1 Prolactin Receptor Signaling Regulates a

0 downloads 0 Views 2MB Size Report
performed using pheatmap package (1.0.8) within R, and VennPlex (24) was used to create the. 145. Venn diagrams and determine overlapping genes. 146.
1

Prolactin Receptor Signaling Regulates a Pregnancy-Specific Transcriptional Program in

2

Mouse Islets

3 4

Authors: Mark E. Pepin1, Adam R. Wende1,2,4 and Ronadip R. Banerjee3,4*

5

Affiliations: 1Department of Biomedical Engineering, University of Alabama at Birmingham,

6

Birmingham, AL; 2Department of Pathology, University of Alabama at Birmingham School of

7

Medicine, Birmingham, AL; 3Department of Medicine, Division of Endocrinology, University of

8

Alabama at Birmingham School of Medicine, Birmingham, AL and the 4University of Alabama at

9

Birmingham Comprehensive Diabetes Center, Birmingham, AL

10

*Corresponding author: [email protected]

11 12

Short title: PRLR Regulates Islet Transcriptome in Pregnancy

13

Keywords: Gestational diabetes, prolactin receptor, pancreatic islets, beta-cell proliferation,

14

transcriptional regulation

15 16

Disclosures: The authors have nothing to disclose.

17

Contact information:

18 19 20 21 22 23 24 25

Ronadip R. Banerjee, M.D., Ph.D. Department of Medicine, Division of Endocrinology, Diabetes and Metabolism University of Alabama School of Medicine, Birmingham, AL Boshell Diabetes Building 730 1808 7th Avenue South Birmingham, AL 35294 lab: (205) 934-6278 email: [email protected]

26

1

27

Abstract

28

Pancreatic β-cells undergo profound hyperplasia during pregnancy to maintain maternal

29

euglycemia. Failure to reprogram β-cells into a more replicative state has been found to underlie

30

susceptibility to gestational diabetes mellitus (GDM). We recently identified a requirement for

31

prolactin receptor (PRLR) signaling in the metabolic adaptations to pregnancy, where mice lacking

32

β-cell PRLR (βPRLRKO) exhibit a metabolic phenotype consistent with GDM. However, the

33

underlying transcriptional program that is responsible for the PRLR-dependent metabolic

34

adaptations during gestation remains incompletely understood. To identify PRLR signaling gene

35

regulatory networks and target genes within β-cells during pregnancy, we performed a

36

transcriptomic analysis of pancreatic islets isolated from either βPRLRKO mice or littermate

37

controls in late gestation. Gene set enrichment analysis identified Forkhead box protein M1

38

(Foxm1) and polycomb repressor complex 2 (PRC2) subunits, Suz12 and Ezh2, as novel candidate

39

regulators of PRLR-dependent β-cell adaptation. GO-term pathway enrichment revealed both

40

established and novel PRLR signaling target genes that together describe a state of increased

41

cellular metabolism and/or proliferation. In contrast to the requirement for β-cell PRLR signaling

42

in maintaining euglycemia during pregnancy, PRLR target genes were not induced following high-

43

fat-diet feeding. Altogether, the current study expands our understanding of which transcriptional

44

regulators and networks mediate gene expression required for islet adaptation during pregnancy.

45

The current work also supports the presence of pregnancy-specific adaptive mechanisms distinct

46

from those activated by nutritional stress.

47

2

48

Introduction

49

Gestational diabetes mellitus (GDM) is a metabolic disorder that emerges only during

50

pregnancy, yet it presents serious health outcomes to mother and offspring even decades after birth

51

(1). As with other forms of diabetes mellitus, a relative deficiency of functional β-cells is a major

52

factor in the pathogenesis of GDM (2). Controversy exists, however, regarding the underlying

53

etiology of GDM when considering its relationship with obesity and nutrition. While glycemic

54

management of GDM can be achieved by nutritional therapy (3), only approximately half of the

55

cases can be attributed to the metabolic effects of obesity (4,5). Furthermore, the risk of both

56

recurrent GDM and type 2 diabetes mellitus is exceedingly high and occurs independently of

57

obesity (6).

58

Unlike various other metabolic stressors that stimulate β-cell expansion, the physiologic

59

adaptation to pregnancy comprises both an interaction between the mother and the feto-placental

60

unit, and a rapidly fluctuating hormonal milieu that induces stereotypic changes in systemic

61

metabolism spanning gestation and postpartum (2). While the proliferative phenotype and

62

augmentation of β-cells seen throughout pregnancy are rapidly reversed postpartum, the adverse

63

consequences of disturbed maternal glucose homeostasis impact both mother and developing

64

offspring during pregnancy and long-term (1,7). These unique features of pregnancy and GDM

65

form the premise for additional studies into the genetic relationship between GDM and type 2

66

diabetes and their shared or distinct pathophysiologic mechanisms (8).

67

Amongst the diverse extracellular and intracellular mechanisms known to contribute to

68

gestational β-cell adaptation, the lactogens are perhaps the most well-studied (9). Most mammals,

69

including rodents and primates, produce pituitary-derived prolactin and placental lactogens, which

70

both signal through the prolactin receptor (PRLR) (9-12). We and others have established a critical

3

71

role for PRLR signaling in rodent gestational β-cell proliferation (13), with the lack of PRLR

72

expression in β-cells resulting in a failure of islets to proliferate during gestation (14). Conversely,

73

β-cell specific overexpression of placental lactogens increases pancreatic islet proliferation and

74

mass (15). Together these studies illustrate that increases in circulating lactogens during pregnancy

75

are essential and sufficient to orchestrate key components of metabolic adaptations to pregnancy.

76

Downstream targets of PRLR signaling that contribute to gestational proliferation include

77

tryptophan hydroxylase 1 (Tph1), the rate limiting enzyme for islet serotonin synthesis (16),

78

transcription factors such as Foxm1 (17) and MafB (14), osteoprotegerin (Tnfrsf11b), menin

79

(Men1), as well as cyclins, cyclin-dependent kinases, cell cycle inhibitors and Prlr itself (14,16,18-

80

20). However, a global assessment of how PRLR signaling regulates β-cell transcription during

81

gestation is lacking.

82

To address this question, in the current study we examined how loss of PRLR signaling within

83

β-cells alters pancreatic islet gene expression during pregnancy. Our findings identify both known

84

and novel PRLR signaling targets and candidate upstream regulators of β-cell transcription.

85

Additionally, we found that the transcriptomic program driven by PRLR signaling and its target

86

genes is distinct from the compensatory response of islets to high-fat diet feeding, indicating

87

pregnancy-specificity of key downstream mediators of this pathway.

88 89

Materials and Methods

90

Mouse Strains, Husbandry, Breeding and Experimentation. Female transgenic mice lacking

91

PRLR within pancreatic -cells (PRLRKO) were used in the current study, as previously

92

developed and described (14). For gestational studies, 8-week-old virgin females were mated with

93

C57Bl6/J males and vaginal plugs scored as GD0.5. Plugged females were single-housed for the

4

94

duration of pregnancy. For nutritional studies, mice were maintained on standard chow (4.7 kcal%,

95

#7917, Envigo, Indianapolis, IN), then given ad libitum access to standard chow or high-fat diet

96

(HFD, 58.0 kcal% fat, #D12492 Research Diets, New Brunswick, NJ) for either 4 or 12 weeks.

