ǣͳͺͳͷͷǦͳͺͳ͸ͲǤ 507 ‹Ž‡•–”‘ ǡ‘‘‹ ǡ‹”‡ŽŽ‹ȋʹͲͲͻȌ‹†‡•’”‡ƒ† Šƒ‰‡•‹•›ƒ’–‹ ƒ”‡”•ƒ•ƒ 508 ˆ— –‹‘‘ˆ•Ž‡‡’ƒ†™ƒ‡ˆ—Ž‡••‹”‘•‘’Š‹ŽƒǤ ‹‡ ‡͵ʹͶǣͳͲͻǦͳͳʹǤ 509 ‘‰ǡƒ‰ǡ‹‡†ƒǡƒ‰ǡ‡‰ ǡŠ‡‰ǡŽƒ‹”ǡƒ”•ŠƒŽŽ ǡƒ‘ȋʹͲͲ͵Ȍ 510 †ͷǦ‡†‹ƒ–‡†‹Š‹„‹–‹‘‘ˆ–Š‡’”‘–‡ –‹˜‡‡ˆˆ‡ –•‘ˆ–”ƒ• ”‹’–‹‘ˆƒ –‘” ʹ‹ 511 ‡—”‘–‘š‹ ‹–›Ǧ‹†— ‡†ƒ’‘’–‘•‹•Ǥ‡—”‘͵ͺǣ͵͵ǦͶ͸Ǥ 512 ‘”‘•–‹œƒǡ‡’‡–”‹•ǦŠƒ—˜‹ǡ ”‡‡Žǡ‹”‡œǡ‡”‹ƒ‹ ȋʹͲͳͶȌ‹” ƒ†‹ƒ 513 ’ƒ ‡ƒ‡”‡—”‘• Šƒ‰‡•›ƒ’–‹  ‘–ƒ –•ƒ ”‘••–Š‡†ƒ›Ǥ—””‹‘ŽʹͶǣʹͳ͸ͳǦ 514 ʹͳ͸͹Ǥ 515 ‡”œ‘‰ǡ ‡”ƒ•–›‡ǡ›ŽŽ‹‡ ǡ ƒ•–‹‰• ȋʹͲͳ͹Ȍ‡‰—Žƒ–‹‰–Š‡ 516 —’”ƒ Š‹ƒ•ƒ–‹ — Ž‡—•ȋȌ‹” ƒ†‹ƒŽ‘ ™‘”ǣ –‡”’Žƒ›„‡–™‡‡‡ŽŽǦ 517 —–‘‘‘—•ƒ†‹” —‹–Ǧ‡˜‡Ž‡ Šƒ‹••Ǥ‘Ž†’”‹‰ ƒ”„‡”•’‡ –‹‘ŽͻǤ 518 ‡•Š‹ƒ ǡ ƒ‹ǡŠ‹‘†ƒǡ ƒ–ƒ ǡƒƒ‘ȋͳͻͻͷȌš’”‡••‹‘‘ˆƒ„‘š‰‡‡ǡ 519  ʹǡ‹‡—”‘•‘ˆ–Š‡‘—•‡ ‡–”ƒŽ‡”˜‘—••›•–‡ǣ‹’Ž‹ ƒ–‹‘‘ˆ‹–•„‹ƒ”› 520 ˆ— –‹‘‹›‘‰‡‹ ƒ†‡—”‘‰‡‹  ‡ŽŽŽ‹‡ƒ‰‡•Ǥ‡—”‘• ‹‡––ʹͲͲǣͳͳ͹ǦͳʹͲǤ 521 ‹ǡŠƒǡ˜ƒ‘‘‹Œǡƒ‰ǡ ƒŽŽ› ǡ‹ǡ‹ Šƒ”†•‘ ǡ ‹ŽŽ ǡƒ••‡ŽǦ—„›ǡ 522 Ž•‘ȋʹͲͲͺȌŠ‡ ʹ–”ƒ• ”‹’–‹‘ˆƒ –‘”‡†‹ƒ–‡••–”‡••Ǧ†‡’‡†‡– 523 ƒ”†‹ƒ ”‡‘†‡Ž‹‰‹‹ ‡Ǥ Ž‹ ˜‡•–ͳͳͺǣͳʹͶǦͳ͵ʹǤ 524 ”œ‡’–‘™•‹ǡ ‡•• ǡ›œƒȋʹͲͳͺȌ‹” ƒ†‹ƒŽƒ•–‹ ‹–›‹–Š‡”ƒ‹‘ˆ •‡ –•ƒ† 525 ‘†‡–•Ǥ ”‘–‡—”ƒŽ‹” —‹–•ͳʹǣ͵ʹǤ 526 ‡‹ˆ‡”ǡ”ƒ‹ ǡ—ǡ ‡”‘–– ǡ”‡‹–„ƒ”–ǡ ‡‰ ǡ‡˜‡ǡ‘•‘ˆ•›ǡƒ†ƒŽǦ 527 ‹ƒ”†ǡ‹’–‘ȋͳͻͻ͵Ȍ ʹǡƒȀ ʹǦˆƒ‹Ž›–”ƒ• ”‹’–‹‘ˆƒ –‘” 528 ‡š’”‡••‡†‹ƒŽƒ‹ƒ”†‹•–”‹„—–‹‘‹ ‡”‡„”ƒŽ ‘”–‡šǤ”‘ ƒ–Ž ƒ† ‹ 529 ͻͲǣͳͷͶ͸ǦͳͷͷͲǤ 530 ‡‹‡–ƒŽǤȋʹͲͲ͹Ȍ ‡‘‡Ǧ™‹†‡ƒ–Žƒ•‘ˆ‰‡‡‡š’”‡••‹‘‹–Š‡ƒ†—Ž–‘—•‡„”ƒ‹Ǥ 531 ƒ–—”‡ͶͶͷǣͳ͸ͺǦͳ͹͸Ǥ 532 ‹ǡ‹•‡ƒǡŽŽ‡ǡ‡‹–œ‡”ǡƒ‰ǡƒ‡••‹‰ǡ‹‡”ƒǡ ‡‹†‡”‡‹ Š 533 ȋʹͲͲͳȌ›‘ ›–‡‡Šƒ ‡”ˆƒ –‘”ʹƒ†ʹ—†‡”‰‘’Š‘•’Š‘”›Žƒ–‹‘ƒ† 534 ƒ•’ƒ•‡Ǧ‡†‹ƒ–‡††‡‰”ƒ†ƒ–‹‘†—”‹‰ƒ’‘’–‘•‹•‘ˆ”ƒ– ‡”‡„‡ŽŽƒ”‰”ƒ—Ž‡‡—”‘•Ǥ  535 ‡—”‘• ‹ʹͳǣ͸ͷͶͶǦ͸ͷͷʹǤ 536 ‹—ǡ‡Ž•Šǡ‘ ǡ”ƒ ǡŠƒ‰ǡ”‹‡•–ǡ—Š”ǡ‹‰‡”ǡ‡‡‡”ǡ 537 ‡”ƒ ǡ‘›Ž‡ ǡ͵”†ǡƒƒŠƒ•Š‹ ǡƒ›ȋʹͲͲ͹Ȍ –‡” ‡ŽŽ—Žƒ” ‘—’Ž‹‰ ‘ˆ‡”• 538 ”‘„—•–‡••ƒ‰ƒ‹•–—–ƒ–‹‘•‹–Š‡ ‹” ƒ†‹ƒ Ž‘ ‡–™‘”Ǥ‡ŽŽͳʹͻǣ͸ͲͷǦ͸ͳ͸Ǥ 539 ‹—ǡƒ˜ƒƒ—‰Š ǡƒ‰ǡƒƒ‰ƒ‹ ǡƒ‘ǡ‹ƒȋʹͲͲ͵Ȍͷƒ –‹˜ƒ–‹‘‘ˆ ʹǦ 540 ‡†‹ƒ–‡†‰‡‡‡š’”‡••‹‘’Žƒ›•ƒ ”‹–‹ ƒŽ”‘Ž‡‹ Ǧ’”‘‘–‡†•—”˜‹˜ƒŽ‘ˆ 541 †‡˜‡Ž‘’‹‰„—–‘–ƒ–—”‡ ‘”–‹ ƒŽ‡—”‘•Ǥ”‘ ƒ–Ž ƒ† ‹ͳͲͲǣͺͷ͵ʹǦ 542 ͺͷ͵͹Ǥ 543 ‹—ǡ‡Ž•‘ǡ‡œ’”‘œ˜ƒƒ›ƒǡŠ‡Ž–‘ ǡ‹ Šƒ”†•‘ ǡƒ••‡ŽǦ—„›ǡŽ•‘ 544 ȋʹͲͳͶȌ‡“—‹”‡‡–‘ˆ ʹǡǡƒ†ˆ‘”•‡Ž‡–ƒŽ—• Ž‡”‡‰‡‡”ƒ–‹‘Ǥ”‘ ƒ–Ž 545  ƒ† ‹ͳͳͳǣͶͳͲͻǦͶͳͳͶǤ

