<<

bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made TCF7L2 plays a complex roleavailable in underhuman aCC-BY-NC-ND adipose 4.0progenitor International biologylicense. which may contribute to

genetic susceptibility to .

Running Title: TCF7L2 and human adipose progenitor biology

Authors: Manu Verma1, Nellie Y. Loh1, Senthil K. Vasan1, Andrea D. van Dam1, Marijana

Todorčević1, Matt J. Neville1, Fredrik Karpe1,2 and Constantinos Christodoulides1.

Affiliations: 1Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of

Medicine, University of Oxford, Oxford OX3 7LE, UK.

2NIHR Oxford Biomedical Research Centre, OUH Foundation Trust, Oxford OX3 7LE, UK.

Address for correspondence to:

Dr Constantinos Christodoulides

Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford OX3 7LE,

UK.

Telephone: +44 (0)1865 857111

E-mail: [email protected]

Keywords: Human Adipose Biology, Adipogenesis, Adult Onset Diabetes, WNT Signalling,

Human Genetics.

Word count: 3992 words

Number of figures: 6

Number of tables: 0

bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made ABSTRACT available under aCC-BY-NC-ND 4.0 International license.

Non-coding genetic variation at TCF7L2 is the strongest genetic determinant of type 2 diabetes (T2D)

risk in humans. TCF7L2 encodes a factor mediating the nuclear effects of WNT

signalling in adipose tissue (AT). Here we mapped the expression of TCF7L2 in human AT and

investigated its role in adipose progenitor (AP) biology. APs exhibited the highest TCF7L2 mRNA

abundance compared to mature adipocytes and adipose-derived endothelial cells. Obesity was

associated with reduced TCF7L2 transcript levels in subcutaneous abdominal AT but increased

expression in APs. In functional studies, TCF7L2 knockdown (KD) in APs led to dose-dependent

activation of WNT/β-catenin signaling, impaired proliferation and dose-dependent effects on

adipogenesis. Whilst partial KD enhanced adipocyte differentiation, complete KD impaired lipid

accumulation and adipogenic expression. Overexpression of TCF7L2 accelerated adipogenesis.

Transcriptome-wide profiling revealed that TCF7L2 can modulate multiple aspects of AP biology

including extracellular matrix secretion, immune signalling and apoptosis. The T2D-risk allele at

rs7903146 was associated with reduced AP TCF7L2 expression and enhanced AT sensitivity.

Our study highlights a complex role for TCF7L2 in AP biology and suggests that in addition to

regulating pancreatic insulin secretion, genetic variation at TCF7L2 may also influence T2D risk by

modulating AP function. bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

1 INTRODUCTION

2 Adipose tissue (AT) plays a central role in the regulation of whole-body energy and glucose

3 homoeostasis. Firstly, it provides a safe depot for the storage of excess calories in the form of

4 triglycerides, thereby protecting extra-adipose tissues from lipotoxicity (1). Secondly, it releases

5 energy as free fatty acids during periods of energy demand such as fasting and exercise. Finally,

6 through the secretion of peptide hormones such as leptin and adiponectin it directly regulates systemic

7 energy balance and insulin sensitivity (2). AT expands through an increase in adipocyte number

8 (hyperplasia) and/or size (hypertrophy). Hyperplastic AT growth is mediated via generation of new

9 adipocytes from adipose progenitors (APs) and is associated with enhanced systemic insulin

10 sensitivity and an improved glucose and lipid profile. In contrast, adipocyte hypertrophy results in

11 AT inflammation and fibrosis which lead to the development of insulin resistance (3,4).

12

13 WNTs are a family of secreted growth factors which function in autocrine and paracrine fashions to

14 regulate stem cell and AP biology (5–7). WNTs signal through frizzled (FZD) receptors to activate

15 multiple downstream signalling cascades. In the best characterized, canonical pathway, WNT binding

16 to FZDs leads to nuclear accumulation of the transcriptional co-activator β-catenin which in

17 conjunction with TCF/LEF transcription factors activates WNT target . In the absence

18 of WNTs, TCF/LEF are bound to members of the Groucho/TLE family of transcriptional

19 co-repressors and suppress WNT/β-catenin signalling. There are four TCF/LEF family members in

20 mammals; TCF7, TCF7L1, TCF7L2 and LEF1 (8). Of these, TCF7L2 is the most highly expressed

21 in AT (9).

22

23 Work from the MacDougald laboratory first highlighted a potential role for TCF7L2 in the regulation

24 of AT function (10). A dominant negative TCF7L2 mutant lacking its β-catenin binding domain was

25 shown to cause spontaneous adipogenesis of 3T3-L1 cells and trans-differentiation of C2C12

26 myoblasts into adipocytes. More recently, the in vitro role of TCF7L2 in the regulation of

27 adipogenesis was revisited with conflicting results. Whilst Chen et al showed that TCF7L2

28 knockdown (KD) in 3T3-L1 preadipocytes leads to impaired adipocyte differentiation (11) in another bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

29 study inducible TCF7L2 deletion in immortalised inguinal mouse APs was associated with enhanced

30 adipogenesis (12). Ex vivo adipose expression of Tcf7l2 was suppressed by high-fat diet (HFD)-

31 induced and genetic obesity (12,13). In vivo, homozygous global Tcf7l2 null mice die perinatally

32 whereas heterozygous null animals are lean with enhanced glucose tolerance and insulin sensitivity

33 compared to wild-type controls both on a chow and following a HFD (14). Conversely, transgenic

34 mice over-expressing Tcf7l2 displayed HFD-induced glucose intolerance (14). Contrasting these

35 findings, targeted deletion of Tcf7l2 in mature adipocytes (mADs) resulted in enhanced adiposity,

36 glucose intolerance and hyperinsulinaemia (11,12). In humans, TCF7L2 expression was decreased in

37 the SC abdominal (hereafter referred to as abdominal) AT of subjects with impaired glucose tolerance

38 (11) and reduced systemic insulin sensitivity (15). Notably, non-coding genetic variation at the

39 TCF7L2 locus was demonstrated to be the strongest genetic determinant of type 2 diabetes (T2D) risk

40 in humans (16,17). The same single nucleotide variation (SNV), rs7903146, was also associated with

41 fat distribution (18). Prompted by these findings we investigated the role of TCF7L2 in human AT

42 function.

43

44 RESEARCH DESIGN AND METHODS

45 Study population: Study subjects were recruited from the Oxford Biobank

46 (www.oxfordbiobank.org.uk), a population-based cohort of healthy 30-50-year-old volunteers (19).

47 Paired abdominal and visceral biopsies were collected from patients undergoing elective surgery as

48 a part of the MolSURG study. Plasma biochemistry (19) and adipocyte sizing (20) were undertaken

49 as described. All studies were approved by the Oxfordshire Clinical Research Ethics Committee, and

50 all volunteers gave written, informed consent.

51

52 Cell culture: Primary APs (derived from AT biopsies) or AP lines were cultured and differentiated

53 as described (21,22). Primary endothelial cells were isolated using a CD31 MicroBead Kit (Miltenyi

54 Biotec). Quantification of intracellular lipid was undertaken using AdipoRed lipid stain (Lonza) and

55 multi-well plate reader (PerSeptive Biosystems, Perkin Elmer).