97

Mice were group-housed (5/cage) on a 12:12-h light-dark cycle at 25 ± 1°C and constant humidity

98

with free access to food and water except as noted. All procedures involving mice were approved

99

and conducted in accordance with the University of Alabama at Birmingham Institutional Animal

100

Care and Use Committee (IACUC).

101 102

Glucose Tolerance Testing. Glucose tolerance tests (GTTs) were performed by intraperitoneal

103

injection of glucose (1 g/kg (Chow-fed) or 2 g/kg (HF-fed) body weight, (glucose stock solution

104

of 20% wt/vol D-glucose [Sigma] in 0.9% saline) to 16-hour overnight fasted mice. Blood glucose

105

was determined using Bayer Contour Glucometer.

106 107

Islet Isolation and Culture. Islets were isolated from donor mice using retrograde perfusion of

108

the pancreatic duct with collagenase and purified using density centrifugation as previously

109

described (14). Donor mice included PRLRKO and littermate controls or C57Bl6/J mice either

110

from virgin females (nonpregnant) or at gestational day (GD) 16.5 (pregnant). For in vitro

111

experiments, hand-picked islets were cultured overnight in RPMI supplemented with 10% FBS.

112

Subsequently, islets from individual mice were picked and divided equally into fresh media

113

supplemented with recombinant mouse prolactin at a final concentration of 500 ng/ml (R&D

114

Systems, Minneapolis, MN), or an equal volume of vehicle. Culture media was changed daily.

115

5

116

Microarray Analysis. Microarray-based mRNA quantification was performed using the

117

Affymetrix GeneChip Mouse 2.0 Array (ThermoFisher, Waltham, MA US). Hybridization and

118

microarray scanning were performed by the Stanford Functional Genomics Facility (Stanford,

119

CA). All microarray data have been deposited within the Gene Expression Omnibus repository

120

(GSE118134). Subsequent microarray annotation, normalization, and differential expression

121

analysis were completed in the R statistical computing environment (version 3.4.3). Briefly, raw

122

microarray data across all samples were compiled and corrected using the robust multi-array

123

average (RMA) of background-adjusted and quantile-normalized microarray expression data using

124

R package Oligo (1.42.0) as previously described (21). Gene annotations were then generated

125

using the MoGene-2.0 Transcript Cluster (mogene20sttranscriptcluster.db). Once expression data

126

were examined for RNA quality, the R package limma (version 3.34.9) was used to generate

127

differential gene expression using generalized linear modelling (KO versus WT) with a Benjamini-

128

Hochberg (B-H) P-value adjustment (22). Due to sample size limitations (n = 3), dispersion

129

estimates were first determined via maximum likelihood, assuming that genes of similar average

130

expression strength possess similar dispersion, as previously described (23). Gene-wise dispersion

131

estimates were then shrunken according to the empirical Bayes approach, providing normalized

132

count data for genes proportional to both the dispersion and sample size. Differential expression

133

was represented as log2 (fold-change) for each annotated gene. Statistical significance was

134

estimated at B-H adjusted P-value (Q-value) < 0.05.

135 136

Bioinformatics and Data Visualization. Details of the R coding scripts and other bioinformatics

137

tools used in the current study are available on GitHub for public reference as supplemental

138

materials (https://github.com/mepepin/bPRLRKO_Pepin.2018). Functional gene set enrichment

6

139

analyses (GSEA) were performed using the interactive web-based platform Enrichr (24) using the

140

Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway database. Gene network analysis

141

and visualization were performed using Cytoscape (3.7.0), with literature-curated pathway

142

enrichment was achieved using QIAGEN’s Ingenuity Pathway Analysis (IPA®, QIAGEN

143

Redwood City, www.qiagen/ingenuity) on differentially expressed genes (DEGs) using a low-

144

stringency statistical threshold of P < 0.05. Heatmap and hierarchical clustering generation was

145

performed using pheatmap package (1.0.8) within R, and VennPlex (24) was used to create the

146

Venn diagrams and determine overlapping genes.

147 148

RNA Isolation for Microarray and Quantitative Real-Time PCR Studies. Total RNA was

149

isolated using TRIzol and reverse transcribed using the Ambion Retroscript Kit according to

150

manufacturers’ instructions. Differential expression by quantitative PCR (qPCR) were performed

151

with FastStart Universal Probe Master (Roche) and Thermofisher TaqMan™ assays (Waltham,

152

MA, Supplemental Table S1 (25)) on BioRad CFX96 Touch™ (Hercules, CA) machine. Assays

153

were performed in technical replicates and normalized to mouse actin (Actb) as a reference

154

standard.

155 156

Statistics. To determine the significance in overlap between the Venn Diagram of differentially-

157

expressed genes identified in our study and those of previous publications, we used a

158

hypergeometric probability distribution with Bonferroni correction to generate an adjusted P-

159

value. For all targets, an unpaired two-tailed student’s t-test was performed using Tukey’s

160

correction for multiple comparisons. Statistical significance was concluded based on a false

161

discovery-adjusted P-value (Q) < 0.05. Functional and network GSEA, along with curated

7

162

literature-supported candidate upstream regulators, were performed using QIAGEN’s Ingenuity

163

Pathway Analysis (IPA®, QIAGEN Redwood City) unless otherwise specified. GTTs were

164

considered significant for repeated measures ANOVA of P-value < 0.05.

165 166

Results

167

Differential gene expression in pancreatic islets from PRLRKO mice during gestation.

168

To identify genes regulated by prolactin receptor (PRLR) signaling in maternal pancreatic

169

islets during pregnancy, we examined global transcriptomic profiles at late gestation using

170

commercial oligonucleotide microarrays (as described in Methods). RNA purified from pancreatic

171

islets isolated from gestational day (GD) 16.5 PRLRKO mice (Prlrf/f; RIP-Cre) and littermate

172

controls (Prlrf/f and Prlrf/+) identified 2,542 differentially-expressed genes (DEGs) at P < 0.05; of

173

these, 70 reached a Bonferroni-adjusted P-value (Q) < 0.05 (Supplemental Table S2 see (25)).