19

546 —’‹ǡ‘‘’‡” ǡ ”‘‡ŠŽ‹ Šǡ–ƒ†ˆ‘”†ǡ ƒŽŽǡ ‘•–‡” ȋͳͻͻͻȌ”ƒ•‰‡‹  547 ƒ„Žƒ–‹‘‘ˆ”‘†’Š‘–‘”‡ ‡’–‘”•ƒŽ–‡”•–Š‡ ‹” ƒ†‹ƒ’Š‡‘–›’‡‘ˆ‹ ‡Ǥ‡—”‘• ‹‡ ‡ 548 ͺͻǣ͵͸͵Ǧ͵͹ͶǤ 549 ›‘• ǡ‹ ƒŽ‡•ǡ Š™ƒ”œ ǡƒ”–‹ ǡŽ•‘ȋͳͻͻͷȌš’”‡••‹‘‘ˆ‡ˆʹ‰‡‡•‹ 550 –Š‡‘—•‡ ‡–”ƒŽ‡”˜‘—••›•–‡•—‰‰‡•–•ƒ”‘Ž‡‹‡—”‘ƒŽƒ–—”ƒ–‹‘Ǥ  551 ‡—”‘• ‹ͳͷǣͷ͹ʹ͹Ǧͷ͹͵ͺǤ 552 ƒ‘ǡ‘‹ǡ‹ƒ ǡƒ†ƒŽǦ‹ ‡•ǡ ”‡‡„‡”‰ȋͳͻͻͻȌ‡—”‘ƒŽƒ –‹˜‹–›Ǧ†‡’‡†‡– 553 ‡ŽŽ•—”˜‹˜ƒŽ‡†‹ƒ–‡†„›–”ƒ• ”‹’–‹‘ˆƒ –‘” ʹǤ ‹‡ ‡ʹͺ͸ǣ͹ͺͷǦ͹ͻͲǤ 554 ‹ ƒǡ–—„„Ž‡ˆ‹‡Ž† ǡ ƒ””‹‰–‘ǡ‡Ž•‘ȋʹͲͲͺȌŠ‘–‘’‡”‹‘†ƒŽ–‡”•’Šƒ•‡ 555 †‹ˆˆ‡”‡ ‡„‡–™‡‡ƒ –‹˜‹–›‘•‡–‹˜‹˜‘ƒ†‡”ʹǣǣŽ— ’‡ƒ‹˜‹–”‘Ǥ Š›•‹‘Ž 556 ‡‰—Ž –‡‰”‘’Š›•‹‘Žʹͻͷǣͳ͸ͺͺǦͳ͸ͻͶǤ 557 ‘Ž›‡—šǡƒŠŽ‰”‡ǡ ƒ””‹‰–‘ȋʹͲͲͺȌ‹” ƒ†‹ƒ‡–”ƒ‹‡–ƒˆ–‡”‡ˆˆ‡ –•‹ 558 •—’”ƒ Š‹ƒ•ƒ–‹ — Ž‡‹ƒ†’‡”‹’Š‡”ƒŽ–‹••—‡•‹˜‹–”‘Ǥ”ƒ‹‡•ͳʹʹͺǣͳʹ͹Ǧͳ͵ͶǤ 559 ‘”‹ǡ‹–ƒ—”ƒǡ‘•Š‹†ƒǡ—™ƒŠƒ”ƒǡŠƒ›ƒǡ ”‹‡ǡ —”—ƒ™ƒȋʹͲͳͷȌ‡ˆʹ†‹• 560 ‡••‡–‹ƒŽˆ‘”–Š‡ƒ–—”ƒ–‹‘ƒ†‹–‡‰”‹–›‘ˆ”‡–‹ƒŽ’Š‘–‘”‡ ‡’–‘”ƒ†„‹’‘Žƒ” ‡ŽŽ•Ǥ 561 ‡‡•‡ŽŽ•ʹͲǣͶͲͺǦͶʹ͸Ǥ 562 ˆ‡‹ˆˆ‡”ǡƒ‰ǡ‹Ž‡”•‘ ǡƒ‹‰— Š‹ǡƒ•‹‘˜ƒǡ‹–Šǡ‘™ƒǡ 563 —„‡”ȋʹͲͳͲȌ ”ƒ‰‹Ž‡‡–ƒŽ”‡–ƒ”†ƒ–‹‘’”‘–‡‹‹•”‡“—‹”‡†ˆ‘”•›ƒ’•‡ 564 ‡Ž‹‹ƒ–‹‘„›–Š‡ƒ –‹˜‹–›Ǧ†‡’‡†‡––”ƒ• ”‹’–‹‘ˆƒ –‘” ʹǤ‡—”‘͸͸ǣͳͻͳǦ 565 ͳͻ͹Ǥ 566 ‘––Š‘ˆˆ ǡŽ•‘ȋʹͲͲ͹Ȍ ʹǣƒ ‡–”ƒŽ”‡‰—Žƒ–‘”‘ˆ†‹˜‡”•‡†‡˜‡Ž‘’‡–ƒŽ 567 ’”‘‰”ƒ•Ǥ‡˜‡Ž‘’‡–ͳ͵ͶǣͶͳ͵ͳǦͶͳͶͲǤ 568 ƒ•Š‹† ǡ‘Ž‡ ǡ ‘••‡Ž›ȋʹͲͳͶȌ‡”‰‹‰”‘Ž‡•ˆ‘” ʹ–”ƒ• ”‹’–‹‘ˆƒ –‘”•‹ 569 ‡‘”›Ǥ ‡‡•”ƒ‹‡Šƒ˜ͳ͵ǣͳͳͺǦͳʹͷǤ 570 ›ƒ‡–ƒŽǤȋʹͲͳ͵Ȍ •‘‰‡‹ Š—ƒ‹ƒ”‹•‘̵•‘†‡Ž•Š‘™•‹–”‘•ƒ–‹˜‡•–”‡••Ǧ 571 ‹†— ‡††›•ˆ— –‹‘‹ ʹǦ ͳƒŽ’Šƒ–”ƒ• ”‹’–‹‘Ǥ‡ŽŽͳͷͷǣͳ͵ͷͳǦͳ͵͸ͶǤ 572 ŠƒŽ‹œ‹ǡ‘‹ȋʹͲͲͷȌ„”ƒ™ˆ‘”„”ƒ‹•ǣ–Š‡”‘Ž‡‘ˆ ʹ’”‘–‡‹•‹–Š‡†‡˜‡Ž‘’‹‰ 573 ‡”˜‘—••›•–‡Ǥ—””‘’‡˜‹‘Ž͸ͻǣʹ͵ͻǦʹ͸͸Ǥ 574 Š‡ ǡƒ‰ǡŠ‡’Š‡”†ǡ‹–Šǡ‹ŽŽ‡” ǡ‡•–ƒǡƒ‘ȋʹͲͳͳȌ‹”‡ –”‡‰—Žƒ–‹‘‘ˆ 575 ‘’Ž‡š „›‹–‘ Š‘†”‹ƒŽ ʹ‹•†‹•”—’–‡†‹ƒ‘—•‡‘†‡Ž‘ˆƒ”‹•‘ 576 †‹•‡ƒ•‡ƒ†‹Š—ƒ’ƒ–‹‡–•Ǥ Ž‹ ˜‡•–ͳʹͳǣͻ͵ͲǦͻͶͲǤ 577 Š‡ƒ”ƒǡ”‹”ƒǡ‡ƒ˜‡”ǡƒ›™‘‘†ǡŠƒ˜‡• ǡŠ‡‰ǡ—‡ǡ‡‡ǡ˜ƒ 578 †‡” ‘”•– ǡ ƒ•–‹‰• ǡ‡’’‡”–ȋʹͲͲͲȌ –‡”ƒ –‹‰‘Ž‡ —Žƒ”Ž‘‘’•‹–Š‡ 579 ƒƒŽ‹ƒ ‹” ƒ†‹ƒ Ž‘ Ǥ ‹‡ ‡ʹͺͺǣͳͲͳ͵ǦͳͲͳͻǤ 580 Š‹‘—”ƒǡ‘™Ǧ‡††‹‡•ǡ‹‰ǡ–‡‡˜‡•ǡŠ‹–‡Ž‡›ǡ—•ŠŽƒ ǡ‡‡‹†‡•ǡ 581 ‹ǡ‹–ƒ–‡”ƒ ǡŠ—” Š‹ŽŽ ǡƒƒŠƒ•Š‹ ȋʹͲͲͳȌ ‡‘‡Ǧ™‹†‡‡’‹•–ƒ–‹  582 ‹–‡”ƒ –‹‘ƒƒŽ›•‹•”‡˜‡ƒŽ• ‘’Ž‡š‰‡‡–‹ †‡–‡”‹ƒ–•‘ˆ ‹” ƒ†‹ƒ„‡Šƒ˜‹‘”‹ 583 ‹ ‡Ǥ ‡‘‡‡•ͳͳǣͻͷͻǦͻͺͲǤ 584 ‹‡’ƒǡ‘‘ ǡƒ” ǡ‘‰ǡ—ƒ”ǡ —ǡ‡‡ǡƒƒŠƒ•Š‹ ȋʹͲͲ͹Ȍ‹” ƒ†‹ƒ 585 —–ƒ–˜‡”–‹‡”‡˜‡ƒŽ• Ǧ„‘š’”‘–‡‹ ͵”‡‰—Žƒ–‹‘‘ˆ ”›’–‘ Š”‘‡ƒ† 586 ’‡”‹‘†‰‡‡‡š’”‡••‹‘Ǥ‡ŽŽͳʹͻǣͳͲͳͳǦͳͲʹ͵Ǥ 587 ‹˜ƒ Š‡‘ǡ‹ǡ„”—œœ‹ǡ‘•„ƒ•ŠȋʹͲͳ͵ȌŠ‡–”ƒ• ”‹’–‹‘ˆƒ –‘”‡ˆʹŽ‹•–Š‡ 588 ”‘•‘’Š‹Žƒ ‘”‡ Ž‘ –‘ ƒ•ʹǡ‡—”‘ƒŽ‘”’Š‘Ž‘‰›ǡƒ† ‹” ƒ†‹ƒ„‡Šƒ˜‹‘”Ǥ 589 ‡—”‘͹ͻǣʹͺͳǦʹͻʹǤ 590 ‹–Šǡ‘—–ǡŠ”‡‡ǡƒŽŽƒ‰ŠƒǡŽƒ ǡ‹•ƒ ǡ‹ ‡– ǡƒ‰ǡƒ‘ǡ 591 ƒ”ȋʹͲͲ͸ȌƒŽ’ƒ‹Ǧ”‡‰—Žƒ–‡†’͵ͷȀ †ͷ’Žƒ›•ƒ ‡–”ƒŽ”‘Ž‡‹†‘’ƒ‹‡”‰‹ 

