MCB Accepts, published online ahead of print on 6 January 2014 Mol. Cell. Biol. doi:10.1128/MCB.01195-13 Copyright © 2014, American Society for Microbiology. All Rights Reserved.

1

Zac1 regulates cell cycle arrest in neuronal

2

progenitors via Tcf4

3 4 5

running title: Zac1 regulates Tcf4 expression

6 7 8

Udo Schmidt-Edelkraut1, Guillaume Daniel1#, Anke Hoffmann and Dietmar

9

Spengler*

10

Max Planck Institute of Psychiatry, Molecular Neuroendocrinology, Kraepelinstr. 2-10,

11

80804 Munich, Germany

12 13

#

14

Switzerland

15

1

these authors contributed equally to this work

17

*

corresponding author

18

Dietmar Spengler, Molecular Neuroendocrinology, Max Planck Institute of Psychiatry,

19

Kraepelinstr. 2-10, 80804 Munich, Germany, Phone: ++49 89 30622559, Fax: ++49 89

20

30622605, Email: [email protected]

present address: École polytechnique fédérale de Lausanne, LMNR, CH-1015 Lausanne,

16

21 22 23

word count (Materials and Methods) : 853

24

word count (Introduction, Results, and Discussion) : 3124

25

1

26

ABSTRACT

27

Imprinted genes play a critical role in brain development and mental health although the

28

underlying molecular and cellular mechanisms remain incompletely understood. The family

29

of basic Helix-Loop-Helix (bHLH) proteins directs the proliferation, differentiation and

30

specification of distinct neuronal progenitor populations. Here, we identified the bHLH factor

31

Tcf4 as a direct target gene of Zac1/Plagl1, a maternally imprinted transcriptional regulator,

32

during early neurogenesis. Zac1 and Tcf4 expression concomitantly increased during neuronal

33

progenitor differentiation; moreover, Zac1 interacts with two cis-regulatory elements in the

34

Tcf4 gene locus and these elements together confer synergistic activation of the Tcf4 gene.

35

Tcf4 upregulation enhances the expression of the cyclin-dependent kinase inhibitor p57Kip2, a

36

paternally imprinted Tcf4 target gene, and increases the number of cells in G1 phase. Overall,

37

we show that Zac1 controls cell cycle arrest function in neuronal progenitors through

38

induction of p57Kip2 via Tcf4 and provide evidence for a co-operation between imprinted

39

genes and a bHLH factor in early neurodevelopment.

40

2

41

INTRODUCTION

42

Multiple factors and signaling pathways control in a concerted manner cell fate decisions and

43

differentiation during brain development. Among these, the family of basic Helix-Loop-Helix

44

(bHLH) proteins coordinates proliferation, specification, differentiation and migration of

45

progenitors during neurogenesis (1, 2). These proteins are characterized by the presence of a

46

basic helix-loop-helix domain that allows them upon homo- or heterodimerization to bind to

47

specific DNA sequences (CANNTG), known as E-box (3). The bHLH family comprises two

48

major classes, the so called specification factors (i.e. Neurogenin, Math, Mash and NeuroD),

49

whose expression is spatiotemporally controlled, and their ubiquitously expressed

50

dimerization partners, the E-proteins (4, 5). Proneural factors are expressed at low levels in

51

proliferating undifferentiated progenitors while with the onset of neurogenesis the expression

52

of specification factors and E-protein family members (the two splice variants of E2A: E12

53

and E47, HEB and TCF4 (alias E2-2, SEF2 or ITF2) increases and inhibits progenitor

54

proliferation and astrogliogenesis through the induction of target genes that are required for

55

terminal neuronal differentiation (6, 7).

56 57

The maternally imprinted Zac1 gene (zinc finger protein regulating apoptosis and cell cycle

58

arrest) is transiently expressed in proliferating stem/progenitor cells of the telencephalic and

59

cerebellar ventricular zones (8–10), the external granular cell layer (11), and the retina (12,

60

13), the function of which is, however, poorly understood. Imprinted genes encompass a

61

subset of mammalian genes that are subject to developmentally determined, parent-of-origin

62

dependent, epigenetic modifications resulting in monoallelic expression. On the other hand,

63

loss of imprinting, i.e. biallelic expression, frequently manifests with severe metabolic and

64

neurodevelopmental syndromes across prenatal and postnatal life (14).

65

3

66

Zac1 encodes a zinc finger protein conferring transcriptional activation and repression

67

following monomer or dimer binding to GC-rich palindromic and repeat DNA elements (15–

68

17). Direct Zac1 target genes identified so far include the G-protein coupled receptor Pac1

69

(pituitary adenylate activating receptor 1) (18–20), the nuclear receptor Pparγ1 (peroxisome

70

proliferator activated receptor) (21), the cyclin-dependent kinase inhibitor p21Waf1/Cip1 (22)

71

and the exchange factor Rasgrf1 (RAS protein-specific guanine nucleotide-releasing factor 1)

72

(23) all of which share a role in the regulation of apoptosis, cell cycle arrest and

73

differentiation across different tissues and stages of development.

74

Moreover, we recently found that Zac1 expression prevents precocious astroglial

75

differentiation by restraining Jak/Stat3 signaling in embryonic and adult neural stem cells

76

(NSC) through transcriptional induction of Socs3 (24). Here, we further show that Tcf4 is a

77

lineage-specific target gene of Zac1 in the control of neuronal progenitor cell cycle arrest

78

function consistent with a role of Zac1 in both lineage decisions.

79 80

MATERIALS AND METHODS

81

Cell culture and transfection experiments. The mouse C17.2 NSC line was cultured in

82

Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% fetal calf serum

83

(both Life Technologies GmbH, Darmstadt, Germany). Tetracycline (Tc) regulated Zac1

84

expression in C17.2 cells was established and proliferation measured as described (21, 23).

85

The mouse embryonic stem cell line 46C was grown and neuronal differentiation performed

86

as reported (25). In addition, cells were differentiated by treatment with all-trans retinoic acid

87

(RA; 0.1 μM) or embryoid body (EB) formation as reported elsewhere (26). The mouse

88

embryonic NS-5 and adult O4ANS NSC lines were grown as reported (24). Primary cells

89

from whole fetal brain (embryonic day 15; E15) of CD1 mice were dissected as described

90

previously (24) and grown as suspension in DMEM/F12 and Neurobasal medium (1:1)

91

supplemented with N2 (1% v/v), B27 (2% v/v) (all Life Technologies GmbH), EGF 4

92

(epidermal growth factor), and FGF (fibroblast growth factor) (both 10 ng/ml; PeproTech,

93

Hamburg, Germany). Neurospheres were dissociated with Accutase (Millipore, Schwalbach,

94

Germany) and grown as monolayers on poly-D-lysine hydrobromide (Sigma, Munich,

95

Germany) coated plates in the presence of EGF and FGF. Neuronal differentiation was

96

initiated with FGF (10 ng/ml) on matrigel coated dishes (0.4 μl/ml; BD Bioscience,

97

Heidelberg, Germany). For astroglial differentiation, cells were kept with 1% FCS. All media

98

contained penicillin/streptomycin (Life Technologies GmbH).