174

Unsupervised principal components analysis (PCA) revealed that the two eigenvectors most

175

responsible for sample variance (51.7%) were sufficient to describe a separation between

176

PRLRKO mice and controls, supporting that prolactin receptor knockout confers a true and

177

measurable shift in global gene expression (Supplemental Figure S1; (26)). Gene set enrichment

178

analysis identified Tryptophan/Serotonin metabolism and Prolactin signaling as enriched

179

pathways (Fig. 1a), both of which are associated with the metabolic adaptation to pregnancy

180

(13,16,27,28). We then compared the DEGs reaching Q-value significance with published studies

181

reporting DEGs in murine islets during pregnancy in wild-type mice. We incorporated microarray

182

data of islet gene expression during pregnancy compared to nonpregnant controls (Schraenen et

183

al.; GD9.5 (29), Layden et al.; GD12.5. (30), and Rieck et al. GD14.5 (31)), and generated a Venn

184

diagram of these studies with the 70 DEGs found in PRLRKO mice. This comparison revealed

8

185

a significant overlap of 26 DEGs (hypergeometric P = 2.8 x 10-61) (Fig. 1b). Amongst these were

186

genes responsible for enriching pathways in Fig. 1a including tryptophan metabolism (Tph1,

187

Tph2), and prolactin receptor signaling (Prlr), as well as diverse cellular physiologic processes

188

(Table 1 and (25)).

189

9

190 Table 1. Genes induced by pregnancy which require β-cell PRLR signaling. Log2 -cell Gene Description q-val. (Fold-Change) physiology Cldn8 claudin 8 -4.7 0.0009 Ivd isovaleryl coenzyme A dehydrogenase -2.4 0.002 (32) Tph1 tryptophan hydroxylase 1 -5.3 0.002 (14,16,33) Matn2 matrilin 2 -3.1 0.002 (32) cytokine inducible SH2-containing Cish -2.4 0.004 (34) protein Tph2 tryptophan hydroxylase 2 -2.6 0.004 (16) Socs2 suppressor of cytokine signaling 2 -1.7 0.005 (35) insulin-like growth factor binding Igfbp5 -2.6 0.005 (36) protein 5 B3galnt1 UDP-GalNAc -2.5 0.008 solute carrier family 2 Slc2a13 -1.4 0.008 (facilitated glucose transporter) apolipoprotein B mRNA editing Apobec1 -1.6 0.011 enzyme, catalytic polypeptide 1 Znrf2 zinc and ring finger 2 -1.3 0.013 tumor necrosis factor receptor Tnfrsf11b -2.2 0.019 (37) superfamily Fmo1 flavin containing monooxygenase 1 -1.8 0.021 solute carrier family 40 (ironSlc40a1 -1.5 0.021 regulated transporter) enoyl-Coenzyme A, hydratase/3Ehhadh hydroxyacyl Coenzyme A -1.8 0.023 (32) dehydrogenase Prlr prolactin receptor -1.8 0.031 (14,16,38) Car15 carbonic anhydrase 15 -1.6 0.033 ectonucleotide pyrophosphatase Enpp2 -1.1 0.033 phosphodiesterase 2 insulin-like growth factor binding Igfals -0.9 0.033 protein, acid labile subunit Gcdh glutaryl-Coenzyme A dehydrogenase -0.9 0.041 WD repeat domain, Wipi1 -1.3 0.043 phosphoinositide interacting 1 Aqp4 aquaporin 4 -2.1 0.045 Chgb chromogranin B -1.4 0.046 (32) solute carrier family 6 Slc6a8 -1.0 0.047 (neurotransmitter transporter, creatine) guanine nucleotide binding protein Gnb1l -0.9 0.049 (G protein), beta polypeptide 1-like 191

10

192

Thus, this approach identified 26 genes that are induced by pregnancy in a PRLR-dependent

193

manner. We also mined microarray data of islet gene expression in global PRLR-knockout females

194

(Prlr-/-; created by (39)) examined by Goyvaerts et al. at early gestation (GD9.5) (40). Although

195

the global Prlr-/- and PRLRKO models differ in several key respects (9,14,39,41), this cross-

196

model comparison showed a significant overlap (hypergeometric P = 2.9 x 10-50) in differential

197

gene expression (defined as Q < 0.05) (Fig. 1c). To further validate our microarray findings, we

198

examined gene expression using qPCR on an independent set of wild-type GD16.5 and

199

nonpregnant C57Bl6/J female mice, which revealed significant changes in expression consistent

200

with array data (Fig. 1d). Altogether, these results indicate that the effects of PRLR signaling on

201

β-cell gene activation occur independently of PRLR signaling in other tissues, and are present at

202

both early and late gestation.

203 204

Prolactin and high-fat feeding stimulate divergent transcriptional cascades in the β-cell.

205

Our analysis establishes that PRLR signaling is necessary for the induction of a subset of genes

206

in islets during healthy gestation. However, PRLR signaling may itself interact with the hormonal

207

milieu of pregnancy to regulate transcription. To determine whether PRLR signaling is sufficient

208

to regulate genes in an islet-autonomous fashion, we treated cultured islets for 24 hours with

209

recombinant prolactin. This treatment significantly upregulated many of the PRLR-dependent

210

DEGs (Fig. 2a), including Tph1 and Tph2 as previously reported (29). Our findings therefore

211

demonstrate that PRLR signaling is both sufficient and required to regulate a subset of DEGs

212

normally induced by pregnancy.

213

Metabolic stressors including nutrition and pregnancy have been theorized to converge on the

214

β-cell, leading to its dysfunction and ultimately diabetes mellitus; however, empirical evidence

11

215

supporting this mechanistic convergence remain inconclusive (1,42). Because pregnancy initiates

216

an adaptive metabolic response via profound increases in lactogenic and other hormones, we

217

hypothesized that PRLR-mediated adaptations activate a pregnancy-specific adaptive mechanism

218

in pancreatic islets. To test this, we first compared our βPRLRKO dataset to published RNA-

219

sequencing data of gene expression changes following high-fat diet (HFD), a widely-used

220

metabolic stress model system (43) (Fig. 2b). Surprisingly, the overlap of DEGs was minimal and

221

not significant (Hypergeometric P = 0.4), comprising only 4 genes (Matn2, 2410021H03Rik,

222

Igfbp5, Cntn3) that were differentially co-expressed by both βPRLRKO and 30-day HFD feeding.

223

As Cruciani et al. only examined male mice (43), we performed a 4-week nutritional study using

224

female mice to determine whether high-fat diet (HFD) feeding displayed sexually dimorphic

225

effects on the 70 DEGs reaching Q-value significance in βPRLRKOs. Consistent with findings in

226

male mice, females exhibited no significant changes in these genes following 4 weeks of high-fat

227

diet (Fig. 2c). Thus, for both sexes, HFD feeding was incapable of inducing PRLR-dependent

228

DEGs.

229

We next sought to determine whether the PRLR is required for β-cell physiologic adaptation

230

to HFD feeding. Unlike the requirement for intact PRLR signaling in maintaining gestational

231

glucose homeostasis, both PRLRKO female mice and littermate controls had no baseline

232

difference in GTT (Fig. 2d) and developed equivalent glucose intolerance following four weeks of

233

HFD feeding (Fig. 2e). These results support our hypothesis that both transcriptional regulation

234

and physiologic adaptation of β-cells by PRLR signaling employs pregnancy-specific mechanisms

235

that differ from those of HFD adaptation.

236 237

Pathway analysis of differential gene expression.