56 bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

57 Generation of de-differentiated fat (DFAT) cells: DFAT cells were generated by selection and de-

58 differentiation of lipid-laden, in vitro differentiated immortalised APs (23) with modifications (See

59 supplemental information).

60

61 Lentiviral constructs and generation of stable AP lines: TCF7L2 (sh843, TRCN0000262843;

62 sh897, TRCN0000061897) and control (scrambled) shRNA plasmid vectors were purchased (Sigma-

63 Aldrich). The TOPflash reporter vector (24) was a gift from Roel Nusse (Addgene #24307). Lentiviral

64 particles were produced in HEK293 cells using MISSION® (Sigma-Aldrich) packaging mix. Stable

65 AP lines were generated by transduction of cells with lentiviral particles and selected using (2µg/ml)

66 puromycin.

67

68 Doxycycline-inducible AP lines: The TCF7L2 sequence (from TCF4E pcDNA3, a kind gift from

69 Frank McCormick, Addgene #32738) (25) was cloned into the tet-pLKO-puro doxycycline-inducible

70 expression lentiviral vector (gift of Dmitri Wiederschain, Addgene #21915) (26). Stable doxycycline-

71 inducible AP lines were generated by transduction of cells with lentiviral particles and selected using

72 (2µg/ml) puromycin.

73

74 Proliferation assays: Equal number of APs were seeded, trypsinised and counted using a Cellometer

75 Auto T4 (Nexcelom Bioscience) every 96 hours or using CyQUANTTM cell proliferation assays.

76 Doubling time was calculated using the formula: Td = (t2 – t1) x [log (2) ÷ log (q2 ÷ q1)], where t =

77 time (days), q = cell number.

78

79 Luciferase assays: To asses cis-regulatory activity, a 150 genomic sequence centred around

80 rs7903146 was cloned into a luciferase reporter vector (pGL4.23[luc2/minP], Promega) and co-

81 transfected with pRL-SV40 (Promega) into HEK293 (Lipofectamine 2000) or DFAT cells (Neon

82 Transfection System). 48h post transfection, luciferase activity was assessed using the Dual-

83 Luciferase® Reporter Assay System (Promega) on Veritas Microplate Luminometer (Turner

84 Biosystems). bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

85

86 TOPflash reporter assays: β-catenin transcriptional activity in TCF7L2 KD or overexpressing

87 DFAT cells was determined as described (21).

88

89 Real time-PCR and western blots: qRT-PCR and western blotting were performed using TaqMan

90 assays and standard protocols (see supplemental information).

91

92 RNA sequencing, pathway enrichment and binding-site motif analysis:

93 RNA sequencing (RNA-Seq) was performed in scrambled control, sh897 and sh843 TCF7L2-KD

94 DFAT abdominal APs (see supplemental information). Differentially regulated with a false

95 discovery rate <0.05 and an absolute fold-change >1.5, were selected for further analysis. Pathway

96 enrichment and transcription factor binding-site motif analysis were undertaken using Metascape (27)

97 and iRegulon (28), respectively.

98

99 Statistical analysis: Statistical analysis was performed using R, SPSS, STATA or GraphPad. For

100 parametric data, Pearson’s correlation, two-tailed student's test with Welch's correction (where

101 appropriate) for 2 groups, or one- or two-way ANOVA followed by appropriate post hoc tests for

102 multiple groups were used. For non-parametric data, Spearman’s correlation, two-tailed Mann-

103 Whitney or Kruskal-Wallis tests were used. Smoothened splines using generalized additive models

104 were used to investigate the relationship between Adipo-IR and BMI in different rs7903146 genotype

105 carriers. p-values were corrected for multiple comparisons or age, BMI and sex (where appropriate)

106 and p <0.05 was considered significant.

107

108 RESULTS

109 TCF7L2 expression is highest in APs. To elucidate the role of TCF7L2 in human AT biology we

110 mapped its expression profile in AT. In biopsy samples from 30 lean and 30 obese individuals,

111 TCF7L2 mRNA abundance tended to be higher in abdominal versus gluteal fat (Fig. 1A). Compared

112 with obese subjects, lean individuals had higher TCF7L2 expression in abdominal AT. TCF7L2 bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

113 transcript levels were similar in the abdominal and visceral AT depots (Fig. 1B). In fractionated AT

114 from over 100 healthy volunteers, TCF7L2 mRNA levels were higher in APs compared with mADs

115 from both the abdominal and gluteal fat depots (Fig. 1C). Furthermore, in a small sample group (n=5-

116 6), TCF7L2 expression was higher in APs versus adipose-derived endothelial cells (Fig. 1D). Finally,

117 TCF7L2 mRNA abundance in abdominal APs correlated positively with body mass index (BMI)

118 whilst an opposite trend was detected in abdominal mADs (Fig. 1E, F). No associations between BMI

119 and TCF7L2 expression in gluteal adipose cell fractions were detected (Fig. S1). Based on these data

120 we focused our subsequent studies on deciphering the function of TCF7L2 in APs.

121

122 TCF7L2 dose-dependently modulates AP differentiation. We investigated the role of TCF7L2 in

123 AP biology using de-differentiated fat (DFAT) cells generated from immortalised abdominal

124 adipocytes (22). These retain their depot-specific gene expression signature (Fig. S2) and have a

125 higher adipogenic capacity than primary and immortalised APs. Stable KD of TCF7L2 in these cells

126 was achieved with two shRNAs targeting the universal 9 of TCF7L2 (Fig. 2A, B). The first

127 shRNA (sh897) led to 30% KD of TCF7L2 at mRNA level and 68% KD at level. The second,

128 more efficient shRNA (sh843), led to 70% reduction in TCF7L2 expression and near complete

129 TCF7L2 protein KD. KD of TCF7L2 led to impaired AP proliferation (Fig. 2C). No differences in

130 the doubling time of moderate- versus high-efficiency TCF7L2-KD cells were detected. Partial

131 TCF7L2-KD was also associated with augmented adipocyte differentiation as ascertained by

132 increased lipid accumulation and enhanced adipogenic gene expression (Fig. 2D - H). In sharp

133 contrast, high-efficiency KD of TCF7L2 resulted in impaired adipogenesis. These findings were

134 confirmed in primary abdominal TCF7L2-KD cells from two subjects. In these latter experiments the

135 anti-adipogenic effect of sh843 was more subtle despite identical TCF7L2-KD efficiency in primary

136 and DFAT cells (Fig. 2I, J). In complimentary experiments we also investigated the effects of

137 inducible TCF7L2 over-expression on AP functional characteristics using a Tet-On system (Fig. 3).

138 The doxycycline (DOX) dose was selected to induce approximately 2- and 5-fold induction in

139 TCF7L2 expression compared to vehicle treated TCF7L2 overexpressing APs which corresponded to

140 an equivalent increase in protein production (and 2.6 and 6.6-fold higher protein production versus bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

141 vehicle treated empty vector controls) (Fig. 3A, B). AP cells were grown in DOX-free media and

142 TCF7L2 expression was induced upon plating for proliferation and differentiation experiments and

143 throughout thereafter. Ectopic expression of TCF7L2 did not influence AP proliferation (Fig. 3C).