99

Transient and stable transfections were performed using Turbofect transfection reagent

100

(Fermentas, St. Leon-Roth, Germany) according to manufacturers’ instructions using 1−5 x

101

105 cells/cm² as described (24). To knock-down Zac1, p57Kip2 or Tcf4 expression the

102

following shRNA vectors (Mission shRNA, Sigma) were used: pLKO.1-Pur-Zac1.pool (clone

103

NM_009538), pLKO.1-Neo-p57Kip2 (clone NM_009876.2-1060s1c1), pLKO.1-Neo-Tcf4

104

(clone NM_013685.1-571s1c1) and pLKO.1-Pur-Non-Target shRNA (SHC016), which

105

served as a control. The region of Tcf4 targeted by shRNA is shown in Figure S1 of the

106

supplemental material.

107

Transfection of primary NSCs (E15) in knock-down experiments was performed with 1 μg of

108

the appropriate expression vectors (24). For quantitative analysis, images of 50 GFP positive

109

cells per transfection corresponding to a total number of 2,000-4,000 cells were scored for

110

Ki67 immunoreactivity by an independent investigator. Nucleofection of primary NSCs (E15)

111

was performed with Amaxa Basic Neuron SCN Nucleofector Kit and the Nucleofector

112

technology (Lonza, Tokyo, Japan) according to manufacturers’ instructions. Luciferase

113

reporter activities were normalized on β-galactosidase activity of a cotransfected expression

114

vector (27). Amounts of transfected plasmids are indicated in the corresponding figure

115

legends.

116

5

117

Plasmids. Fragments of the Tcf4 promoter, intron and regulatory element (RE) were

118

amplified by polymerase chain reaction (PCR), sequence verified and cloned in the pGL3

119

basic vector (Promega, Mannheim, Germany). Details on the generation of the constructs are

120

available on request. Zac1 expression constructs are described elsewhere (17). The

121

pCDEF3.Flag-Tcf4 expression vector was described previously (28).

122 123

Chromatin immunoprecipitation (ChIP), RNA extraction and PCR experiments. ChIP

124

assays were performed as described (29) with rabbit polyclonal Zac1 (21, 25) or rabbit

125

polyclonal Tcf4 (aa 32-154) (Pineda, Berlin, Germany) antisera. The specificity of the Tcf4

126

antibody was verified by immunoblotting (see Fig. S1 in the supplemental material). Brain

127

punches enclosing the neocortical VZ/SVZ were obtained by micro-dissection under

128

histological control, whereby tissues from five (E15) or three (E18) animals were pooled for

129

each experiment. Data are diagrammed as percent of input normalized to control sera (IgG or

130

preimmune-serum). RNA extraction and PCR experiments were done as described (25).

131

Primers for PCR and ChIP experiments are listed in Table S1 of the supplemental material.

132

The housekeeping genes Gapdh (glycerinaldehyde-3-phosphat-dehydrogenase), Atp5j (ATP

133

synthase-coupling factor 6) or β-actin served for normalization.

134 135

Immunoblotting, immunocytochemistry and immunohistochemistry. Whole cell extracts

136

(30 to 100 µg) were fractionated by SDS-PAGE gel electrophoresis and tested with the

137

following antibodies: mouse monoclonal Tcf4 [clone Ri-3B9, aa 1-183, see Fig. S1 in the

138

supplemental material (30)], actin (sc-8432, Santa Cruz), Flag (F3165, Sigma), or Zac1

139

(rabbit polyclonal) (21, 25). For immunocytochemistry, cells were grown on poly-D-lysine

140

hydrobromide coated coverslips, fixed with 4% formaldehyde, permeabilized with 0.1%

141

saponin (Sigma) in phosphate buffered saline (PBS), blocked with donkey normal serum

142

(Sigma) and subjected to indirect immunofluorescence with antibodies to Tcf4 (clone Ri6

143

3B9), Gfap (Z0334, Dako, Hamburg, Germany), Tuj1 (ab7751, Abcam), Nestin (ab6142,

144

Abcam), p57Kip2 (sc-8298, Santa Cruz) and Zac1 (guinea pig polyclonal) (23, 24). Nuclei

145

were stained with 2-(4-Amidinophenyl)-6-indolecarbamidine-dihydrochloride (DAPI).

146

Immunohistochemistry was performed on 4% paraformaldehyde-fixed, cryopreserved brain

147

sections (30 μm) from CD1 mice with antibodies to Nestin, Dcx (sc-8066, Santa Cruz),

148

p57Kip2, Tcf4 (clone Ri-3B9) and Zac1 (guinea pig polyclonal). Nuclei were stained with

149

DAPI.

150

Microscopic analysis was done with fluorescence microscope (BX61, Olympus, Hamburg,

151

Germany) and CCD camera (E-620 SRL, Olympus) or confocal microscope (FluoView 1000,

152

Olympus). All images were captured using identical laser power and gain settings.

153 154

Fluorescence-Activated Cell Sorting Analysis

155

Cells (2 x 106) were stained with propidium iodide and analyzed on a Beckman Coulter Epics

156

XL using Expo32 ADC analysis as described previously (24).

157 158

Software and statistical analysis. Computational analysis of the Tcf4 and p57Kip2 gene was

159

done with Genomatix MatInspector™. Results represent the means and standard deviations

160

from at least four independent experiments. Numerical data were analyzed by unpaired

161

Student’s t test and ANOVA with post-hoc Tukey test. The threshold for significance was set

162

at * P < 0.05 and ** P < 0.01.

163 164

RESULTS

165

Zac1 transactivates Tcf4. To identify Zac1 target genes in neural progenitors, we generated

166

a panel of inducible Zac1 clones in the C17.2 cerebellar neural stem cell line using a Tet-off

167

system. Comparative, genome-wide expression analysis for two representative Zac1 clones

168

(21) showed enhanced expression of Tcf4 (1.7-fold, data not shown) after Zac1 induction. 7

169

Quantitative RT-PCR (qRT-PCR) corroborated a transient increase in Zac1 expression

170

peaking at 9 h and declining at 24 h after tetracycline removal. At the same time, a similar

171

pattern of regulation was detected for Tcf4 (Fig. 1A). Moreover, transient transfection of a

172

Zac1 expression vector into parental C17.2 cells caused a robust increase in both Tcf4 gene

173

expression and protein levels of the b-isoform (hereafter referred to as Tcf4), while the a-

174

isoform was unaffected (Fig. 1B and C, see Fig. S1 in the supplemental material and data not

175

shown). Together, these findings raised the possibility that Zac1 up-regulates Tcf4.