12

238

To identify potential mechanisms underlying pregnancy-specific adaptations in islets, we next

239

sought to understand the global transcriptional landscape that is regulated by PRLR signaling in

240

pregnancy. Because the FDR-adjusted threshold yielded only 70 DEGs, we considered whether

241

reducing the statistical stringency of our analysis (P < 0.05) would capture nodal networks and

242

regulatory clusters. Hierarchical clustering and heatmap visualization justified this approach,

243

displaying a distinct expression profile even at this cutoff (Fig. 3a). KEGG Pathway enrichment

244

of the 2,540 genes (P < 0.05) revealed a clear signature of cellular proliferation amongst all of the

245

top-5 most enriched networks: G2/M DNA Damage Checkpoint (P = 1.7 x 10-5), GADD45

246

Signaling (P = 5.6 x 10-5), Regulation of Cellular Mechanics (P = 2.3 x 10-4), Estrogen-mediated

247

S-phase Entry (P = 2.8 x 10-4), and Mitotic Roles of Polo-Like Kinase (P = 4.9 x 10-4). Volcano

248

plot revealed a negatively skewed signature of transcriptional changes by βPRLRKO relative to

249

littermate controls (Fig. 3b), consistent with known upregulation of β-cell genes during pregnancy

250

(30,31).

251

Our gene set enrichment analysis identified established pathways differentially affected by loss

252

of PRLR signaling. However, to identify novel networks affected in βPRLRKO, we used the

253

STRING database (44) to generate a network of DEGs based on known and predicted interactions

254

among their encoded proteins (Fig. 3c). After ranking nodes by degree of interaction, the largest

255

cluster was identified to functionally enrich pathways associated with cellular proliferation and

256

developmental programs (Fig. 3d): cell cycle (BH-adjusted P = 10-9), progesterone-mediated

257

oocyte maturation (BH-adjusted P = 10-6.2), p53 signaling (BH-adjusted P = 10-5.3), FoxO signaling

258

(10-5.2), and oocyte meiosis (BH-adjusted P = 10-4.3). By contrast, when protein-protein interaction

259

analysis was performed on the dataset from mice fed high-fat diet (43), the network instead

260

enriched pathways mediating fatty acid metabolism and signaling, with peroxisome proliferator-

13

261

activated receptors (PPARs) as the most-enriched transcriptional regulators (Supplemental Figure

262

S2; (26)). These data demonstrate that βPRLRKO impairs a pregnancy-specific program of β-cell

263

proliferation during pregnancy.

264 265

EZH2/PRC2 and FOXM1 are PRLR-dependent candidate transcriptional regulators of β-

266

cell adaptation in pregnancy.

267

To identify upstream regulators capable of driving this proliferative signature, we used the

268

Ingenuity® (IPA) causal network analysis to leverage the directionality of gene changes when

269

performing gene set enrichment (45). IPA upstream functional analysis disproportionately

270

enriched genes known to interact with PRLR itself, both directly and indirectly (Fig. 4a). Other

271

upstream regulators included Growth and DNA Damage Inducible 45 γ (Gadd45g, 2.1-fold

272

suppressed) and DNA methyltransferase 3B (Dnmt3b, 2.2-fold suppressed), both regulators of de

273

novo DNA methylation (46,47). Examination of the genes reported to interact with PRLR revealed

274

increased expression of upstream factors and decreased expression of PRLR target genes (Fig. 4b).

275

Of the PRLR target genes, we noted that the histone methyltransferase Enhancer of Zeste

276

Homologue 2 (Ezh2) was suppressed in βPRLRKO.

277

As a transmembrane receptor resident at the cell surface, PRLR indirectly regulates

278

transcription through signal transduction cascades, including the canonical JAK2-STAT5 pathway

279

(48). However, as mice with a β-cell specific deletion of Stat5 do not exhibit defective glucose

280

homeostasis or GDM during pregnancy (49), we hypothesized that multiple transcription factors

281

coordinately regulate transcription for PRLR signaling-mediated gestational adaptations. To

282

identify candidates, we inspected the proximal promoter region (-1kB to +500kB) of differentially-

283

regulated genes (P < 0.05) for disproportionate enrichment of transcription factor binding sites.

14

284

Using the ENCODE ChIP-sequencing database (50), we identified promoter binding sites for

285

Foxm1 and E2f4 among down-regulated DEGs (Fig. 4c), a finding consistent with prior studies

286

(14,17). Stat5a was also statistically enriched (FDR < 0.05), though not among the 10 most-

287

enriched putative regulators. Conversely, promoter enrichment of genes expressed at higher levels

288

in βPRLRKO identified promoter binding sites for Suz12 (BH-adjusted P = 10-7.8) and Ezh2 (BH-

289

adjusted P = 10-4.4), two components of the polycomb repressor complex (PRC2) (Fig. 4d). Co-

290

localization of Ezh2 and Suz12 promoter binding sites DEGs was also noted, demonstrating a 90%

291

(28/31) overlap. To determine whether PRC2 methyltransferase activity likely controls gene

292

expression in βPRLRKO, we then enriched DEG promoters using ChIP-sequencing analysis of

293

chromatin modifications using the human histone modification database (51). Histone

294

modification enrichment identified H3K27 trimethylation (H3K27me3), the modification

295

enzymatically mediated by EZH2/PRC2, as the only histone modification localized to up-regulated

296

DEG promoters (Fig. 4e). Reciprocally, the promoter regions of down-regulated DEGs contained

297

sites of H3K27 acetylation (H3K27ac), the counter-regulatory modification of H3K27me3.

298

Consistent with other research (52,53), these observations support that the PRC2-mediated gene

299

silencing that occurs during pregnancy is de-repressed in the setting of βPRLRKO.

300

Ezh2 has been classically identified as a transcriptional repressor via histone-methyltransferase

301

activity in the PRC2 (54). However, we and others have identified a paradoxical role for Ezh2 as

302

a positive regulator of gene expression upon heterodimerization with Foxm1 and other

303

transcriptional regulators (55-57). Therefore, to understand whether this dual role of Ezh2 might

304

be simultaneously present in pancreatic islets, we plotted the 75 DEGs (P < 0.05) that are targets

305

of Ezh2 according to the Ingenuity® database (Fig. 4f). The resulting distribution depicted a broad

306

array of target genes both increased and decreased by βPRLRKO. Additionally, genes co-targeted

15

307

by Ezh2 and Foxm1 feature key regulators of cellular proliferation (Fig. 4g): Top2a (log2fold = -

308

1.4, P = 0.001 ), Mki67 (log2fold = -1.4, P = 0.001), Ccne2 (log2fold = -0.30, P = 0.04), Myc

309

(log2fold = -0.81, P = 0.0007), Esr1 (log2fold = -0.60, P = 0.005), Cdk1 (log2fold = -0.6, P =

310

0.001), and Cdkn1a (log2fold = 0.87, P = 0.0096). In sum, our analyses support a role of Foxm1

311

and Ezh2 as co-regulators of the proliferative signature within β-cells during pregnancy.