144 Low-dose TCF7L2 overexpression similarly did not affect adipocyte differentiation (Fig. S3)

145 although, the DOX-induced increase in TCF7L2 expression was not sustained throughout

146 adipogenesis (Fig. S4). On the other hand, 5-fold increase in TCF7L2 protein production accelerated

147 adipocyte differentiation (Fig. S5). By the end of the differentiation time course however, TCF7L2

148 over-expressing adipocytes displayed only a subtle increase in lipid accumulation concomitant with

149 increased CEBPA expression versus vehicle treated controls (Fig. 3D-H). We conclude that, in vitro

150 TCF7L2 has dose-dependent actions on adipogenesis.

151

152 TCF7L2 dose-dependently modulates WNT/β-catenin signalling. We next determined whether the

153 effects of TCF7L2 on AP function were driven by changes in WNT/β-catenin signalling (Fig. 4).

154 Partial KD of TCF7L2 did not result in altered expression of the universal WNT target gene AXIN2

155 (Fig. 4A). In contrast, (near) complete TCF7L2-KD was associated with increased AXIN2 levels.

156 Using TCF7L2-KD cells stably expressing the TOPflash promoter reporter construct which monitors

157 endogenous β-catenin transcriptional activity we confirmed that complete depletion of TCF7L2 was

158 associated with robust stimulation of canonical WNT signalling whilst weak activation was also seen

159 following partial KD both in the absence and presence of WNT3A (Fig. 4B). Interestingly, despite

160 increased β-catenin transcriptional activity, active β-catenin protein levels were decreased in both

161 sh897 and sh843 cells (Fig. 4C). Given the paradoxical activation of WNT/β-catenin signalling in

162 TCF7L2-KD APs we examined the expression of other TCF/LEF family members in these cells (Fig.

163 4D & Fig. S6A). Only TCF7 and TCF7L1 were expressed in abdominal APs. Notably, TCF7L2-KD

164 was associated with a dose-dependent increase in TCF7 mRNA abundance in both DFAT and primary

165 abdominal APs (Fig. 4D). In gain-of-function experiments, 48-hour induction of 2- and 5-fold

166 TCF7L2 production did not influence AP AXIN2 mRNA levels (Fig. 4E). In TOPflash promoter

167 assays, TCF7L2 over-expression led to inhibition of WNT/β-catenin signalling (Fig. 4F, G).

168 However, DOX treatment also suppressed TOPflash activity in empty vector control cells. Over- bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

169 expression of TCF7L2 did not influence active β-catenin protein levels (Fig. 4I). Collectively these

170 findings suggest that TCF7L2 functions to antagonize the transcriptional activity of β-catenin in

171 abdominal APs.

172

173 Transcriptome-wide profiling reveals that TCF7L2 regulates multiple aspects of AP biology. To

174 identify the genes and biological processes regulated by TCF7L2 in APs we undertook transcriptomic

175 analyses of TCF7L2-KD cells using RNA-Seq (Fig. 5A). High-efficiency TCF7L2-KD altered the

176 expression of 2,806 genes (18% of all expressed genes) whilst partial KD resulted in a more modest

177 change in the transcriptome with 1,024 genes differentially regulated (FDR < 5%, absolute fold-

178 change > 1.5) (Fig. 5B). Gene set enrichment analysis revealed that the cluster of genes suppressed

179 in complete TCF7L2-KD cells was enriched for pathways and processes involved in cell cycle,

180 response to hypoxia, signalling and ribosome biogenesis (Fig. 5E). Interestingly, cell cycle and

181 ribosome biogenesis are two key pathways that are transcriptionally positively regulated by canonical

182 WNT signalling (29). Consistent with these findings, the expression of many well-established

183 WNT/β-catenin target genes was reduced in high-efficiency TCF7L2-KD APs (Fig. S7). The cluster

184 of upregulated genes in high-efficiency KD cells was enriched for genes involved in interferon

185 signalling and extracellular matrix (ECM) organization. In moderate-efficiency TCF7L2-KD cells

186 the downregulated gene set was also enriched for pathways and processes involved in response to

187 hypoxia and p53 signalling (Fig. 5F). Additionally, we detected enrichment for circadian regulation

188 of gene expression. The upregulated gene cluster in sh897 cells was enriched for genes involved in

189 ECM organization and immune and inflammatory processes. We also performed transcription factor-

190 binding site motif analysis on the promoters of genes differentially expressed in TCF7L2-KD APs

191 (Fig. 5E, F). The promoters of genes suppressed following both complete and partial TCF7L2-KD

192 were enriched for binding sites of and FOXM1 which regulate cell proliferation. Genes whose

193 expression was decreased in high-efficiency TCF7L2-KD cells were also enriched for TFDP1,

194 TFDP3 and binding sites in their promoters. TFDP family members co-operatively regulate

195 cell cycle genes with members of the family whilst KLF4 suppresses p53 expression and is a β-

196 catenin target gene. The promoters of genes suppressed in partial TCF7L2-KD cells were also bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

197 enriched for HIF1A binding sites. Genes, whose expression was increased in both sh897 and sh843

198 cells, exhibited enrichment for binding sites of transcription factors involved in cytokine and

199 particularly, interferon signalling in their promoters namely, IRF1, IRF7, STAT1, and STAT2.

200 Finally, real-time PCR revealed good concordance between changes in the transcriptome of DFAT

201 and primary APs following moderate- but not after high-efficiency TCF7L2-KD (Fig. 5G, H).

202

203 The T2D-risk allele at rs7903146 reduces AP TCF7L2 expression. Genome wide association study

204 (GWAS) meta-analyses have revealed that non-coding genetic variation at TCF7L2 is the strongest

205 genetic determinant of T2D risk in humans (16,17). Previous efforts to characterize the mechanism

206 of action at TCF7L2 revealed that the fine-mapped T2D-risk allele at rs7903146 overlaps enhancer

207 histone marks in both islets and APs (30). Whilst this variant has been shown to increase TCF7L2

208 expression in islets (31), evidence demonstrating that it is associated with a cis-expression

209 quantitative trait locus (eQTL) in APs has been missing. By examining age-, BMI- and sex-adjusted

210 TCF7L2 mRNA abundance data in isolated mADs and ex vivo cultured abdominal and gluteal APs

211 from up to 21 homozygous carriers of the T2D-risk variant (T) and 59 subjects homozygous for the

212 wild-type allele (C) at rs7903146 we found that the T allele reduces TCF7L2 expression in abdominal

213 APs (Fig. 6A, B). A similar trend was detected in gluteal APs. We substantiated this finding in a

214 subset of samples by demonstrating lower TCF7L2 protein levels in abdominal APs derived from

215 T2D-risk variant carriers versus ancestral allele carriers (Fig. 6C). In cell-based luciferase assays a

216 150 base pair nucleotide genomic sequence centred around rs7903146 exhibited allele-specific

217 enhancer properties in abdominal APs and HEK293 cells with the T allele abolishing enhancer

218 activity (Fig. 6E, F). Histological assessment of abdominal AT biopsies from 19 age- and BMI-

219 matched pairs of males homozygous either for the T2D-risk allele or the protective allele at rs7903146

220 did not reveal any difference in median adipocyte size between the two genotypes (Fig. 6G).