176

Zac1’s function as transactivator (15, 16) or coactivator (25, 31) was assessed by transfection

177

of C17.2 cells with constructs either defective in DNA binding or in transactivation due to a

178

mutation in zinc finger seven (ZF7mt) or to absence of the central transactivation domain

179

(ΔLPR) (17). RNA (qRT-PCR) and immunoblot analysis evidenced that Tcf4 was barely

180

induced by either ZF7mt or ΔLPR when compared to wild type Zac1 (Fig. 1D and E)

181

although these constructs were expressed at similar levels (Fig. 1E). Because Zac1’s

182

coactivator function is maintained in the absence of the central transactivation domain (25),

183

we conclude that Zac1’s transactivation and DNA binding domains are required for Tcf4

184

regulation pointing to a role as transcription factor.

185

Computational analysis of the Tcf4 locus revealed multiple potential GC-rich palindromic

186

and repeat Zac1 binding sites (17) at both the promoter and first intron as schematically

187

depicted in Figure 1F. In contrast, no corresponding sites could be identified at the Tcf4-a

188

promoter (data not shown). Chromatin immunoprecipitation (ChIP) experiments using a Zac1

189

antiserum showed that Zac1 occupies exclusively the Tcf4 proximal promoter and the center

190

of the first intron (Fig. 1G).

191

In sum, these findings suggest that Zac1 directly regulates Tcf4 following binding at the

192

proximal promoter and/or the first intron in mouse C17.2 stem cells.

193

8

194

Zac1 and Tcf4 are upregulated during neuronal differentiation of mouse embryonic

195

stem cells. Mouse embryonic stem cells (ESC) were used as a model for early neuronal

196

differentiation (25). Upon withdrawal of Leukemia inhibitory factor (Lif), cell clusters

197

flattened and spread to give rise to single cells that acquired progressively neuronal

198

morphology within 6 days (Fig. 2A). At the same time, expression of the pluripotency marker

199

Oct4 and the late neuronal differentiation marker Tuj1 rapidly decreased and increased,

200

respectively (Fig. 2B and C). Moreover, both Zac1 and Tcf4 mRNA and protein expression

201

steadily rose with progressive neuronal differentiation (Fig. 2D and E).

202

We further studied co-induction of Zac1 and Tcf4 under embryoid body formation or retinoid

203

acid treatment, simulating early embryo formation (32) and posterior brain development (33),

204

respectively. Both protocols promoted efficient neuronal differentiation (loss of Oct4

205

expression, upregulation of the progenitor marker Nestin and the neuronal markers N-

206

cadherin and Tuj1) and upregulation of Zac1 and Tcf4 (see Fig. S2 in the supplemental

207

material). Together, these results indicate that co-induction of Zac1 and Tcf4 is common to

208

different models of neuronal stem cell differentiation.

209 210

Zac1 binding transactivates Tcf4. ChIP experiments revealed barely detectable Zac1

211

binding at the Tcf4 locus under Lif conditions, whereas differentiation caused a marked

212

increase at the proximal promoter, particularly at the core region (-485 and -89 bp), and at the

213

center of the first intron (+1933 bp) (Fig. 3A). The contribution of the different binding sites

214

to transactivation was studied by cloning various fragments encoding either the proximal

215

promoter or the first intron separately, or jointly (including the intervening exon) in front of a

216

luciferase reporter gene (Fig. 3B). Each fragment showed strong transcriptional activity when

217

compared to the parent vector with a strong decrease (50 to 100 fold) in the reverse (3´ to 5´)

218

orientation (Fig. 3C). Increasing doses of Zac1 transactivated the separate promoter and intron

219

constructs about 2.5-fold and 1.5-fold, respectively, and resulted in a 5.5-fold transactivation 9

220

in case of their joint presence. None of these reporter plasmids responded to the DNA binding

221

defective construct ZF7mt (Fig. 3D).

222

The intron was additionally cloned downstream of the thymidine kinase (TK) promoter

223

(devoid of Zac1 binding sites) and showed robust basal promoter activity when compared to

224

the parent vector (data not shown). However, Zac1 was ineffective to confer transactivation to

225

this chimeric reporter (Fig. 3E) and similar results were obtained when the intron was cloned

226

downstream of the SV40 promoter (data not shown). Moreover, insertion of the intron in the

227

reverse orientation adjacent to the promoter region prevented transactivation when compared

228

to the parent composite Tcf4 reporter construct (Fig. 3E).

229

In sum, these experiments suggest that Zac1 transactivates Tcf4 in a synergistic manner

230

through binding to the promoter and intron. Excluding a role as autonomous enhancer, the

231

intron operates solely in conjunction with the homologous promoter to confer Zac1

232

transactivation in an orientation-dependent manner.

233 234

Zac1-dependent regulation of Tcf4 is lineage-specific. Neural stem cell lines derived from

235

mouse embryonic (NS-5) and adult (O4ANS) brain (24) served to investigate Zac1-dependent

236

induction of Tcf4 across different developmental stages. Although Zac1 expression was up-

237

regulated under either neuronal or astroglial differentiation, Tcf4 increased solely in the

238

neuronal lineage (Fig. 4A and data not shown). Consistent with this finding, enhanced Zac1

239

occupancy at the Tcf4 promoter and first intron occurred exclusively under neuronal

240

differentiation as evidenced by ChIP experiments (Fig. 4B).

241

Epigenetic mechanisms, comprising DNA methylation and chromatin modifications among

242

others, are recognized for their role in directing NSC specification. To elucidate if lineage-

243

specific Zac1 binding and Tcf4 regulation is controlled at the level of DNA methylation, the

244

two CpG islands overlapping the 5’ end of Tcf4 (see Fig. S3 in the supplemental material)

245

were analyzed by MeDIP. We detected minor DNA methylation at these two regions that did 10

246

not change following neuronal or astroglial differentiation (see Fig. S3 in the supplemental

247

material).

248

Therefore, we additionally performed sequential ChIP experiments with antisera against

249

active (pan-acetyl histone 3, acH3) or repressive (dimethyl lysine 9 of histone 3, H3K9me2)

250

histone marks followed by immunoprecipitation with Zac1 antiserum. An increase in acH3

251

concomitant to a decrease in H3K9me2 was measured at the Tcf4 5’ end regulatory region

252

under neuronal differentiation whereas the opposite pattern emerged upon astroglial

253

differentiation (Fig. 4C and D). Moreover, Zac1 preferentially associated with chromatin

254

containing the active histone mark at the Tcf4 gene (Fig. 4E and F).

255

Taken together, changes in DNA methylation are unlikely to contribute directly to lineage-

256

specific Tcf4 regulation by Zac1. Neuronal differentiation induces however a more open

257

chromatin configuration at the Tcf4 5’ end regulatory region and associated with Zac1

258

binding.

259 260

Zac1 regulates Tcf4 in primary NSCs. Long-term cultivation of stem cells can compromise

261

aspects of cellular identity and differentiation potential (24). Therefore, Zac1-dependent Tcf4

262

induction was studied in primary cells of embryonic mice brain from day 15 (E15).

263

Neurosphere-derived cells show the capacity for self-renewal, Nestin immunoreactivity and

264

the ability to differentiate into neurons, astrocytes and oligodendrocytes (34). After 2 days of

265

monolayer culture, the cell population consisted mainly of Nestin positive radial glial-like

266

cells, which co-expressed Zac1 and Tcf4 (Fig. 5A). Cells were subsequently maintained under

267

different conditions to enhance neuronal versus astroglial differentiation (Fig. 5B). Across 6

268

days of differentiation, Zac1 was up-regulated under either condition with concomitant

269

increases in Tcf4 occurring solely in neurons (Fig. 5C and D).

270

In agreement with the results from above, Zac1 transactivated the composite Tcf4 reporter

271

construct in primary NSCs under neuronal, but barely under astroglial differentiation or 11

272

undifferentiated conditions (Fig. 5E). Moreover, Zac1 occupancy at the Tcf4 promoter and

273

first intron increased exclusively during neuronal differentiation as evidenced by ChIP

274

experiments (Fig. 5F) despite similar amounts of Zac1 protein expression under either

275

condition (Fig. 5D). These findings corroborate that Zac1 binding to the Tcf4 locus is

276

confined to the neuronal lineage.

277 278

Zac1 and Tcf4 colocalize in the neocortical ventricular zone. Neurogenesis generally

279

precedes astrogliogenesis in the developing mammalian brain, with the same progenitor

280

domains switching developmental programs from neuron to oligodendrocyte or astrocyte

281

production. Progenitors produce predominantly neuronal lineages in the ventricular and

282

subventricular zone (VZ/SVZ) from early (E9) to late (E17) embryonic stages in mice. To

283

investigate Zac1 and Tcf4 expression in the neocortical ventricular zone, we co-stained

284

coronal sections of E11 (early neurogenesis) and E15 (mid-neurogenesis) mice brains using

285

Zac1 and Tcf4 antisera. We detected colocalization of the two proteins in progenitor cells of

286

the VZ/SVZ hinting to a shared function between the two transcription factors (Fig. 6A and

287

B). As expected, Tcf4 positive cells colocalized with the progenitor marker Nestin and the

288

neuroblast marker Doublecortin (Dcx) in agreement with the findings from above (see Fig. S4

289

in the supplemental material).

290

Zac1 binding to the Tcf4 gene locus during neurogenesis at E15 was studied by ChIP

291

experiments with brain punches of the VZ/SVZ; thereby punches from subsequent

292

astrogliogenesis at E18 served as a negative control. Zac1 efficiently occupied the Tcf4 locus

293

at the time of neurogenesis but barely during astrogliogenesis. (Fig. 6C). Moreover,

294

transfection of dissociated cells encompassing the neocortical VZ/SVZ (E15) with increasing

295

amounts of Zac1 caused a robust increase in the activity of the composite Tcf4 reporter

296

construct (Fig. 6D).

12

297

In sum, Zac1 and Tcf4 colocalize in the VZ/SVZ during neurogenesis. At the same time, Zac1

298

occupies the Tcf4 regulatory regions and confers transactivation to a Tcf4 reporter construct.

299 300

Zac1, Tcf4 and p57Kip2 are co-regulated during brain development. The biological

301

function of Tcf4 regulation by Zac1 was addressed by the study of Tcf4-dependent target

302

genes during neurogenesis. The paternally imprinted cyclin-dependent kinase inhibitor p57Kip2

303

is an interesting candidate in this respect. Firstly, it is co-regulated with Zac1 in the context of

304

an imprinted gene network (35). Secondly, p57Kip2 plays an important role in differentiation

305

and migration of radial glia and progenitor cells during early and mid-neurogenesis (36).

306

Thirdly, Zac1, Tcf4 and p57Kip2 share a cell cycle arrest function (30, 37). In this respect, E-

307

proteins induce cell cycle arrest via p57Kip2 in the human neuroectodermal cell line SK-N-SH

308

(38).

309

Several potential E-box motifs were predicted in the distal p57Kip2 promoter region by

310

computational analysis (see Fig. S5 in the supplemental material). Following transfection of

311

C17.2 cells with a Zac1 or Tcf4 expression vector, we measured a 2-fold and 1.6-fold

312

induction of p57Kip2, respectively, as evidenced by qRT-PCR (Fig. 7A).

313

The role of endogenous Zac1 for Tcf4 and p57Kip2 expression was further studied by knock-

314

down experiments. Transfection of shRNA-Zac1, but not of scrambled shRNA, depleted Zac1

315

mRNA levels by 40 % and resulted in a reduced expression of Tcf4, and p57Kip2 (Fig. 7B and

316

C). Of note, knock-down of Tcf4 decreased the expression of p57Kip2 without altering the

317

expression of Zac1 (Fig. 7D and E).

318 319

Having shown that Zac1 regulates p57Kip2 via Tcf4 in C17.2 stem cells, we asked whether this

320

applies also to early (E11) and mid (E15) neurogenesis. Zac1 and Tcf4 were widely expressed

321

in neuronal progenitor cell populations of the caudal brain regions, the pallium and

322

prethalamic eminence at E11 and showed hereby a broad co-expression (see Fig. S6 in the 13

323

supplemental material) in accord with previous studies (5, 8, 9). On the other hand, p57Kip2

324

expression was more restricted and localized particularly to the subpallium and peduncular

325

hypothalamus in which triple positive moderately p57Kip2 expressing cells were visualized

326

(see Fig. S6 in the supplemental material).