312 313

Discussion

314

Prior studies have identified transcriptional changes within pancreatic islets during pregnancy,

315

a period of rapid β-cell proliferation and mass expansion (29-31). Many hormonal and metabolic

316

changes accompany gestation, with lactogens serving a critical role in physiologic adaptation of

317

β-cells (14,15). While the mitogenic effects of lactogen signaling have been widely studied, the

318

specific gene targets and transcriptional networks under lactogen control remain poorly

319

understood. Using a β-cell specific prolactin receptor knockout (βPRLRKO) mouse model, we

320

confirm the requirement of intact β-cell PRLR signaling for induction of both well-established and

321

novel genes and gene networks during pregnancy.

322

We identified and cross-validated a number of known and novel pro-proliferative genes as

323

induced by pregnancy in a PRLR-dependent manner (Table 1). Our findings are consistent with a

324

recent proteomic analysis of islets during pregnancy, which independently found proteins encoded

325

by our DEGs to be differentially regulated in late gestation: Ivd, Matn2, Ehhadh, Myc, and Chgb

326

(32). Amongst these, we confirmed the requirement of intact β-cell PRLR signaling for induction

327

of well-established gestational pro-proliferative genes (e.g. Tph1, Tph2, Prlr, Opg). In addition to

328

previously identified genes of pregnancy, we identified many novel gene targets of PRLR

329

signaling that provide exciting avenues for future study into the mechanisms of islet gestational 16

330

adaptation. For instance, pregnancy increases gap junction coupling between β-cells (11), which

331

has been theorized to regulate insulin secretion and the alterations in insulin secretion during

332

pregnancy (58). Claudin-8 (Cldn8), here identified as induced by PRLR in pregnancy, is an integral

333

membrane protein and tight junction component that has not been studied in β-cells in any context.

334

In our search for regulatory networks impacted by PRLR disruption in pregnancy, we

335

identified Foxm1 and Ezh2 as putative nodal upstream co-regulators of gestational gene expression

336

and cellular proliferation (Fig. 5). Within the pancreatic islet, Ezh2 has previously been shown to

337

silence the tumor suppressor Ink4a/Arf (p16) in aging and diabetes to increase β-cell proliferation

338

(59,60). Conversely, selective β-cell Ezh2 knockout in mice has been shown to produce

339

hyperglycemia in mice (59). In the current study, we found that pregnancy induces Ezh2 in a

340

PRLR-dependent manner. Therefore, gestational induction of Ezh2 likely creates a pro-

341

proliferative state required for metabolic adaptation. Our results thus broaden our understanding

342

of Ezh2 as a regulator of β-cell function.

343

Though canonically associated with gene silencing via histone 3 trimethylation (H3K27me3),

344

Ezh2 has also been shown to heterodimerize with transcription factors, such as Foxm1, to activate

345

gene expression in a methylation-independent mechanism (57,61). In breast and prostate tissue,

346

Ezh2 is induced by PRLR activation via STAT5, and mediates the mitogenic effects of prolactin

347

in these tissues (62,63). Our results therefore support a broader role for Ezh2 in the regulation of

348

β-cell proliferation during pregnancy, analogous to its role in other tissues (57).

349

As GDM and type 2 diabetes share many similarities and risk factors, it has been widely

350

assumed that they share underlying pathophysiologic mechanisms. However, our data demonstrate

351

that the reprogramming of pancreatic islets during pregnancy is distinct from that of nutritional

352

stressors. We found that the robust PRLR-dependent transcriptional changes in gestation were not

17

353

regulated by HFD feeding. Co-expression network analysis further identified distinct functional

354

enrichment in pregnancy associated with cellular proliferation, whereas HFD feeding enriched

355

fatty acid signaling via PPARs. Our analysis therefore provides several avenues for investigation

356

whereby disease-specificity may be realized. Stable modifications to gene accessibility and

357

expression, termed epigenetic changes, that we have identified as etiology-specific drivers of other

358

diseases (57). Here, we identify many PRLR-dependent epigenetic modifiers that regulate

359

chromatin remodeling (via Ezh2 and Suz12) and/or differential DNA methylation (via Gadd45g

360

and Dnmt3b). Therefore, the pregnancy-specific response to PRLR signaling likely requires many

361

genetic and epigenetic regulators to coordinate β-cell adaptation. Nevertheless, future studies are

362

needed to mechanistically define these programs.

363

While our findings underscore the importance of PRLR signaling as a mediator of β-cell

364

function in pregnancy, we recognize several limitations to our studies. We and others have

365

established that PRLR is expressed specifically in β-cells of rodent islets (14,64). However, we

366

cannot exclude the possibility of non- β-cell effects of βPRLRKO. Therefore, future studies should

367

employ single-cell or cell-sorted approaches to analyze gestational islet transcription. The

368

computational approach we used to define transcriptional regulators of PRLR signaling provide

369

preliminary support for their role in gestational adaptation; however, future mechanistic studies

370

are needed to determine the nature of their influence. Additionally, although gestational induction

371

of DEGs in our data is likely to be directly downstream of PRLR signaling, we recognize that

372

parallel compensatory signaling mechanisms exist which may contribute to the differential

373

regulation of gene expression. Consistent with this possibility, we observed a subtle increase in

374

GHR expression on microarray analysis (1.3-fold, P = 0.03), a known inducer of the prolactin

375

receptor (65).

18

376

In conclusion, the current study examines novel pregnancy-specific gene networks and PRLR-

377

dependent transcriptional programs that describe a state of augmented cellular proliferation.

378

Despite the stated limitations, we believe that the computational approach, combined with both in

379

vivo and in vitro experimentation, serves to significantly advance our understanding of how β-cells

380

respond to the metabolic demands of pregnancy. These lactogen-mediated regulatory networks

381

likely reflect broader adaptive mechanisms that apply to other contexts. We propose PRLR-

382

dependent transcriptional targets and upstream regulators as exciting new candidates to examine

383

in gestational diabetes pathogenesis.

384 385

Acknowledgements: Financial support for this work provided to R.R.B. by University of Alabama

386

School of Medicine Start-up funds and the UAB Diabetes Research Center Pilot and Feasibility

387

Grant (P30 DK079626) and NIH R01 HL133011 to A.R.W. Training support was provided to

388

M.E.P. by the NIH F30 HL137240. R.R.B. designed the study. M.E.P and R.R.B performed

389

experiments, collected data, and analyzed the results. All authors contributed to the writing and

390

editing of the manuscript. The authors would like to thank Drs. Kirk Habegger, Stuart Frank, Chad

391

Hunter, and Sushant Bhatnagar for helpful discussions and critical reading of the manuscript.