221 However, interrogation of plasma biochemistry data from 600 age- and BMI-matched pairs of

222 subjects from the Oxford Biobank showed that T2D-risk variant carriers had reduced AT insulin

223 resistance (Adipo-IR) (Fig. 6H) which was more pronounced in obese individuals (Fig. 6I). We bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

224 conclude that in addition to its established role in regulating pancreatic insulin secretion, genetic

225 variation at rs7903146 may also influence T2D risk through effects on AP biology.

226

227 DISCUSSION

228 Our study highlights a complex role for TCF7L2 in AP biology and AT function. We show that

229 TCF7L2 expression is similar across different fat depots and diminishes in abdominal AT in obesity.

230 The lower AT TCF7L2 mRNA abundance in obese subjects could be mediated via increased

231 methylation at TCF7L2 (32) and may contribute to the AT dysfunction that is associated with greater

232 adiposity. Our results also revealed that TCF7L2 was highly expressed in all adipose cell lineages

233 examined, namely APs, mature adipocytes and endothelial cells with the former exhibiting the highest

234 transcript levels. These data suggest that TCF7L2 has pleiotropic roles in AT ranging from the

235 regulation of AP biology and adipogenesis, to the modulation of mature adipocyte function and

236 angiogenesis. Finally, we showed that increased adiposity was paradoxically associated with higher

237 TCF7L2 expression in isolated abdominal APs despite reduced TCF7L2 mRNA abundance in whole

238 AT. It is likely that the reduction in TCF7L2 mRNA levels in the abdominal depot in obesity is driven

239 by lower expression in adipocytes given that they are the principal cellular component of AT and as

240 hinted by the correlation data in Fig. 1F. Thus, obesity seems to lead to directionally opposite changes

241 in TCF7L2 expression in different AT cellular fractions.

242

243 To elucidate the role of TCF7L2 in AP biology, we undertook functional studies in immortalised and

244 primary APs. These revealed that TCF7L2 protein production may be regulated both transcriptionally

245 and at the level of protein translation because TCF7L2-KD was associated with more pronounced

246 changes in protein than mRNA levels. Thus in addition to regulating its own transcription (33)

247 TCF7L2 may regulate its own translation. The loss-of-function studies also showed that TCF7L2-

248 KD impairs AP proliferation. Additionally, they demonstrated that TCF7L2 dose-dependently

249 regulates adipogenesis. We speculate that the primary role of TCF7L2 in APs is to restrain

250 adipogenesis since only complete TCF7L2-KD led to impaired adipocyte differentiation whilst bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

251 downregulation of its expression within a physiological range, in both DFAT and primary abdominal

252 APs, led to enhanced adipogenesis.

253

254 To decipher the signalling pathways mediating the actions of TCF7L2 in AP biology we examined

255 canonical WNT signalling in TCF7L2-KD cells. These experiments revealed that in abdominal APs

256 TCF7L2 may function to inhibit WNT pathway activity. Whilst prima facie paradoxical, this result

257 is in keeping with previous findings demonstrating that TCF7L2-KD in 3T3L1 cells was associated

258 with a robust increase in Axin2 expression (11). Tang et al (34) also showed that TCF7L2 displayed

259 a cell-type specific activity to both enhance and inhibit WNT signalling and localized the

260 transcriptional repressive ability of TCF7L2 to its C-terminal tail, which is present in some, so-called

261 E isoforms, but not all TCF7L2 transcript species. However, expression of E-type TCF7L2 splice

262 variants is low in AT (35,36). Additionally, none of these studies examined the expression of other

263 TCF/LEF family members in TCF7L2-KD cells (see below). WNT/β-catenin signalling is known to

264 inhibit adipogenesis. Accordingly, high-level KD of TCF7L2 was associated with impaired

265 adipogenesis concomitant with canonical WNT signalling activation. However, downregulation of

266 TCF7L2 expression within a more physiological range (using sh897) led to increased adipocyte

267 differentiation despite mild WNT/β-catenin pathway activation. These data suggest that WNT

268 signalling may have dose-dependent effects on adipogenesis and cell fate determination (21,37).

269 Additionally/alternatively TCF7L2 may engage other signalling pathways to influence AP biology.

270

271 To further elucidate the genes, pathways and biological processes regulated by TCF7L2 in APs we

272 undertook genome-wide transcriptional profiling of TCF7L2-KD cells. Consistent with our in vitro

273 studies these experiments showed that KD of TCF7L2 activates transcriptional programmes which

274 inhibit the proliferation and modulate the adipogenic capacity of APs. Furthermore, they revealed

275 that TCF7L2 can regulate many aspects of AP biology including ECM secretion, immune and

276 inflammatory signalling, and apoptosis and/or senescence. TCF7L2-KD in APs also led to

277 suppression of HIF1A signalling which promotes fibrosis and AT dysfunction (3). These findings

278 expand the potential functional repertoire of TCF7L2 in AT although, they require experimental bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

279 confirmation. Contrary to our expectations, the RNA-Seq experiments also revealed that TCF7L2

280 promotes canonical WNT signalling in APs. How can we reconcile this finding with our in vitro

281 studies? In TCF7L2-KD cells expression of TCF7 was upregulated. We speculate that TCF7 is a more

282 potent activator of WNT signalling in APs given that functional redundancy is common amongst

283 TCF/LEF family members (8) and based on the increased TOPflash promoter activity and AXIN2

284 expression in TCF7L2-KD cells. Nonetheless, increased TCF7 levels cannot compensate for the

285 absence of TCF7L2 at many WNT target gene promoters. Consequently, AP TCF7L2-KD leads to

286 downregulation of several classic WNT target genes and pathways. Transcription factor-binding site

287 analysis was in keeping with the results of the gene set enrichment analysis. Interestingly we saw no

288 enrichment for TCF7L2 binding in the promoters of genes suppressed in TCF7L2-KD cells.