327

In view of the limited amount of tissue available from this region for functional studies, we

328

focused on neuronal progenitors from the neocortical zone (E15), which co-express Zac1 and

329

Tcf4 (Fig. 6B). Immunohistochemistry also evidenced p57Kip2 co-expression in Tcf4 positive

330

cells (Fig. 7F).

331

Consistent with this finding, in vivo ChIP experiments revealed Tcf4 binding to the p57Kip2

332

gene at the predicted E-boxes in the distal promoter. A region devoid of E-box motifs served

333

as a negative control in this experiments (Fig. 7G and Fig. S5 in the supplemental material).

334 335

Zac1 regulates cell cycle arrest in NSCs. Recombinant pools of O4ANS cells that contained

336

either Flag-tagged Zac1 or Zac1 shRNA expression vectors showed a doubling or reduction

337

by 80% of Zac1 mRNA levels with corresponding changes in protein expression (24). Here,

338

we detected concordant changes in Tcf4 and p57Kip2 mRNA levels in proliferating and

339

differentiating O4ANS cells consistent with the role of Tcf4 as lineage-specific Zac1 target

340

gene (Fig. 8A). In contrast, expression of the isoform Tcf4-a did not change under either

341

condition and served as a negative control in these experiments.

342

Zac1 overexpression increased the number of cells in G1-phase concomitant to a decrease in

343

G2/M-phase during early time points (Fig. 8B). On the other hand, Zac1 knock-down

344

diminished the number of cells in G1-phase and led to concomitant increases in S- and G2/M-

345

phase.

346

Zac1’s role in antiproliferation was additionally investigated by cotransfection experiments in

347

primary NSCs (E15). Following 1 day of neuronal differentiation, Zac1 transfection reduced

348

the number of Ki67 positive cells consistent with a role in antiproliferation (Fig. 8C and D). 14

349

In contrast, cotransfection of a Tcf4 shRNA expression vector restored Ki67

350

immunoreactivity in Zac1 positive cells similar to the one detected in p57Kip2 shRNA

351

transfected cells.

352

Together, these findings are consistent with the hypothesis that Tcf4 and p57Kip2 are

353

downstream to Zac1 antiproliferation in NSCs.

354 355

DISCUSSION

356

The present study uncovers a co-operation between imprinted genes and the bHLH factor

357

Tcf4 in cell cycle regulation. Zac1 is induced in differentiating neural progenitors and confers

358

in a lineage-specific manner transactivation of Tcf4. Subsequently, Tcf4 drives upregulation

359

of the cyclin-dependent kinase inhibitor p57Kip2, a critical mediator of G1 arrest function

360

during neurodevelopment.

361 362

Expression profiling in C17.2 NSCs identified Tcf4 as potential Zac1 target gene. Given

363

Zac1’s expression pattern in the developing nervous system (8, 9), we focused on Zac1-

364

dependent Tcf4 regulation in ESCs and NSCs. Although Zac1 is expressed in both glia and

365

neurons, upregulation of Tcf4 was restricted to neuronal differentiation under in vitro and in

366

vivo conditions. Thereby, Zac1 coordinately occupied the proximal promoter and first intron

367

of Tcf4 and these two elements together conferred synergistic transactivation of the Tcf4 gene.

368

The first intron did not fulfill, however, a role as an autonomous Zac1 enhancer.

369 370

Tcf4 is known to control proliferation, migration and differentiation of distinct neuronal

371

progenitors, pointing to the importance of this factor for brain development (39).

372

Mechanistically, increased Tcf4 expression might counteract the association of its natural

373

inhibitors, the Id proteins, with the proneural factors (40), or the lineage-specific repressor

374

function of Hes proteins (41, 42). 15

375

Alternatively, we suggest that Zac1 transactivation of Tcf4 contributes to enhanced p57Kip2

376

expression in differentiating neuronal precursors as evidenced by overexpression and knock-

377

down experiments in C17.2 and primary NSCs. Cyclin-dependent kinase inhibitors are key

378

effectors of cell cycle arrest during differentiation. In support of this view, we detected

379

colocalization between Zac1, Tcf4 and p57Kip2 during early and late neurogenesis and showed

380

that in the neocortical ventricular zone Tcf4 directly binds to E-box motifs present in the

381

p57Kip2 promoter.

382

The p57Kip2 gene regulates cell cycle dynamics of radial glia and progenitor cells, controls

383

precursor pool size, neuronal differentiation, cortical size and laminar patterning (36, 43, 44).

384

Because cell cycle exit of precursor cells correlates with their laminar destination, neuronal

385

fate is coupled to the cell cycle machinery. Hence, we propose that Zac1 might finetune the

386

timing and fate of neuronal differentiation by enhancing p57Kip2 expression via Tcf4.

387 388

In support of our present findings, Zac1 and p57Kip2 have been hypothesized to be connected

389

in the context of an imprinting gene network (IGN) that regulates early mouse development

390

(35). The mechanisms underlying such connectivity remained, however, incompletely

391

understood. In this regard, we recently showed that Zac1 directly represses the paternally

392

imprinted gene Rasgrf1 leading to an inhibition of glucose-stimulated insulin secretion in

393

pancreatic β-cells (23). Here we show that imprinted genes can be also connected via

394

regulatory networks that involve proneural factors such as Tcf4. On the other hand, the

395

proneural genes Neurogenin 2 and Archaete scute homolog 1 have been found to regulate the

396

imprinted Dlk1-Gtl2 locus in the developing telencephalon (45). Overall, we propose that

397

such reciprocal interactions between imprinted and proneural genes can critically contribute to

398

progenitor cell fate decisions and early neural development.

399

16

400

ACKNOWLEDGEMENT

401

The help of A. Patchev and A. Varga in animal experiments is appreciated. The NSC lines

402

and the Tcf4 antibody were generously provided by A. Smith (Cambridge University, UK)

403

and A. Herbst (University of Munich, Germany), respectively. The pCDEF3.Flag-Tcf4-b

404

expression vector was kindly gifted by S. Itoh (University of Tsukuba, Japan). This work was

405

supported by the Deutsche Forschungsgemeinschaft (SP 386/5-1 to D.S.) The authors declare

406

no competing financial interests.