392

19

393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437

References 1. Buchanan TA, Xiang AH, Page KA. Gestational diabetes mellitus: risks and management during and after pregnancy. Nat Rev Endocrinol. 2012;8(11):639-649. 2. Rieck S, Kaestner KH. Expansion of beta-cell mass in response to pregnancy. Trends Endocrinol Metab. 2010;21(3):151-158. 3. American Diabetes A. 13. Management of Diabetes in Pregnancy: Standards of Medical Care in Diabetes-2018. Diabetes Care. 2018;41(Suppl 1):S137-S143. 4. Kim SY, England L, Wilson HG, Bish C, Satten GA, Dietz P. Percentage of gestational diabetes mellitus attributable to overweight and obesity. Am J Public Health. 2010;100(6):1047-1052. 5. Wahabi HA, Fayed AA, Alzeidan RA, Mandil AA. The independent effects of maternal obesity and gestational diabetes on the pregnancy outcomes. BMC Endocr Disord. 2014;14:47. 6. Getahun D, Fassett MJ, Jacobsen SJ. Gestational diabetes: risk of recurrence in subsequent pregnancies. Am J Obstet Gynecol. 2010;203(5):467 e461-466. 7. Ruchat SM, Hivert MF, Bouchard L. Epigenetic programming of obesity and diabetes by in utero exposure to gestational diabetes mellitus. Nutr Rev. 2013;71 Suppl 1:S88-94. 8. Lowe WL, Jr., Scholtens DM, Sandler V, Hayes MG. Genetics of Gestational Diabetes Mellitus and Maternal Metabolism. Curr Diab Rep. 2016;16(2):15. 9. Banerjee RR. Piecing together the puzzle of pancreatic islet adaptation in pregnancy. Ann N Y Acad Sci. 2018;1411(1):120-139. 10. Parsons JA, Brelje TC, Sorenson RL. Adaptation of islets of Langerhans to pregnancy: increased islet cell proliferation and insulin secretion correlates with the onset of placental lactogen secretion. Endocrinology. 1992;130(3):1459-1466. 11. Sorenson RL, Brelje TC. Adaptation of islets of Langerhans to pregnancy: beta-cell growth, enhanced insulin secretion and the role of lactogenic hormones. Horm Metab Res. 1997;29(6):301-307. 12. Soares MJ. The prolactin and growth hormone families: pregnancy-specific hormones/cytokines at the maternal-fetal interface. Reprod Biol Endocrinol. 2004;2:51. 13. Huang C, Snider F, Cross JC. Prolactin receptor is required for normal glucose homeostasis and modulation of beta-cell mass during pregnancy. Endocrinology. 2009;150(4):16181626. 14. Banerjee RR, Cyphert HA, Walker EM, Chakravarthy H, Peiris H, Gu X, Liu Y, Conrad E, Goodrich L, Stein RW, Kim SK. Gestational Diabetes Mellitus From Inactivation of Prolactin Receptor and MafB in Islet beta-Cells. Diabetes. 2016;65(8):2331-2341. 15. Vasavada RC, Garcia-Ocana A, Zawalich WS, Sorenson RL, Dann P, Syed M, Ogren L, Talamantes F, Stewart AF. Targeted expression of placental lactogen in the beta cells of transgenic mice results in beta cell proliferation, islet mass augmentation, and hypoglycemia. J Biol Chem. 2000;275(20):15399-15406. 16. Kim H, Toyofuku Y, Lynn FC, Chak E, Uchida T, Mizukami H, Fujitani Y, Kawamori R, Miyatsuka T, Kosaka Y, Yang K, Honig G, van der Hart M, Kishimoto N, Wang J, Yagihashi S, Tecott LH, Watada H, German MS. Serotonin regulates pancreatic beta cell mass during pregnancy. Nat Med. 2010;16(7):804-808. 17. Zhang H, Zhang J, Pope CF, Crawford LA, Vasavada RC, Jagasia SM, Gannon M. Gestational diabetes mellitus resulting from impaired beta-cell compensation in the

20

438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481

18. 19.

20.

21. 22.

23. 24.

25.

26.

27.

28. 29.

30.

absence of FoxM1, a novel downstream effector of placental lactogen. Diabetes. 2010;59(1):143-152. Hughes E, Huang C. Participation of Akt, menin, and p21 in pregnancy-induced beta-cell proliferation. Endocrinology. 2011;152(3):847-855. Karnik SK, Chen H, McLean GW, Heit JJ, Gu X, Zhang AY, Fontaine M, Yen MH, Kim SK. Menin controls growth of pancreatic beta-cells in pregnant mice and promotes gestational diabetes mellitus. Science. 2007;318(5851):806-809. Kondegowda NG, Fenutria R, Pollack IR, Orthofer M, Garcia-Ocana A, Penninger JM, Vasavada RC. Osteoprotegerin and Denosumab Stimulate Human Beta Cell Proliferation through Inhibition of the Receptor Activator of NF-kappaB Ligand Pathway. Cell Metab. 2015;22(1):77-85. Carvalho BS, Irizarry RA. A framework for oligonucleotide microarray preprocessing. Bioinformatics. 2010;26(19):2363-2367. Diboun I, Wernisch L, Orengo CA, Koltzenburg M. Microarray analysis after RNA amplification can detect pronounced differences in gene expression using limma. BMC Genomics. 2006;7:252. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15(12):550. Cai H, Chen H, Yi T, Daimon CM, Boyle JP, Peers C, Maudsley S, Martin B. VennPlex-a novel Venn diagram program for comparing and visualizing datasets with differentially regulated datapoints. PLoS One. 2013;8(1):e53388. Pepin ME, Wende A. R., Banerjee, R.R. . Data from: Supplemental Tables: Impact of pregnancy-specific Prolactin receptor signaling on pancreatic islet gene expression in mice. 2018. https://github.com/mepepin/bPRLRKO_Pepin.2018/raw/master/Supplemental.Tables.xls x. Pepin ME, Wende A. R., Banerjee, R.R. . Data from: Supplemental Figures: Impact of pregnancy-specific Prolactin receptor signaling on pancreatic islet gene expression in mice. 2018. https://github.com/mepepin/bPRLRKO_Pepin.2018/raw/master/Supplemental%20Figure s.pptx. Arumugam R, Horowitz E, Lu D, Collier JJ, Ronnebaum S, Fleenor D, Freemark M. The interplay of prolactin and the glucocorticoids in the regulation of beta-cell gene expression, fatty acid oxidation, and glucose-stimulated insulin secretion: implications for carbohydrate metabolism in pregnancy. Endocrinology. 2008;149(11):5401-5414. Dalgaard LT, Billestrup N, Nielsen JH. STAT5 activity in pancreatic β-cells. Expert Review of Endocrinology and Metabolism. 2008;3(4):423-439. Schraenen A, Lemaire K, de Faudeur G, Hendrickx N, Granvik M, Van Lommel L, Mallet J, Vodjdani G, Gilon P, Binart N, in't Veld P, Schuit F. Placental lactogens induce serotonin biosynthesis in a subset of mouse beta cells during pregnancy. Diabetologia. 2010;53(12):2589-2599. Layden BT, Durai V, Newman MV, Marinelarena AM, Ahn CW, Feng G, Lin S, Zhang X, Kaufman DB, Jafari N, Sorensen GL, Lowe WL, Jr. Regulation of pancreatic islet gene expression in mouse islets by pregnancy. J Endocrinol. 2010;207(3):265-279.