20

592 ‡—”‘†‡ƒ–Š–Š”‘—‰Š‘†—Žƒ–‹‘‘ˆ–Š‡–”ƒ• ”‹’–‹‘ˆƒ –‘”›‘ ›–‡‡Šƒ ‡” 593 ˆƒ –‘”ʹǤ ‡—”‘• ‹ʹ͸ǣͶͶͲǦͶͶ͹Ǥ 594 ƒƒŠƒ•Š‹ ȋʹͲͳ͹Ȍ”ƒ• ”‹’–‹‘ƒŽƒ” Š‹–‡ –—”‡‘ˆ–Š‡ƒƒŽ‹ƒ ‹” ƒ†‹ƒ Ž‘ Ǥƒ– 595 ‡˜ ‡‡–ͳͺǣͳ͸ͶǦͳ͹ͻǤ 596 ƒƒŠƒ•Š‹ ǡ ‘‰ ǡ‘ ǡ ‡ƒ”‘ȋʹͲͲͺȌŠ‡‰‡‡–‹ •‘ˆƒƒŽ‹ƒ ‹” ƒ†‹ƒ 597 ‘”†‡”ƒ††‹•‘”†‡”ǣ‹’Ž‹ ƒ–‹‘•ˆ‘”’Š›•‹‘Ž‘‰›ƒ††‹•‡ƒ•‡Ǥƒ–‡˜ ‡‡–ͻǣ͹͸ͶǦ 598 ͹͹ͷǤ 599 ƒ‰ǡƒ‰ǡ ‘‰ǡ‘‰ǡƒ”ǡ‹ƒǡƒ‘ȋʹͲͲͷȌ› Ž‹Ǧ†‡’‡†‡–‹ƒ•‡ͷ 600 ‡†‹ƒ–‡•‡—”‘–‘š‹Ǧ‹†— ‡††‡‰”ƒ†ƒ–‹‘‘ˆ–Š‡–”ƒ• ”‹’–‹‘ˆƒ –‘”›‘ ›–‡ 601 ‡Šƒ ‡”ˆƒ –‘”ʹǤ ‡—”‘• ‹ʹͷǣͶͺʹ͵ǦͶͺ͵ͶǤ 602 ‘•‹‹ ǡ‰—œœ‹ ǡ—ŽŽ‘ ǡ‹—ǡƒ•ƒƒ–•—ȋʹͲͲ͹Ȍˆˆ‡ –‘ˆ’Š‘–‘”‡ ‡’–‘” 603 †‡‰‡‡”ƒ–‹‘‘ ‹” ƒ†‹ƒ’Š‘–‘”‡ ‡’–‹‘ƒ†ˆ”‡‡Ǧ”—‹‰’‡”‹‘†‹–Š‡‘›ƒŽ 604 ‘ŽŽ‡‰‡‘ˆ—”‰‡‘•”ƒ–Ǥ”ƒ‹‡•ͳͳͶͺǣ͹͸ǦͺʹǤ 605 ‡†ƒ ǡ ƒ›ƒ•Š‹ǡŠ‡ǡƒ‘ǡƒ Š‹†ƒǡŠ‹‰‡›‘•Š‹ǡ ‹‘ǡ ƒ•Š‹‘–‘ȋʹͲͲͷȌ 606 ›•–‡ǦŽ‡˜‡Ž‹†‡–‹ˆ‹ ƒ–‹‘‘ˆ–”ƒ• ”‹’–‹‘ƒŽ ‹” —‹–•—†‡”Ž›‹‰ƒƒŽ‹ƒ 607 ‹” ƒ†‹ƒ Ž‘ •Ǥƒ– ‡‡–͵͹ǣͳͺ͹ǦͳͻʹǤ 608 ‡Ž•ŠǡƒƒŠƒ•Š‹ ǡƒ›ȋʹͲͳͲȌ—’”ƒ Š‹ƒ•ƒ–‹ — Ž‡—•ǣ ‡ŽŽƒ—–‘‘›ƒ† 609 ‡–™‘”’”‘’‡”–‹‡•Ǥ—‡˜Š›•‹‘Ž͹ʹǣͷͷͳǦͷ͹͹Ǥ 610 ƒƒœƒ‹ǡŽ‘‡•ǡ‡ƒ‡”ȋʹͲͲʹȌ –‡”ƒ –‹‘‘ˆ–Š‡”‡–‹ƒ™‹–Š•—’”ƒ Š‹ƒ•ƒ–‹  611 ’ƒ ‡ƒ‡”•‹–Š‡ ‘–”‘Ž‘ˆ ‹” ƒ†‹ƒ„‡Šƒ˜‹‘”Ǥ ‹‘ŽŠ›–Š•ͳ͹ǣ͵ͳͷǦ͵ʹͻǤ 612 ƒ‰ǡ ƒ‘ǡ— ǡƒ‰ǡ ƒ‘ ǡŠ— ǡƒ‹ǡƒ‹ ǡƒ‰ȋʹͲͳͷȌ”ƒ• ”‹’–‹‘ˆƒ –‘” 613 ›‘ ›–‡‡Šƒ ‡”ˆƒ –‘”ʹ”‡‰—Žƒ–‡•‹–‡”Ž‡—‹ǦͳͲ’”‘†— –‹‘‹‹ ”‘‰Ž‹ƒ–‘ 614 ’”‘–‡ –‡—”‘ƒŽ ‡ŽŽ•ˆ”‘‹ˆŽƒƒ–‹‘Ǧ‹†— ‡††‡ƒ–ŠǤ ‡—”‘‹ˆŽƒƒ–‹‘ 615 ͳʹǣ͵͵Ǥ 616 ‘‘ ǡƒƒœƒ‹ǡ‘™”‡›ǡŠ‹‘—”ƒǡ‘ ǡ—Š”ǡ‹‡’ƒǡ ‘‰ ǡŠ ǡ 617 ‘‘ ǡ‡ƒ‡”ǡƒƒŠƒ•Š‹ ȋʹͲͲͶȌ ʹǣǣ ”‡ƒŽǦ–‹‡”‡’‘”–‹‰ 618 ‘ˆ ‹” ƒ†‹ƒ†›ƒ‹ •”‡˜‡ƒŽ•’‡”•‹•–‡– ‹” ƒ†‹ƒ‘• ‹ŽŽƒ–‹‘•‹‘—•‡’‡”‹’Š‡”ƒŽ 619 –‹••—‡•Ǥ”‘ ƒ–Ž ƒ† ‹ͳͲͳǣͷ͵͵ͻǦͷ͵Ͷ͸Ǥ 620 621