407

17

408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455

REFERENCES 1. Cai L, Morrow EM, Cepko CL. 2000. Misexpression of basic helix-loop-helix genes in the murine cerebral cortex affects cell fate choices and neuronal survival. Development 127:3021–3030. 2. Ge W, He F, Kim KJ, Blanchi B, Coskun V, Nguyen L, Wu X, Zhao J, Heng JI, Martinowich K, Tao J, Wu H, Castro D, Sobeih MM, Corfas G, Gleeson JG, Greenberg ME, Guillemot F, Sun YE. 2006. Coupling of cell migration with neurogenesis by proneural bHLH factors. Proc. Natl. Acad. Sci. U.S.A. 103:1319–1324. 3. Murre C, McCaw PS, Baltimore D. 1989. A new DNA binding and dimerization motif in immunoglobulin enhancer binding, daughterless, MyoD, and myc proteins. Cell 56:777–783. 4. Bertrand N, Castro DS, Guillemot F. 2002. Proneural genes and the specification of neural cell types. Nat. Rev. Neurosci. 3:517–530. 5. Bhattacharya A, Baker NE. 2011. A network of broadly expressed HLH genes regulates tissue-specific cell fates. Cell 147:881–892. 6. Sun Y, Nadal-Vicens M, Misono S, Lin MZ, Zubiaga A, Hua X, Fan G, Greenberg ME. 2001. Neurogenin promotes neurogenesis and inhibits glial differentiation by independent mechanisms. Cell 104:365–376. 7. Ross SE, Greenberg ME, Stiles CD. 2003. Basic helix-loop-helix factors in cortical development. Neuron 39:13–25. 8. Valente T, Auladell C. 2001. Expression pattern of Zac1 mouse gene, a new zinc-finger protein that regulates apoptosis and cellular cycle arrest, in both adult brain and along development. Mech. Dev. 108:207–211. 9. Alam S, Zinyk D, Ma L, Schuurmans C. 2005. Members of the Plag gene family are expressed in complementary and overlapping regions in the developing murine nervous system. Dev. Dyn. 234:772–782. 10. Chung S, Marzban H, Aldinger K, Dixit R, Millen K, Schuurmans C, Hawkes R. 2011. Zac1 plays a key role in the development of specific neuronal subsets in the mouse cerebellum. Neural Dev 6:25. doi:10.1186/1749-8104-6-25. 11. Ciani E, Frenquelli M, Contestabile A. 2003. Developmental expression of the cell cycle and apoptosis controlling gene, Lot1, in the rat cerebellum and in cultures of cerebellar granule cells. Brain Res. Dev. Brain Res. 142:193–202. 12. Ma L, Cantrup R, Varrault A, Colak D, Klenin N, Götz M, McFarlane S, Journot L, Schuurmans C. 2007. Zac1 functions through TGFbetaII to negatively regulate cell number in the developing retina. Neural Dev 2:11. doi:10.1186/1749-8104-2-11.

18

456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505

13. Ma L, Hocking JC, Hehr CL, Schuurmans C, McFarlane S. 2007. Zac1 promotes a Müller glial cell fate and interferes with retinal ganglion cell differentiation in Xenopus retina. Dev. Dyn. 236:192–202. 14. Constância M, Kelsey G, Reik W. 2004. Resourceful imprinting. Nature 432:53–57. 15. Varrault A, Ciani E, Apiou F, Bilanges B, Hoffmann A, Pantaloni C, Bockaert J, Spengler D, Journot L. 1998. hZAC encodes a zinc finger protein with antiproliferative properties and maps to a chromosomal region frequently lost in cancer. Proc. Natl. Acad. Sci. U.S.A. 95:8835–8840. 16. Bilanges B, Varrault A, Mazumdar A, Pantaloni C, Hoffmann A, Bockaert J, Spengler D, Journot L. 2001. Alternative splicing of the imprinted candidate tumor suppressor gene ZAC regulates its antiproliferative and DNA binding activities. Oncogene 20:1246–1253. 17. Hoffmann A, Ciani E, Boeckardt J, Holsboer F, Journot L, Spengler D. 2003. Transcriptional activities of the zinc finger protein Zac are differentially controlled by DNA binding. Mol. Cell. Biol. 23:988–1003. 18. Ciani E, Hoffmann A, Schmidt P, Journot L, Spengler D. 1999. Induction of the PAC1-R (PACAP-type I receptor) gene by p53 and Zac. Brain Res. Mol. Brain Res. 69:290–294. 19. Rodríguez-Henche N, Jamen F, Leroy C, Bockaert J, Brabet P. 2002. Transcription of the mouse PAC1 receptor gene: cell-specific expression and regulation by Zac1. Biochim. Biophys. Acta 1576:157–162. 20. Spengler D, Villalba M, Hoffmann A, Pantaloni C, Houssami S, Bockaert J, Journot L. 1997. Regulation of apoptosis and cell cycle arrest by Zac1, a novel zinc finger protein expressed in the pituitary gland and the brain. EMBO J. 16:2814–2825. 21. Barz T, Hoffmann A, Panhuysen M, Spengler D. 2006. Peroxisome proliferatoractivated receptor gamma is a Zac target gene mediating Zac antiproliferation. Cancer Res. 66:11975–11982. 22. Liu P, Hsieh T, Liu S, Chang Y, Lin W, Wang W, Huang S. 2011. Zac1, an Sp1-like protein, regulates human p21(WAF1/Cip1) gene expression in HeLa cells. Exp. Cell Res. 317:2925–2937. 23. Hoffmann A, Spengler D. 2012. Transient neonatal diabetes mellitus gene Zac1 impairs insulin secretion in mice through Rasgrf1. Mol. Cell. Biol. 32:2549–2560. 24. Schmidt-Edelkraut U, Hoffmann A, Daniel G, Spengler D. 2013. Zac1 regulates astroglial differentiation of neural stem cells through Socs3. Stem Cells 31:1621–1632. 25. Hoffmann A, Spengler D. 2008. A new coactivator function for Zac1's C2H2 zinc finger DNA-binding domain in selectively controlling PCAF activity. Mol. Cell. Biol. 28:6078– 6093.