289 However, genome-wide chromatin occupancy data have shown that TCF7L2 regulates gene

290 expression primarily by binding to intergenic regions (29,33). Lastly, the RNA-Seq data provided

291 additional evidence that partial TCF7L2-KD in DFAT cells may be more physiologically relevant as

292 we were better able to replicate changes in the transcriptome of partial versus complete TCF7L2-KD

293 DFAT cells in moderate- and high-efficiency TCF7L2-KD primary cells respectively.

294

295 GWAS meta-analyses have identified eight independent signals at TCF7L2 which are associated with

296 T2D susceptibility (17). At least three of these overlap regions of active chromatin in AT (17). Of

297 these signals, only rs7903146 has received attention to date. It has been demonstrated through

298 analysis of GWAS metadata of T2D-related traits (38,39) and human physiological studies (40) that

299 the risk allele at this SNV increases T2D susceptibility via impaired insulin secretion. Additionally,

300 this variant was shown to increase TCF7L2 expression in pancreatic islets (31,41). These data have

301 conclusively established that rs7903146 increases T2D risk primarily through islet dysfunction

302 probably driven by increased TCF7L2 expression in pancreatic beta-cells. Nevertheless, this SNV

303 may also influence T2D predisposition via actions in AT as it was shown to overlap active enhancer

304 histone marks in APs (30) and to be associated with fat distribution (18). Using a “soft clustering”

305 method to group variant-trait associations ascertained from GWAS for 94 independent T2D signals

306 and 47 diabetes-related traits another study also provided some evidence that “lipodystrophy-like” bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

307 insulin resistance may contribute to T2D predisposition at rs7903146 (39). The same SNV was further

308 shown to overlap a DNA-methylated genomic region exhibiting increased methylation in AT from

309 obese versus lean subjects which was reversed post gastric bypass surgery (32). Extending these

310 findings, we now demonstrate that rs7903146 is an eQTL for TCF7L2 in abdominal APs with the T

311 allele reducing TCF7L2 expression. Whilst the eQTL signal at rs7903146 was only nominally

312 significant our gene expression data were derived from ex vivo expanded APs. This would have added

313 noise to the eQTL dataset in addition to the noise introduced by inter-individual variability in TCF7L2

314 AP expression. We also corroborated our data by demonstrating that the T2D-risk variant at this SNV

315 was associated with reduced AP TCF7L2 protein levels and displayed allele specific enhancer activity

316 in luciferase assays which was directional consistent with the eQTL data. Interestingly rs7903146

317 confers higher T2D susceptibility in lean versus obese subjects (17). Our results offer a possible

318 explanation for this paradox. In our study the T2D-risk variant at this SNV was associated with

319 enhanced age- and BMI-adjusted AT insulin sensitivity which was more pronounced in obese

320 subjects. Whilst we detected no difference in adipocyte size based on rs7903146 genotype, our study

321 sample (n=19) comprised mostly of lean (n=7) and overweight subjects (n=11) due to limited

322 numbers of obese homozygous T2D-risk allele carriers in the Oxford Biobank (see Fig. 6I). Based

323 on the dose-dependent effects of TCF7L2 on adipogenesis demonstrated herein, it will be interesting

324 to determine whether the T2D-predisposing allele at rs7903146 is associated with differential effects

325 on adipocyte size in lean versus obese subjects; with smaller fat cells specifically seen in obese

326 individuals which have higher baseline AP TCF7L2 expression.

327

328 In summary, our work highlights that TCF7L2 plays an important but complex role in AP biology.

329 We also demonstrate that rs7903146 has cis regulatory effects outside the pancreas and reduces

330 TCF7L2 expression in abdominal APs. Thus, in addition to islet dysfunction, altered AP function

331 consequent to changes in TCF7L2 expression might also influence the T2D predisposition conferred

332 by this SNV. Future studies should define the role of TCF7L2 in human adipocytes and characterize

333 the impact of T2D-associated regulatory variants at TCF7L2, beyond rs7903146, which overlap open

334 chromatin in adipose cells on adipose TCF7L2 expression and AT function. bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

335

336 ACKNOWLEDGEMENTS

337 We thank the volunteers from the Oxford BioBank (www.oxfordbiobank.org.uk) for their

338 participation in this recall study. We thank the nurses at the Clinical Research Unit, in particular, Mrs.

339 Jane Cheeseman, for their help in recruitment and sample collection from Oxford BioBank

340 volunteers. The OBB and Oxford BioResource are funded by the NIHR Oxford Biomedical Research

341 Centre (BRC). The views expressed are those of the author(s) and not necessarily those of the NIHR

342 or the Department of Health and Social care. This research is supported by the British Heart

343 Foundation through an Intermediate Clinical Research Fellowship to CC (FS/16/45/32359) and a

344 programme grant (RG/17/1/32663) to FK. MV and ADVD are supported by a Novo Nordisk

345 Postdoctoral Fellowship run in partnership with the University of Oxford. Funding support was also

346 received from the National Institute for Health Research, Oxford Biomedical Research Centre (BRC).

347 We thank the Oxford Genomics Centre at the Wellcome Centre for Human Genetics (funded by

348 Wellcome Trust grant reference 203141/Z/16/Z) for the generation of the Sequencing data. The

349 Genotype-Tissue Expression (GTEx) Project was supported by the Common Fund of the Office of

350 the Director of the National Institutes of Health, and by NCI, NHGRI, NHLBI, NIDA, NIMH, and

351 NINDS. The data used for the analyses described in this manuscript were obtained from the GTEx

352 Portal on 10/20/19.

353

354 CONTRIBUTION STATEMENT

355 Conceptualization, C.C.; Methodology, M.V., N.Y.L., S.K.V.; Investigation, M.V., N.Y.L., S.K.V.,

356 A.D.V.D., M.T., M.J.N., C.C., Writing – Original Draft, M.V., C.C.; Writing – Review & Editing,

357 All authors; Funding Acquisition, C.C., F.K., Resources, C.C., F.K.; Supervision, C.C., F.K. C.C is

358 the guarantor of this work and, as such, had full access to all the data in the study and takes

359 responsibility for the integrity of the data and the accuracy of the data analysis.

360

361 DUALITY OF INTEREST bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

362 MV and ADVD are supported by a Novo Nordisk Postdoctoral Fellowship run in partnership with

363 the University of Oxford. The funders had no role in study design, analysis or reporting of the current

364 work. The authors declare that there is no duality of interest associated with this manuscript.

365

366 PRIOR PRESENTATIONS: Parts of the study were presented as an oral presentation at the 54th

367 Annual Meeting of European Association for the Study of Diabetes, Berlin, Germany, 1-5th October

368 2018 and at the 44th Adipose Tissue Discussion Group meeting, Edinburgh, UK, 6-7th December

369 2018.