21

622 FIGURE LEGENDS 623 624 625 Fig. 1: Global loss of Mef2d Lengthens Circadian Free-Running Period and Alters

626 Behavioral Activity Patterns. Representative actograms of WT (a) and KO (b) mice on

627 running-wheels. Actograms are double plotted, with each horizontal line representing 48h of

628 activity. Mice were initially housed in LD12:12, as indicated by the yellow shading (lights on) and

629 results analyzed by student’s t-tests. (c) Summary data (mean ± SEM) for activity onset in LD

630 (n= 13 WT, 18 KO; t(29) = 3.762, *p=0.0008); (d) Summary data (mean ± SEM) for free-running

631 period in DD (n = 18 WT, 22 KO; t(38) = 8.340, *p < 0.0001). (e) Mean ± SEM number (t(29) =

632 4.257, *p =0.0002) and duration (t(29) = 4.844, *p < 0.0001) of activity bouts as measured in DD

633 (n = 12 WT, 19 KO). (f, g) Representative ethograms from video analyses of WT (f) and KO (g)

634 mice (n=2/group). Ethograms are double plotted, with each horizontal line representing 48h of

635 activity. Mice were housed in LD12:12, as indicated by the yellow shading (lights on). (h).

636 Representative scored ethograms from WT (blue) and KO (red) mice, representing 15.5h of

637 data beginning at ZT12 (lights-off) on the day indicated by the red arrows in (f, g).

638

639 Fig. 2: Mef2d Activity in the Brain is Necessary for Normal Circadian Behavior. CamKIIα-

640 iCre mice were crossed with Mef2a, Mef2c, and Mef2d floxed lines to produce mice with a

641 varying combination of floxed Mef2 alleles. Representative actograms from (a) Mef2dfx/fx control

642 and (b) CamKIIα-iCre:Mef2dfx/fx mice. Actograms are double plotted, with each horizontal line

643 representing 48h of activity. Mice were initially housed in LD12:12, as indicated by the yellow

644 shading (lights on). (c) Free-running periods in DD (mean ± SEM) from floxed and control mice

645 (n = 14 Mef2a, 4 Mef2c, 13 Mef2d, 32 control; analyzed by one-way ANOVA, F(3, 59) = 25.19,

646 *p < 0.0001, different from all other conditions assessed by Tukey post hoc comparisons).

647

22

648 Fig. 3: Mef2d KO Causes a Short Free-Running Period in the SCN. (a) Detrended,

649 smoothed, bioluminescence traces from representative WT and KO mouse SCNs. Mice were

650 housed in T24 (LD12:12) prior to tissue harvest and results analyzed with student’s t-tests. (b)

651 Periods (mean ± SEM) of LD12:12 SCN explants (n= 7 WT, 7 KO; t(12) = 3.443, *p=0.0049).

652 (c) Periods (mean ± SEM) of LD12:12 peripheral explants (n= 3-10 mice/tissue/genotype; each

653 tissue analyzed separately, t(17)=3.279, *p=0.0044). (d) Detrended, smoothed,

654 bioluminescence traces from representative WT and KO mouse SCNs. Mice were housed in DD

655 prior to tissue harvest. (e) Periods (mean ± SEM) of DD SCN explants (n = 4 WT, 4 KO; t(6) =

656 0.598, p = 0.572). (f) Periods (mean ± SEM) of DD peripheral tissue explants (n= 2-4

657 mice/tissue/genotype). (g, h) Detrended, smoothed, bioluminescence traces from representative

658 WT and KO mouse SCNs. Mice in g were housed in T22 (LD 11:11) and mice in h were housed

659 in T26 (LD 13:13) prior to tissue harvest and results analyzed by two-way ANOVA. (i) Periods

660 (mean ± SEM) of SCN explants (T22: n = 6 WT, 9 KO; T24: n = 7 WT, 7 KO (also in b); T26: n =

661 6 WT, 6 KO; main effect of genotype F(1, 35) = 12.24, * p =0.0013 for T24; no main effect of T

662 length).