19

506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554

26. Martín-Ibáñez R, Urbán N, Sergent-Tanguy S, Pineda JR, Garrido-Clua N, Alberch J, Canals JM. 2007. Interplay of leukemia inhibitory factor and retinoic acid on neural differentiation of mouse embryonic stem cells. J. Neurosci. Res. 85:2686–2701. 27. Hoffmann A, Barz T, Spengler D. 2006. Multitasking C2H2 zinc fingers link Zac DNA binding to coordinated regulation of p300-histone acetyltransferase activity. Mol. Cell. Biol. 26:5544–5557. 28. Tanaka A, Itoh F, Itoh S, Kato M. 2009. TAL1/SCL relieves the E2-2-mediated repression of VEGFR2 promoter activity. J. Biochem. 145:129–135. 29. Murgatroyd C, Hoffmann A, Spengler D. 2012. In vivo ChIP for the analysis of microdissected tissue samples. Methods Mol. Biol. 809:135–148. 30. Herbst A, Bommer GT, Kriegl L, Jung A, Behrens A, Csanadi E, Gerhard M, Bolz C, Riesenberg R, Zimmermann W, Dietmaier W, Wolf I, Brabletz T, Göke B, Kolligs FT. 2009. ITF-2 is disrupted via allelic loss of chromosome 18q21, and ITF-2B expression is lost at the adenoma-carcinoma transition. Gastroenterology 137:639-648. 31. Huang SM, Stallcup MR. 2000. Mouse Zac1, a transcriptional coactivator and repressor for nuclear receptors. Mol. Cell. Biol. 20:1855–1867. 32. Kamiya D, Banno S, Sasai N, Ohgushi M, Inomata H, Watanabe K, Kawada M, Yakura R, Kiyonari H, Nakao K, Jakt LM, Nishikawa S, Sasai Y. 2011. Intrinsic transition of embryonic stem-cell differentiation into neural progenitors. Nature 470:503– 509. 33. Guan K, Chang H, Rolletschek A, Wobus AM. 2001. Embryonic stem cell-derived neurogenesis. Retinoic acid induction and lineage selection of neuronal cells. Cell Tissue Res. 305:171–176. 34. Ahmed S. 2009. The culture of neural stem cells. J. Cell. Biochem. 106:1–6. 35. Varrault A, Gueydan C, Delalbre A, Bellmann A, Houssami S, Aknin C, Severac D, Chotard L, Kahli M, Le Digarcher A, Pavlidis P, Journot L. 2006. Zac1 regulates an imprinted gene network critically involved in the control of embryonic growth. Dev. Cell 11:711–722. 36. Tury A, Mairet-Coello G, DiCicco-Bloom E. 2011. The cyclin-dependent kinase inhibitor p57Kip2 regulates cell cycle exit, differentiation, and migration of embryonic cerebral cortical precursors. Cereb. Cortex 21:1840–1856. 37. Matsuoka S, Edwards MC, Bai C, Parker S, Zhang P, Baldini A, Harper JW, Elledge SJ. 1995. p57KIP2, a structurally distinct member of the p21CIP1 Cdk inhibitor family, is a candidate tumor suppressor gene. Genes Dev. 9:650–662. 38. Rothschild G, Zhao X, Iavarone A, Lasorella A. 2006. E Proteins and Id2 converge on p57Kip2 to regulate cell cycle in neural cells. Mol. Cell. Biol. 26:4351–436.1

20

555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585

39. Sobrado VR, Moreno-Bueno G, Cubillo E, Holt LJ, Nieto MA, Portillo F, Cano A. 2009. The class I bHLH factors E2-2A and E2-2B regulate EMT. J. Cell. Sci. 122:1014– 1024. 40. Jung S, Park R, Kim S, Jeon Y, Ham D, Jung M, Kim S, Lee Y, Park C, Suh-Kim H. 2010. Id proteins facilitate self-renewal and proliferation of neural stem cells. Stem Cells Dev. 19:831–841. 41. Nakamura Y, Sakakibara Si, Miyata T, Ogawa M, Shimazaki T, Weiss S, Kageyama R, Okano H. 2000. The bHLH gene hes1 as a repressor of the neuronal commitment of CNS stem cells. J. Neurosci. 20:283–293. 42. Holmberg J, Hansson E, Malewicz M, Sandberg M, Perlmann T, Lendahl U, Muhr J. 2008. SoxB1 transcription factors and Notch signaling use distinct mechanisms to regulate proneural gene function and neural progenitor differentiation. Development 135:1843–1851. 43. Mairet-Coello G, Tury A, van Buskirk E, Robinson K, Genestine M, DiCicco-Bloom E. 2012. p57(KIP2) regulates radial glia and intermediate precursor cell cycle dynamics and lower layer neurogenesis in developing cerebral cortex. Development 139:475–487. 44. Tury A, Mairet-Coello G, DiCicco-Bloom E. 2012. The multiple roles of the cyclindependent kinase inhibitory protein p57(KIP2) in cerebral cortical neurogenesis. Dev Neurobiol 72:821–842. 45. Seibt J, Armant O, Le Digarcher A, Castro D, Ramesh V, Journot L, Guillemot F, Vanderhaeghen P, Bouschet T. 2012. Expression at the imprinted dlk1-gtl2 locus is regulated by proneural genes in the developing telencephalon. PLoS ONE 7:e48675. doi:10.1371/journal.pone.0048675.

21

586 587 588

FIGURE LEGENDS

589

Zac1 transactivates Tcf4. (A) C17.2 neural stem cells harbouring Tet-off regulated Zac1

590

expression were grown in the presence or absence of tetracycline (Tc). Zac1 and Tcf4

591

expression following Tc removal for the indicated periods were measured by qRT-PCR. (B)

592

Parental C17.2 cells were transfected with the indicated amount of Zac1 vector and

593

expression of Tcf4-a and Tcf4-b isoforms was detected by qRT-PCR analysis. (C)

594

Immunoblot analysis (30 μg whole cell extract, WCE) of Zac1, Tcf4-b and β-actin expression

595

following Zac1 transfection (100 ng). (D) Expression of Tcf4 in C17.2 cells was measured by

596

qRT-PCR following transfection of the indicated Zac1 constructs (100 ng each). (E) Protein

597

levels of Tcf4 and the different Zac1 constructs (100 ng each) were assessed by immunoblot

598

analysis (30 μg WCE). The asterisk indicates the ΔLPR specific signal. (F) Schematic

599

representation of the Tcf4 promoter and first intron. The 2 first alternative exons I and exon II

600

are represented by grey boxes and Roman numbers. Putative Zac1 binding sites are depicted

601

as black squares and triangles. Underlined regions and numbers indicate the genomic fractions

602

amplified in ChIP experiments with respect to the transcriptional start site (+1). (G) ChIP

603

assays revealed Zac1 occupancy at the Tcf4 proximal promoter and first intron, but not at the

604

promoter of the isoform a (Tcf4-a). Representative results from three (C and E) or means ±

605

SD from four (B and G) or six (A and D) independent experiments are shown. * P < 0.05 and

606

** P < 0.01.

Figure 1

607 608

Figure 2

609

Zac1 and Tcf4 are upregulated during neuronal differentiation of mouse embryonic

610

stem cells. (A) Bright-light microscopy of 46C ESCs during neuronal differentiation

611

following leukemia inhibitory factor (Lif) withdrawal for the indicated number of days. Scale

612

bar, 100 μm. (B) Expression of the pluripotency marker Oct4 and (C) neuronal marker Tuj1 22

613

following Lif withdrawal for the indicated number of days was measured by qRT-PCR. (D)

614

Time course analysis of Zac1 and Tcf4 mRNA expression upon Lif withdrawal as detected by

615

qRT-PCR. (E) Immunoblot analysis (100 μg WCE) of ESCs grown for the indicated number

616

of days in the absence of Lif revealed co-regulation of Zac1 and Tcf4. Representative results

617

from three (A and E) or means ± SD from four (B and C) or six (D) independent experiments

618

are shown.