370

371 REFERENCES

372 1. Mann JP, Savage DB. What lipodystrophies teach us about the metabolic syndrome. J Clin 373 Invest. 2019;129(10):4009–21. 374 2. Kahn CR, Wang G, Lee KY. Altered adipose tissue and adipocyte function in the pathogenesis 375 of metabolic syndrome. J Clin Invest. 2019;129(10):3990–4000. 376 3. Sun K, Tordjman J, Clément K, Scherer PE. Fibrosis and adipose tissue dysfunction. Cell 377 Metab. 2013;18(4):470–7. 378 4. Marcelin G, Silveira ALM, Martins LB, Ferreira AVM, Clément K. Deciphering the cellular 379 interplays underlying obesity-induced adipose tissue fibrosis. J Clin Invest. 380 2019;129(10):4032–40. 381 5. Prestwich TC, MacDougald OA. Wnt/β-catenin signaling in adipogenesis and metabolism. 382 Curr Opin Cell Biol. 2007;19(6):612–7. 383 6. Christodoulides C, Lagathu C, Sethi JK, Vidal-Puig A. Adipogenesis and WNT signalling. 384 Trends Endocrinol Metab. 2009;20(1):16–24. 385 7. Nusse R, Clevers H. Wnt/β-Catenin Signaling, Disease, and Emerging Therapeutic Modalities. 386 Cell [Internet]. 2017;169(6):985–99. Available from: 387 http://dx.doi.org/10.1016/j.cell.2017.05.016 388 8. Cadigan KM, Waterman ML. TCF/LEFs and Wnt signaling in the nucleus. Cold Spring Harb 389 Perspect Biol. 2012;4(11):1–22. 390 9. Aguet F, Brown AA, Castel SE, Davis JR, He Y, Jo B, et al. Genetic effects on gene expression 391 across human tissues. Nature. 2017;550(7675):204–13. 392 10. Ross SE, Hemati N, Longo KA, Bennett CN, Lucas PC, Erickson RL, et al. Inhibition of 393 Adipogenesis by Wnt Signaling. Science (80- ). 2000;289(5481). 394 11. Chen X, Ayala I, Shannon C, Fourcaudot M, Acharya NK, Jenkinson CP, et al. The diabetes 395 gene and wnt pathway effector TCF7L2 regulates adipocyte development and function. 396 Diabetes. 2018;67(4):554–68. 397 12. Geoghegan G, Simcox J, Seldin MM, Parnell TJ, Stubben C, Just S, et al. Targeted deletion of 398 Tcf7l2 in adipocytes promotes adipocyte hypertrophy and impaired glucose metabolism. Mol 399 Metab [Internet]. 2019;24(March):44–63. Available from: 400 https://doi.org/10.1016/j.molmet.2019.03.003 401 13. Chen ZL, Shao WJ, Xu F, Liu L, Lin BS, Wei XH, et al. Acute Wnt pathway activation 402 positively regulates leptin gene expression in mature adipocytes. Cell Signal [Internet]. 403 2015;27(3):587–97. Available from: http://dx.doi.org/10.1016/j.cellsig.2014.12.012 404 14. Yang H, Li Q, Lee JH, Shu Y. Reduction in Tcf7l2 expression decreases diabetic susceptibility 405 in mice. Int J Biol Sci. 2012;8(6):791–801. 406 15. Karczewska-Kupczewska M, Stefanowicz M, Matulewicz N, Nikołajuk A, Straczkowski M. bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

407 Wnt signaling genes in adipose tissue and skeletal muscle of humans with different degrees of 408 insulin sensitivity. J Clin Endocrinol Metab. 2016;101(8):3079–87. 409 16. Cauchi S, El Achhab Y, Choquet H, Dina C, Krempler F, Weitgasser R, et al. TCF7L2 is 410 reproducibly associated with type 2 diabetes in various ethnic groups: a global meta-analysis. 411 J Mol Med [Internet]. 2007 Jul 3 [cited 2016 Nov 17];85(7):777–82. Available from: 412 http://link.springer.com/10.1007/s00109-007-0203-4 413 17. Mahajan A, Taliun D, Thurner M, Robertson NR, Torres JM, Rayner NW, et al. Fine-mapping 414 type 2 diabetes loci to single-variant resolution using high-density imputation and islet-specific 415 epigenome maps. Nat Genet. 2018;50(11):1505–13. 416 18. Pulit SL, Stoneman C, Morris AP, Wood AR, Glastonbury CA, Tyrrell J, et al. Meta-Analysis 417 of genome-wide association studies for body fat distribution in 694 649 individuals of 418 European ancestry. Hum Mol Genet. 2019;28(1):166–74. 419 19. Karpe F, Vasan SK, Humphreys SM, Miller J, Cheeseman J, Dennis AL, et al. Cohort profile: 420 The Oxford Biobank. Int J Epidemiol. 2018;47(1):21-21g. 421 20. Small KS, Todorčević M, Civelek M, El-Sayed Moustafa JS, Wang X, Simon MM, et al. 422 Regulatory variants at KLF14 influence type 2 diabetes risk via a female-specific effect on 423 adipocyte size and body composition. Nat Genet. 2018;50(4):572–80. 424 21. Loh NY, Neville MJ, Marinou K, Hardcastle SA, Fielding BA, Duncan EL, et al. LRP5 425 regulates human body fat distribution by modulating adipose progenitor biology in a dose- and 426 depot-specific fashion. Cell Metab [Internet]. 2015 Feb 3 [cited 2016 Oct 10];21(2):262–72. 427 Available from: http://dx.doi.org/10.1016/j.cmet.2015.01.009 428 22. Todorčević M, Hilton C, McNeil C, Christodoulides C, Hodson L, Karpe F, et al. A cellular 429 model for the investigation of depot specific human adipocyte biology. Adipocyte [Internet]. 430 2017;6(1):40–55. Available from: 431 https://www.tandfonline.com/doi/full/10.1080/21623945.2016.1277052 432 23. Lessard J, Côté JA, Lapointe M, Pelletier M, Nadeau M, Marceau S, et al. Generation of human 433 adipose stem cells through dedifferentiation of mature adipocytes in ceiling cultures. J Vis 434 Exp. 2015;2015(97):1–5. 435 24. Fuerer C, Nusse R. Lentiviral vectors to probe and manipulate the . 436 PLoS One. 2010;5(2). 437 25. Tetsu O, McCormick F. β-catenin regulates expression of cyclin D1 in colon carcinoma cells. 438 Nature. 1999;398(6726):422–6. 439 26. Wiederschain D, Wee S, Chen L, Loo A, Yang G, Huang A, et al. Single-vector inducible 440 lentiviral RNAi system for oncology target validation. Cell Cycle. 2009;8(3):498–504. 441 27. Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, Tanaseichuk O, et al. Metascape 442 provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun 443 [Internet]. 2019;10(1). Available from: http://dx.doi.org/10.1038/s41467-019-09234-6 444 28. Janky R, Verfaillie A, Imrichová H, van de Sande B, Standaert L, Christiaens V, et al. 445 iRegulon: From a Gene List to a Gene Regulatory Network Using Large Motif and Track 446 Collections. PLoS Comput Biol. 2014;10(7). 447 29. Madan B, HARMSTON N, Nallan G, Montoya A, Faull P, Petretto E, et al. Temporal 448 dynamics of Wnt-dependent transcriptome reveals an oncogenic Wnt / / ribosome axis 449 Graphical abstract Find the latest version : J Clin Invest [Internet]. 2018;128(12):5620–33. 450 Available from: http://www.jci.org/articles/view/122383 451 30. Varshney A, Scott LJ, Welch RP, Erdos MR, Chines PS, Narisu N, et al. Genetic regulatory 452 signatures underlying islet gene expression and type 2 diabetes. Proc Natl Acad Sci U S A. 453 2017;114(9):2301–6. 454 31. Lyssenko V, Lupi R, Marchetti P, Del Guerra S, Orho-Melander M, Almgren P, et al. 455 Mechanisms by which common variants in the TCF7L2 gene increase risk of type 2 diabetes. 456 JClinInvest. 2007;117(8):2155–63. 457 32. Multhaup ML, Seldin MM, Jaffe AE, Lei X, Kirchner H, Mondal P, et al. Mouse-human 458 experimental epigenetic analysis unmasks dietary targets and genetic liability for diabetic 459 phenotypes. Cell Metab. 2015;21(1):138–49. 460 33. Hatzis P, van der Flier LG, van Driel MA, Guryev V, Nielsen F, Denissov S, et al. Genome- 461 Wide Pattern of TCF7L2/TCF4 Chromatin Occupancy in Cells. Mol Cell bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