663

664 Fig. 4: Clock Gene Expression is Not Greatly Affected by loss of Mef2d. Quantitative RT-

665 PCR analysis of clock gene expression in WT and Mef2d KO cerebellum (a) and heart (b) taken

666 from mice housed in T24, LD12:12 (LD). Data are presented as relative quantification compared

667 to WT0; n = 2-3/group; and analyzed by two-way ANOVA. Per1: interaction (F(1,7)= 8.502,

668 p=0.0225) and main effect of time (F(1,7)=21.19, p=0.0025); * p =0.0089 WT12 different from

669 WT0; Reverba: interaction (F(1,7)=8.076, p=0.0250, main effect of time (F(1,7)=43.10,

670 p=0.0003, and main effect of genotype (F(1,7)=6.001, p=0.0441); *p = 0.0218 KO12 different

671 from WT12; Mef2d: t(3)=3.296, *p=0.0459 via student’s t-test.

672

23

673 Fig. 5: MEF2D Does not Directly Interact with Core Clock Components. Co-transfection of

674 Mef2d does not lead to activation of (a) Per1::Luc (average of 2 replicates across 3

675 experiments; 125 ng Mef2d transfected), (b) Per2::Luc (average of 2 replicates across 2

676 experiments; increasing doses of Mef2d from 5-50 ng), or (c) Bmal1:Luc (average of 2

677 replicates from 1 experiment; increasing doses of Mef2d from 5-50ng) in reporter assays. Both

678 Per1::Luc and Per2::Luc activation is observed in the presence of known transcriptional

679 activators, BMAL1 and CLOCK (Per1: f(3,20)= 10.80; ** p=0.0002; Per2: f(10,33)=3.42,

680 p=0.0037, ns after correction for multiple comparisons) (d) Co-transfection of Mef2d activates a

681 Mef2 binding domain fused to a TATA-box (average of 2 replicates across 2 experiments; 125

682 ng Mef2d transfected; t(3.127)=8.472; *p=0.029). (e) MEF2D is not pulled down with BMAL1 in

683 co-IP experiments, whereas the known binding partner, CLOCK, was successfully co-IP’d and

684 FLAG (BMAL1) enrichment is observed.

685

686 Fig. 6: Loss of Mef2d Leads to Fragmented Sleep. Patterns of wakefulness (a), NREM (b),

687 and REM sleep (c) are shifted towards a later phase in the KO mice, mirroring the delayed

688 phase observed from running-wheel behavior. (d) Mean ± SEM number of transitions over the

689 full day. Data are presented as mean ± SEM (n= 8 WT, 8 KO) and analyzed by student’s t-

690 tests; there were no significant differences between genotypes. However, KO mice tended to

691 have more transitions from wakefulness to NREM sleep (t(14)= 2.077; p = 0.0567) and from

692 NREM sleep to wakefulness (t(14)= 2.015; p = 0.0635) or REM sleep (t(14)= 2.002; p = 0.0651).

693 (e) Mean ± SEM REM latency is shorter in Mef2d KO mice (n’s same as above; results

694 analyzed with student’s t-tests). Data are presented over the full day (24; t(14)=3.147,

695 *p=0.0071), the light portion of the LD cycle (Light; t(14)=2.848, **p=0.0129), or the dark portion

696 of the LD cycle (Dark; t(14)=3.108, ***p=0.0077). (f) NREM episode duration is shorter in Mef2d

697 KO mice (n’s same as above; analyzed with student’s t-tests). Data are presented over the full

698 day (NREM24; t(14)=2.829, *p=0.0134), the light portion of the LD cycle (NREMLight;

24

699 t(14)=2.557, **p=0.0228), or the dark portion of the LD cycle (NREMDark: t(14)=2.910,

700 ***p=0.0114) (g) NREM episode duration as a percentage of all NREM episodes reveals

701 fragmented sleep patterns in KO mice.

702

703 TABLES

704

705 706 Table 1: Primers for Real-time PCR quantification of mRNA expression.

Gene Primer Set F 5' -CCA CCT CAG AGC CAT TGA TAC A- 3' Bmal1 R 5' -CGA AGA ACG TCA TGA TGC AG- 3' F 5' -CCC AGC TTT ACC TGC AGA AG- 3' Per1 R 5' -ATG GTC GAA AGG AAG CCT CT- 3' F 5' -TGT GCG ATG ATG ATT CGT GA- 3' Per2 R 5' -GGT GAA GGT ACG TTT GGT TTG C- 3' F 5' -CGA AGA ACG TCA TGA TGC AG- 3' Dbp R 5' -GGT TCC CCA ACA TGC TAA GA- 3' Nr1d1 F 5' -AGA CTT CCC GCT TCA CCA AG- 3' (Reverba) R 5' -AGC TTC TCG GAA TGC ATG TT- 3' F 5' -ACG AGA GCC GCA CCA ATG CT- 3' Mef2d R 5' -GGA ACA GAT GAC CCA TAG CGC CTG- 3' F 5' -CAA GGA GTA AGA AAC CCT GGA CC- 3' Gapdh R 5' -CGA GTT GGG ATA GGG CCT CT- 3' 707