619 620

Figure 3

621

Coordinated Zac1 binding regulates Tcf4 expression. (A) ChIP assays in 46C ESCs with a

622

Zac1 antibody were conducted in the presence (+) or absence (-) of Lif for the indicated

623

number of days. In the presence of Lif, Zac1 binding is barely detectable at the Tcf4 locus,

624

whereas upon Lif withdrawal a strong association between Zac1 and the Tcf4 proximal

625

promoter (-89) and first intron (+1933) was detected. (B) Schematic representation of the Tcf4

626

5’ end and the various Tcf4 promoter fragments used in reporter assays. (C) Reporter assays

627

(note the logarithmic ordinate) with the different Tcf4 promoter fragments (500 ng each) in

628

46C ESCs grown for 3 days in the absence of Lif. Reverse orientation of Tcf4 promoter

629

constructs strongly reduced luciferase activity. Luciferase activity of the parent vector was set

630

to 1. (D) Promoter reporter assays in 46C ESCs. Cells were grown for 3 days in the absence

631

of Lif and subsequently cotransfected with the indicated Tcf4 promoter constructs (500 ng

632

each) and increasing amounts of Zac1 and ZF7mt (10 and 25 ng each) or the empty vector,

633

which was set to 1. Synergistic Zac1 transactivation depended on the joint promoter and

634

intronic regions. (E) Activity of the first Tcf4 intron was measured by reporter assays in 46C

635

cells. Cells were grown for 3 days in the absence of Lif and subsequently cotransfected with

636

the indicated Tcf4 constructs (500 ng each) and Zac1 (25 ng) or the empty vector, which was

637

set to 1. The Tcf4 first intron does not behave as an autonomous Zac1 enhancer when fused to

638

the thymidine kinase (TK) promoter and prevented transactivation when inserted in the 23

639

reverse orientation adjacent to the Tcf4 promoter region. The arrow illustrates the orientation

640

of the intron. Means ± SD from four (A and C) or five (D and E) independent experiments are

641

shown. * P < 0.05 and ** P < 0.01.

642 643

Figure 4

644

Zac1 associates with active chromatin at the Tcf4 locus during neuronal differentiation.

645

(A) Neuronal and astroglial differentiation of O4ANS cells.

646

expression was measured by qRT-PCR across 1 week of differentiation. Note the separate

647

ordinate for Zac1 and Tcf4 regulation. (B-D) ChIP assays in O4ANS cells following 4 days of

648

neuronal or astroglial differentiation using antibodies against Zac1 (B), acH3 (C), and

649

H3K9me2 (D). (E and F) Sequential ChIP analysis following neuronal or astroglial

650

differentiation for 4 days. During neuronal differentiation, Zac1 associated with chromatin

651

containing active marks (acH3/Zac1) at the Tcf4 locus, whereas association with repressive

652

chromatin marks (H3K9me2/Zac1) decreased. Means ± SD from four (C to F) or six (A and

653

B) independent experiments are shown. * P < 0.05 and ** P < 0.01.

Zac1 and Tcf4 mRNA

654 655

Figure 5

656

Zac1 regulates Tcf4 expression during neuronal differentiation of primary NSCs. (A)

657

Immunocytochemistry of NSCs maintained under growth conditions (NSC) or following

658

neuronal or astroglial differentiation for 6 days. Cells were subjected to indirect

659

immunofluorescence with antibodies against Nestin (green), Tuj1 (green), Gfap (green), Zac1

660

(red) or Tcf4 (c, g, k: red and d, h, l: green). Nuclei were stained with DAPI (blue). Scale bar

661

50 μm. (B) Quantitative analysis of the NSC specific marker Nestin, and the neuronal and

662

astroglial markers Tuj1 and Gfap, respectively. Cells were kept under growth conditions or in

663

the appropriate differentiation medium for 6 days. Unknown cellular phenotypes are indicated

664

as non-defined (n/d). Mean of five analyzed images of the type shown in (A). (C) Time 24

665

course analysis of Zac1 and Tcf4 mRNA expression during neuronal and astroglial

666

differentiation as measured by qRT-PCR. (D) Immunoblot analysis (100 μg WCE) of Zac1

667

and Tcf4 expression during neuronal or astroglial differentiation. (E) Promoter reporter assays

668

in undifferentiated NSCs or following neuronal or astroglial differentiation for 6 days. Cells

669

were cotransfected with the composite Tcf4 promoter construct (500 ng) and the indicated

670

amounts of Zac1 or the empty vector, which was set to 1. Highest induction occurred

671

following neuronal differentiation. (F) ChIP analysis following neuronal or astroglial

672

differentiation for 6 days evidenced Zac1 occupancy at the Tcf4 promoter and first intron

673

solely during the former. Representative results from three (A and D) or means ± SD from

674

four (F) or five (C and E) independent experiments are shown. * P < 0.05 and ** P < 0.01.

675 676

Figure 6

677

Zac1 and Tcf4 colocalize in the neocortical ventricular zone. (A and B) Immunohisto-

678

chemistry of mouse brain sections. Upper panel, sections from E11 (A) and E15 (B) were

679

stained with antibodies for Zac1 (red) and Tcf4 (green). Representative images of the

680

subventricular and ventricular zones are shown. Nuclei were stained with DAPI (blue). Scale

681

bar 100 μm. Lower panels show regions with higher magnification corroborating

682

colocalization of Zac1 and Tcf4. Scale bar 40 μm. (C) ChIP analysis of Zac1 occupancy at the

683

Tcf4 promoter in the neocortical VZ/SVZ during neurogenesis (E15) or astrogliogenesis

684

(E18). (D) Induction of the composite Tcf4 promoter construct (500 ng) following

685

cotransfection of increasing amounts of Zac1 or the empty vector, which was set to 1, in cells

686

dissected from the neocortical VZ/SVZ at E15. Representative results from three (A and B) or

687

means ± SD from five (C and D) independent experiments are shown. * P < 0.05 and ** P

Zac1 regulates cell cycle arrest in neuronal progenitors via Tcf4.

Imprinted genes play a critical role in brain development and mental health, although the underlying molecular and cellular mechanisms remain incomple...
3MB Sizes 1 Downloads 0 Views