462 Biol. 2008;28(8):2732–44. 463 34. Tang W, Dodge M, Gundapaneni D, Michnoff C, Roth M, Lum L. A genome-wide RNAi 464 screen for Wnt/beta-catenin pathway components identifies unexpected roles for TCF 465 transcription factors in cancer. Proc Natl Acad Sci U S A [Internet]. 2008;105(28):9697–702. 466 Available from: 467 http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2453074&tool=pmcentrez&rend 468 ertype=abstract 469 35. Kaminska D, Kuulasmaa T, Venesmaa S, Käkelä P, Vaittinen M, Pulkkinen L, et al. Adipose 470 tissue TCF7L2 Splicing is regulated by weight loss and associates with glucose and fatty acid 471 metabolism. Diabetes. 2012;61(11):2807–13. 472 36. Mondal AK, Das SK, Baldini G, Chu WS, Sharma NK, Hackney OG, et al. Genotype and 473 tissue-specific effects on of the transcription factor 7-like 2 gene in 474 humans. J Clin Endocrinol Metab. 2010;95(3):1450–7. 475 37. Basham KJ, Rodriguez S, Turcu AF, Lerario AM, Logan CY, Rysztak MR, et al. A ZNRF3- 476 dependent Wnt/β-catenin signaling gradient is required for adrenal homeostasis. Genes Dev. 477 2019;33(3–4):209–20. 478 38. Mahajan A, Wessel J, Willems SM, Zhao W, Robertson NR, Chu AY, et al. Refining the 479 accuracy of validated target identification through coding variant fine-mapping in type 2 480 diabetes article. Nat Genet. 2018;50(4):559–71. 481 39. Udler MS, Kim J, von Grotthuss M, Bonàs-Guarch S, Cole JB, Chiou J, et al. Type 2 diabetes 482 genetic loci informed by multi-trait associations point to disease mechanisms and subtypes: A 483 soft clustering analysis. PLoS Med. 2018;15(9):1–23. 484 40. Ingelsson E, Langenberg C, Hivert MF, Prokopenko I, Lyssenko V, Dupuis J, et al. Detailed 485 physiologic characterization reveals diverse mechanisms for novel genetic loci regulating 486 glucose and insulin metabolism in humans. Diabetes. 2010;59(5):1266–75. 487 41. Viñuela A, Varshney A, Bunt M van de, Prasad RB, Asplund O, Bennett A, et al. Influence of 488 genetic variants on gene expression in human pancreatic islets – implications for type 2 489 diabetes. bioRxiv [Internet]. 2019;(d):655670. Available from: 490 https://www.biorxiv.org/content/10.1101/655670v1 491

492 FIGURE LEGENDS

493 Fig. 1 Ex vivo TCF7L2 expression in human adipose depots and adipose cell fractions. TCF7L2

494 expression in paired samples of (A) subcutaneous abdominal and gluteal adipose tissue (AT) biopsies

495 from lean and obese subjects (n=30 [15F] / group; Lean – Age 43.9±4.7 years, BMI 22.0±1.1 kg/m2;

496 Obese – Age 43.9±3.6 years, BMI 34.5±2.4 kg/m2), (B) abdominal subcutaneous and visceral AT

497 biopsies (n=27 [16F] / group; Age - 59.1±11.8 years, BMI - 28.2±7.2 kg/m2), (C) cultured APs and

498 mature adipocytes (mADs) from subcutaneous abdominal and gluteal AT biopsies (n=108-114 [59F]

499 / group; Age - 45.3±8.7 years, BMI - 27.2±4.5 kg/m2), and (D) cultured APs and adipose-derived

500 endothelial cells from subcutaneous abdominal and gluteal AT biopsies (n=5-6 [5-6F] / group; Age -

501 51.9±7.9 years, BMI - 28.8±4.0 kg/m2). Errror bars are mean ± SD. (E, F) Correlations between

502 TCF7L2 expression in cultured APs and mADs from subcutaneous abdominal AT biopsies and donor

503 BMI (n=110-113 [59F] / group; Age - 45.3±8.7 years, BMI - 27.2±4.5 kg/m2). qRT-PCR data were bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

504 normalized to geometric mean of (A) PPIA, PGK1, PSMB6, and IPO8, (B) PPIA and PGK1, or (C-

505 F) to 18S rRNA levels. ***p<0.001; #p<0.05 (adjusted for multiple comparisons). Histograms are

506 means ± SEM. Age and BMI data are means ± SD.

507

508 Fig. 2 TCF7L2 KD in abdominal APs impairs proliferation and dose-dependently regulates

509 differentiation. (A-H) TCF7L2 KD in DFAT abdominal APs. shCN = scrambled control, sh897 =

510 moderate and sh843 = high TCF7L2 KD DFAT abdominal APs. TCF7L2 KD was confirmed by (A)

511 qRT-PCR and (B) western blot in DFAT abdominal APs. (C) Doubling time of control, sh897 and

512 sh843 TCF7L2 KD APs. (D) Representative micrographs of control, sh897 and sh843 APs at day 14

513 of adipogenic differentiation. The histogram shows the relative lipid accumulation (as a marker for

514 differentiation) assessed by AdipoRed lipid stain (n=24 wells/group), (E-H) Relative mRNA levels

515 of adipogenic genes CEBPA, PPARG2, FABP4 and ADIPOQ at baseline (day 0), day 7 and day 14

516 of adipogenic differentiation. (I-J) TCF7L2 KD in human primary abdominal APs. (I) Confirmation

517 of TCF7L2 KD by qRT-PCR and western blot in human primary abdominal APs (western blots from

518 one experiment). (J) Representative micrographs of control, moderate (sh897) and high (sh843)

519 TCF7L2 KD human primary abdominal APs at day 14 of adipogenic differentiation. The histogram

520 shows the relative lipid accumulation, assessed by AdipoRed (n=16 wells/group). qRT-PCR data

521 were normalized to 18S rRNA levels. Histograms are means ± SEM. Data obtained from 3

522 independent experiments. ***p<0.001, **p<0.01; ###p<0.001, ##p<0.01, #p<0.05 (adjusted for

523 multiple comparisons). α-tubulin was used as a loading control for western blots.