708

25 Figure 1

abWT KO Day Day

0 8 16 0 8 16 0 0 8 16 0 8 16 0 Time (h) Time (h) c de

6 Ave Bout Length (min) 24.5 * * 250 0.0 24.0 200 4 -0.2 23.5 150

-0.4 2 * Period (h) 23.0 # Bouts/Day 100 WT -0.6 KO 22.5 0 50 Onset (h prior to lights off) * fgWT KO Day Day

18 2 10 18 2 10 18 18 2 10 18 2 10 18 Time (h) Time (h) h WT KO

Rest (sleep) 8 At end of cage (possible sleep) 7 General activity (far end of cage) 6 General activity (near wheel) 5 Near food hopper 4 At food hopper (eating) 3 At sipper (drinking) 2 On wheel 1

3 3 13 15 17 19 21 23 1 3 1 15 17 19 21 2 1 3 Time (h) Time (h) Figure 2

a Mef2dfx/fx

c 24.5 * Day 24.0

23.5 0 8 16 0 8 16 0

Time (h) Period (h) b CamKIIα-iCre x Mef2dfx/fx 23.0

22.5 Mef2c Mef2a Mef2d Day Control

0 8 16 0 8 16 0 Time (h) Figure 3 abc WT 200 LD 26 25 KO * 100 24 24 * (h)

0 Period Period (h) Period 23 22 -100

-200 22 20 Bioluminescence (detrended) Bioluminescence 1 2 3 4 5 6 7 LD SCN g ry Days in Culture Heart Liver Lun Kidney Pituita def

200 DD 25 26

100

24 (h) 24

0 Period (h) Period 23 Period 22 -100

-200 22 20 Bioluminescence (detrended) Bioluminescence 1 2 3 4 5 6 7 DD SCN ary Days in Culture Heart Liver Lung Kidney Pituit ghi

200 T22 200 T26 25 * 100 100 24 0 0

Period (h) Period 23 -100 -100

-200 -200 22 Bioluminescence (detrended) Bioluminescence 1234567 1234567 4 T22 T2 Days in Culture Days in Culture T26 Figure 4 WT a Cerebellum (LD) KO Bmal1 Per1 Per2 2.0 1.5 1.5 * 1.5 1.0 1.0

1.0

0.5 0.5 Relative 0.5

Quantification (RQ) 0.0 0.0 0.0 0 12 012 012 Dbp Rev-erbα Mef2d 1.5 1.5 1.5

* 1.0 1.0 1.0

0.5 0.5 0.5 Relative

Quantification (RQ) 0.0 0.0 0.0 012 012 012 Time (h)Time (h) Time (h)

b Heart (LD) WT KO Bmal1 Per1 Per2 10.0 1.5 1.5

8.0 1.0 1.0 6.0

4.0 0.5 0.5 Relative 2.0

Quantification (RQ) 0.0 0.0 0.0 012 012 012 Dbp Rev-erbα Mef2d 2.0 2.5 * 1.5 2.0 1.5 1.0 1.5 1.0 1.0 0.5 Relative 0.5 0.5

Quantification (RQ) 0.0 012012012 Time (h)Time (h) Time (h) Figure 5

abPer1::Luc Per2::Luc 200 12 ** 150 8

100

4 LUC/REN 50 LUC/REN

0 0 Mef2d-HA + + Mef2d-HA Flag-Bmal1 + + Flag-Bmal1 + + + + + + Flag-Clock + + Flag-Clock + + + + + + cde Bmal1::Luc desMef2::Luc Input IP: FLAG 200 10 * Mef2d-HA + + + + + + 8 Flag-Bmal1 + + + + + + 150 Clock + + 6 100 WB: HA 4 LUC/REN LUC/REN WB: CLOCK 50 2 WB: FLAG 0 0 WB: TUBULIN Mef2d-HA Mef2d-HA + Figure 6

abWake NREM c REM

60 60 10 WT KO 40 40 6

20 20 2 Time in state (mins) 0 0 0 0 2 4 6 8 6 0 2 4 6 8 10 12 14 16 18 20 22 10 12 14 16 18 20 22 0 2 4 6 8 10 12 14 1 18 20 22 Hour (ZT) Hour (ZT) Hour (ZT) de 400 WT 8 300 KO *** 6 200 * ** 100 4 40 30 # Transitions 2

20 REM latency (min) 10 0 0 M 24 Dark RE Light e to REM EM to Wake Wak R Wake to NREM NREM to WakeNREM to REM to NREM f g 20 100 15 10 80 8 60 6 *** 40 4 * ** 2 20 Episode Duration (min) 0 0 % of All NREM Episodes % of 0 5 10 15 20 25 30 35 40 45 5

REM24 NREM Episode Duration (epochs) Wake24NREM24 REMLightWakeDark REMDark WakeLightNREMLight NREMDark