21

482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526

31.

32.

33. 34.

35.

36.

37.

38. 39.

40.

41.

42.

43.

44.

Rieck S, White P, Schug J, Fox AJ, Smirnova O, Gao N, Gupta RK, Wang ZV, Scherer PE, Keller MP, Attie AD, Kaestner KH. The transcriptional response of the islet to pregnancy in mice. Mol Endocrinol. 2009;23(10):1702-1712. Horn S, Kirkegaard JS, Hoelper S, Seymour PA, Rescan C, Nielsen JH, Madsen OD, Jensen JN, Kruger M, Gronborg M, Ahnfelt-Ronne J. Research Resource: A Dual Proteomic Approach Identifies Regulated Islet Proteins During beta-Cell Mass Expansion In Vivo. Mol Endocrinol. 2016;30(1):133-143. Huang C. Wild-type offspring of heterozygous prolactin receptor-null female mice have maladaptive beta-cell responses during pregnancy. J Physiol. 2013;591(5):1325-1338. Anderson ST, Isa NN, Barclay JL, Waters MJ, Curlewis JD. Maximal expression of suppressors of cytokine signaling in the rat ovary occurs in late pregnancy. Reproduction. 2009;138(3):537-544. Street ME, Viani I, Ziveri MA, Volta C, Smerieri A, Bernasconi S. Impairment of insulin receptor signal transduction in placentas of intra-uterine growth-restricted newborns and its relationship with fetal growth. Eur J Endocrinol. 2011;164(1):45-52. Liao S, Vickers MH, Stanley JL, Ponnampalam AP, Baker PN, Perry JK. The Placental Variant of Human Growth Hormone Reduces Maternal Insulin Sensitivity in a DoseDependent Manner in C57BL/6J Mice. Endocrinology. 2016;157(3):1175-1186. Akinci B, Celtik A, Yuksel F, Genc S, Yener S, Secil M, Ozcan MA, Yesil S. Increased osteoprotegerin levels in women with previous gestational diabetes developing metabolic syndrome. Diabetes Res Clin Pract. 2011;91(1):26-31. Baeyens L, Hindi S, Sorenson RL, German MS. beta-Cell adaptation in pregnancy. Diabetes Obes Metab. 2016;18 Suppl 1:63-70. Ormandy CJ, Camus A, Barra J, Damotte D, Lucas B, Buteau H, Edery M, Brousse N, Babinet C, Binart N, Kelly PA. Null mutation of the prolactin receptor gene produces multiple reproductive defects in the mouse. Genes Dev. 1997;11(2):167-178. Goyvaerts L, Lemaire K, Arijs I, Auffret J, Granvik M, Van Lommel L, Binart N, in't Veld P, Schuit F, Schraenen A. Prolactin receptors and placental lactogen drive male mouse pancreatic islets to pregnancy-related mRNA changes. PLoS One. 2015;10(3):e0121868. Freemark M, Avril I, Fleenor D, Driscoll P, Petro A, Opara E, Kendall W, Oden J, Bridges S, Binart N, Breant B, Kelly PA. Targeted deletion of the PRL receptor: effects on islet development, insulin production, and glucose tolerance. Endocrinology. 2002;143(4):1378-1385. Halban PA, Polonsky KS, Bowden DW, Hawkins MA, Ling C, Mather KJ, Powers AC, Rhodes CJ, Sussel L, Weir GC. beta-cell failure in type 2 diabetes: postulated mechanisms and prospects for prevention and treatment. Diabetes Care. 2014;37(6):1751-1758. Cruciani-Guglielmacci C, Bellini L, Denom J, Oshima M, Fernandez N, Normandie-Levi P, Berney XP, Kassis N, Rouch C, Dairou J, Gorman T, Smith DM, Marley A, Liechti R, Kuznetsov D, Wigger L, Burdet F, Lefevre AL, Wehrle I, Uphues I, Hildebrandt T, Rust W, Bernard C, Ktorza A, Rutter GA, Scharfmann R, Xenarios I, Le Stunff H, Thorens B, Magnan C, Ibberson M. Molecular phenotyping of multiple mouse strains under metabolic challenge uncovers a role for Elovl2 in glucose-induced insulin secretion. Mol Metab. 2017;6(4):340-351. Szklarczyk D, Morris JH, Cook H, Kuhn M, Wyder S, Simonovic M, Santos A, Doncheva NT, Roth A, Bork P, Jensen LJ, von Mering C. The STRING database in 2017: quality-

22

527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571

45. 46. 47. 48. 49.

50. 51. 52.

53.

54.

55.

56. 57.

58. 59.

60.

controlled protein-protein association networks, made broadly accessible. Nucleic Acids Res. 2017;45(D1):D362-D368. Kramer A, Green J, Pollard J, Jr., Tugendreich S. Causal analysis approaches in Ingenuity Pathway Analysis. Bioinformatics. 2014;30(4):523-530. Niehrs C, Schafer A. Active DNA demethylation by Gadd45 and DNA repair. Trends Cell Biol. 2012;22(4):220-227. Okano M, Bell DW, Haber DA, Li E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell. 1999;99(3):247-257. Brooks CL. Molecular mechanisms of prolactin and its receptor. Endocr Rev. 2012;33(4):504-525. Lee JY, Gavrilova O, Davani B, Na R, Robinson GW, Hennighausen L. The transcription factors Stat5a/b are not required for islet development but modulate pancreatic beta-cell physiology upon aging. Biochim Biophys Acta. 2007;1773(9):1455-1461. Consortium EP. The ENCODE (ENCyclopedia Of DNA Elements) Project. Science. 2004;306(5696):636-640. Zhang Y, Lv J, Liu H, Zhu J, Su J, Wu Q, Qi Y, Wang F, Li X. HHMD: the human histone modification database. Nucleic Acids Res. 2010;38(Database issue):D149-154. De Gobbi M, Garrick D, Lynch M, Vernimmen D, Hughes JR, Goardon N, Luc S, Lower KM, Sloane-Stanley JA, Pina C, Soneji S, Renella R, Enver T, Taylor S, Jacobsen SE, Vyas P, Gibbons RJ, Higgs DR. Generation of bivalent chromatin domains during cell fate decisions. Epigenetics Chromatin. 2011;4(1):9. Leung TW, Lin SS, Tsang AC, Tong CS, Ching JC, Leung WY, Gimlich R, Wong GG, Yao KM. Over-expression of FoxM1 stimulates cyclin B1 expression. FEBS Lett. 2001;507(1):59-66. Chen H, Gu X, Liu Y, Wang J, Wirt SE, Bottino R, Schorle H, Sage J, Kim SK. PDGF signalling controls age-dependent proliferation in pancreatic beta-cells. Nature. 2011;478(7369):349-355. Kim SH, Joshi K, Ezhilarasan R, Myers TR, Siu J, Gu C, Nakano-Okuno M, Taylor D, Minata M, Sulman EP, Lee J, Bhat KP, Salcini AE, Nakano I. EZH2 protects glioma stem cells from radiation-induced cell death in a MELK/FOXM1-dependent manner. Stem Cell Reports. 2015;4(2):226-238. Mahara S, Chng WJ, Yu Q. Molecular switch of EZH2 in hypoxia. Cell Cycle. 2016;15(22):3007-3008. Pepin ME, Ha CM, Crossman DK, Litovsky SH, Varambally S, Barchue JP, Pamboukian SV, Diakos NA, Drakos SG, Pogwizd SM, Wende AR. Genome-wide DNA methylation encodes cardiac transcriptional reprogramming in human ischemic heart failure. Lab Invest. 2018. Lain KY, Catalano PM. Metabolic changes in pregnancy. Clin Obstet Gynecol. 2007;50(4):938-948. Chen H, Gu X, Su IH, Bottino R, Contreras JL, Tarakhovsky A, Kim SK. Polycomb protein Ezh2 regulates pancreatic beta-cell Ink4a/Arf expression and regeneration in diabetes mellitus. Genes Dev. 2009;23(8):975-985. Zhou JX, Dhawan S, Fu H, Snyder E, Bottino R, Kundu S, Kim SK, Bhushan A. Combined modulation of polycomb and trithorax genes rejuvenates beta cell replication. J Clin Invest. 2013;123(11):4849-4858.

23

572 573 574 575 576 577 578 579 580 581 582 583 584 585 586

61. 62.

63.

64.

65.

Yamaguchi H, Hung MC. Regulation and Role of EZH2 in Cancer. Cancer Res Treat. 2014;46(3):209-222. Yoo KH, Oh S, Kang K, Hensel T, Robinson GW, Hennighausen L. Loss of EZH2 results in precocious mammary gland development and activation of STAT5-dependent genes. Nucleic Acids Res. 2015;43(18):8774-8789. Tan D, Tan S, Zhang J, Tang P, Huang J, Zhou W, Wu S. Histone trimethylation of the p53 gene by expression of a constitutively active prolactin receptor in prostate cancer cells. Chin J Physiol. 2013;56(5):282-290. Brelje TC, Svensson AM, Stout LE, Bhagroo NV, Sorenson RL. An immunohistochemical approach to monitor the prolactin-induced activation of the JAK2/STAT5 pathway in pancreatic islets of Langerhans. J Histochem Cytochem. 2002;50(3):365-383. Galsgaard ED, Nielsen JH, Moldrup A. Regulation of prolactin receptor (PRLR) gene expression in insulin-producing cells. Prolactin and growth hormone activate one of the rat prlr gene promoters via STAT5a and STAT5b. J Biol Chem. 1999;274(26):18686-18692.

587

24

588

25

589

26

590

27

591 28

592

29

593

Figure Legends

594

Figure 1. Identifying the prolactin-dependent transcriptional effects of pregnancy. (a) KEGG

595

pathway enrichment using differentially-expressed genes (DEGs)* of isolated pancreatic islets

596

from pregnant β-cell-specific prolactin receptor knockout (βPRLRKO) mice relative to littermate

597

controls (n = 3). (b) Comparison of genes differentially regulated in pregnant βPRLRKO mice to

598

published studies of islets from pregnant females at various stages: Schraenen et al. (PMID:

599

20886204), Layden et al. (PMID: 20847227), Rieck et al. (PMID: 19574445), and the current

600

study (βPRLRKO). (c) Cross-comparison of βPRLR-dependent DEGs* identified in the current

601

study with those identified by Goyvaerts et al. (PMID: 25816302) using a constitutive whole-body

602

PRLR knockout mouse model (Q < 0.05). (d) qPCR validation of genes found to be induced by

603

pregnancy and suppressed in βPRLR-KO, reported as average relative fold-change  SEM.

604

*Significance was assumed using Bonferroni-adjusted P-value < 0.05.

605 606

Figure 2. Pregnancy and high-fat diet feeding activate divergent transcriptional programs.

607

(a) qPCR quantification of PRLR-dependent pregnancy genes in primary isolated pancreatic islets

608

treated with/without 500 ng/ml recombinant mouse prolactin (PRL+/-) for 24 hours (n = 4). (b)

609

Venn diagram illustrating the degree of overlap between βPRLR-dependent DEGs and those

610

induced by high-fat diet (HFD) based on RNA-sequencing dataset generated by Cruciani et al.

611

(PMID: 28377873). (c) qPCR quantification of PRLR-dependent pregnancy genes in primary

612

isolated pancreatic islets from chow- or HFD-fed mice (n = 4). (d) Intraperitoneal glucose

613

tolerance testing of βPRLRKO mice and littermate controls at baseline (e) following four weeks

614

of standard chow (Chow) or 60% HFD feeding (n = 6). *Significance of transcriptomic data was

615

assumed using Bonferroni-adjusted P-value < 0.05.

30

616 617

Figure 3. Gene expression analysis of PRLRKO identifies proliferative impairment.

618

(a) Hierarchical clustering of normalized beta values via Wald.D2 test and dendrogram constructed

619

by Euclidean distance with heatmap visualization of DEGs*. (b) Volcano plot illustrating the

620

distribution of genes based on -Log10(P-value) as a function of Log10(Fold—Change), labelling

621

the most differentially-regulated genes (|Fold-change| > 2, FDR < 0.05) (c) Protein-protein

622

interaction network generated from DEGs* based on the STRING database. (d) Gene set

623

enrichment of the gene module with highest degree of protein interactivity using KEGG pathways

624

of DEGs. *significance threshold of P < 0.05 was used for gene expression, followed by BH-

625

adjusted P (“Q”) < 0.05 for subsequent gene set enrichment analysis.

626 627

Figure 4. EZH2 and Foxm1 as likely upstream regulators of PRLR-dependent gene

628

expression. (a) PRLR identified as top differentially-expressed upstream regulator via Ingenuity

629

database (IPA® Redwood City, CA). (b) Regulatory network of DEGs* associated with PRLR

630

(blue = down-regulated; yellow = up-regulated). *P < 0.05 for pathway and network enrichment.

631

(c) Upstream DNA-binding protein enrichment of up-regulated differentially-expressed genes

632

(DEGs) and (d) down-regulated DEGs in βPRLRKO relative to WT was performed using

633

published ChIP-Sequencing datasets based on the Encyclopedia of DNA Elements (ENCODE).

634

(e) Up-regulated DEGs were enriched for histone modifications using ENCODE database. (f)

635

Circular genome plot of differentially-regulated direct downstream targets of EZH2. (g) Co-

636

regulatory network of downstream DEGs of EZH2-FOXM1 regulation.

637 638 639

31

640

Figure 5. Proposed model of pregnancy-specific transcriptional reprogramming. Placental

641

and pituitary lactogens induce transcriptional activation of serotonin signaling and proliferation

642

within β-cells via Ezh2 and Foxm1. In contrast, high-fat diet feeding induces transcriptional

643

reprogramming via PPARα independent of the prolactin receptor and serotonin signaling.

32