524

525 Fig. 3 Doxycycline-induced TCF7L2 overexpression in DFAT abdominal APs regulates

526 adipogenesis. DFAT abdominal APs, stably transduced with the empty vector (EV) or TCF7L2

527 overexpression vector (TCF7L2), were cultured in the presence of vehicle (Veh) or DOX (final

528 concentration: 0.01 g/ml or 0.05 g/ml) to induce TCF7L2 overexpression. TCF7L2 overexpression

529 was confirmed by (A) qRT-PCR and (B) western blot in DFAT abdominal APs. (C) Doubling time

530 of DFAT abdominal APs. (D) Representative micrographs of DFAT abdominal APs at day 14 of

531 adipogenic differentiation. The histogram shows the relative lipid accumulation, assessed by bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

532 AdipoRed lipid stain (n=24 wells/group). (E-H) Relative mRNA levels of adipogenic genes CEBPA,

533 PPARG2, FABP4 and ADIPOQ at day 14 of adipogenic differentiation. qRT-PCR data were

534 normalized to 18S rRNA levels. Histograms are means ± SEM and expressed relative to vehicle-

535 treated cells (arbitrarily set to 1). Data obtained from 3 independent experiments. ***p<0.001,

536 **p<0.01, *p<0.05; ###p<0.001, #p<0.05 (adjusted for multiple comparisons). Actin was used as

537 western blot loading control.

538

539 Fig. 4 TCF7L2 dose-dependently modulates WNT/β-catenin signalling. (A-D) Effects of

540 TCF7L2 KD on: (A) AXIN2 mRNA levels, (B) TOPFlash promoter activity following 6h treatment

541 with vehicle (Veh) or 50ng/ml WNT3A (n=12 wells/group), (C) active β-catenin protein levels, and

542 (D) TCF7 mRNA levels. shCN = scrambled control, sh897 = moderate and sh843 = high TCF7L2

543 KD DFAT abdominal APs. (E-I) Effects of TCF7L2 overexpression on: (E) AXIN2 mRNA levels,

544 (F and G) TOPFlash promoter activity following 20h treatment with (F) vehicle (Veh) or (G) 50ng/ml

545 WNT3A (n=12 wells/group), (H) TCF7 mRNA levels, and (I) active β-catenin protein levels. DFAT

546 abdominal APs expressing either the empty vector (EV) or TCF7L2 overexpression vector (TCF7L2)

547 were cultured in the presence of vehicle (Veh) or DOX (final concentration of 0.01 µg/ml or 0.05

548 µg/ml). Data expressed relative to vehicle treatment (arbitrarily set to 1) for EV or TCF7L2

549 overexpressing DFAT abdominal AP line, respectively. Actin was used as a loading control for

550 western blots. qRT-PCR data were normalized to 18S rRNA levels. Histograms are means ± SEM.

551 Data obtained from 3 independent experiments. ***p<0.001, **p<0.01, *p<0.05; ###p<0.001,

552 ##p<0.01, #p<0.05 (adjusted for multiple comparisons).

553

554 Fig. 5 Global transcriptional profiling reveals that TCF7L2 regulates multiple aspects of AP

555 biology. (A) Principal component analysis (PCA) of the global transcriptomic profile of control

556 (scrambled), moderate (sh897) and high (sh843) TCF7L2 KD DFAT abdominal APs. (B) Venn

557 diagram showing the overlap between differentially regulated genes from paired comparisons

558 (FDR<0.05; absolute fold change >1.5). (C and D) Volcano plots showing the genes differentially

559 regulated between control and (C) sh897; or (D) sh843, TCF7L2 KD DFAT abdominal APs. bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

560 Highlighted are the top 20 differentially regulated genes. (E and F) Pathway enrichment analyses of

561 genes downregulated (red) and upregulated (green) in (E) sh897 and (F) sh843 DFAT abdominal

562 APs, vs. controls. Shown also are transcription factor binding-site motif analysis (inset) of

563 downregulated (red) and upregulated (green) genes. (G and H) qRT-PCR validation of differentially

564 regulated genes. qRT-PCR data were normalized to 18S rRNA levels and are from 3 independent

565 experiments. Histograms are means ± SEM. FDR is annotated for RNA-Seq fold-change

566 measurements. ***p<0.001, **p<0.01, *p<0.05 for qRT-PCR measurements.

567

568 Fig. 6 The type 2 diabetes risk allele at rs7903146 reduces TCF7L2 expression in human

569 abdominal APs. (A and B) TCF7L2 expression in paired cultured APs and mature adipocytes

570 (mADs) from subcutaneous (A) abdominal and (B) gluteal AT biopsies from homozygous carriers of

571 the T2D allele T (n=19-21 [4F]) vs. carriers of the non-risk allele C (n=54-59 [29F]). TT carriers,

572 Age - 42.8±6.4 years, BMI - 26.2±6.0 kg/m2. CC carriers, Age - 45.8±9.5 years, BMI - 27.3±3.8

573 kg/m2. p-value adjusted for age, BMI and sex. (C) TCF7L2 protein levels in cultured abdominal APs

574 from age-, BMI- and sex-matched homozygous carriers of the T2D risk allele T vs. carriers of the

575 non-risk allele C (n=11/group). Actin was used as western blot loading control. (D) Chromatin state

576 map showing that rs7903146 overlaps a weak enhancer in APs (image reproduced from

577 type2diabetesgenetics.org). Yellow – weak enhancer, orange – active enhancer, green – transcription

578 (E and F) Luciferase reporter assay in (E) DFAT abdominal APs and (F) HEK293 cells transfected

579 with empty vector (EV) or the luciferase reporter vector containing 151 bp genomic sequence with

580 the T2D risk allele T or the non-risk allele C (n=12-16 replicates/group). ***p<0.001; ###p<0.001,

581 #p<0.05 (adjusted for multiple comparisons). (G) Median adipocyte area (µm2) calculated from the

582 histological sections of abdominal AT from 19 pairs of age- and BMI-matched males grouped by

583 rs7903146 genotype. CC carriers, Age - 44.2±6.3 years, BMI - 25.4±2.8 kg/m2. TT carriers, Age -

584 44.1±7.3 years, BMI - 25.1±2.7 kg/m2. Error bars are median with 95% CI. (H) Comparison of

585 adipose tissue insulin resistance (Adipo-IR) between age-, BMI- and sex-matched homozygous

586 carriers of the T2D risk allele (T) and the non-risk allele (C) (n= 600 pairs). **p<0.01. Data are

587 medians with boxplot showing IQR and whiskers are range of values within 1.5*IQR. (I) Smoothened bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

588 splines showing the relationship between adipo-IR and BMI for homozygous carriers of the T2D risk

589 allele (T) and carriers of the non-risk allele (C) at rs7903146. See Table S1 for anthropometric and

590 plasma biochemistry data for (H and I). bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

Fig. 1 A B ns p=0.051 1.5

Lean )

U

A (

Obese )

2.5 # A

U

N

R

A (

1.0

m

A 2.0 ns

2

N

L

R

7

F m

1.5 C

2 T

L 0.5

e

7

v

F

i t

C 1.0

a

T

l

e

e 27 27

R

v i

t 0.5 0.0

a l

e T T 30 30 30 30 A R A t l 0.0 u ra c e Abdominal AT Gluteal AT b c u is C D S V

Abdominal 2.0 1.5 *** *** Abdominal Gluteal ns Gluteal

#

) )

U 1.5

U

A

A

( (

1.0

S

S

8

8

1

1

1.0

p=0.052 /

/

ns

2

2

L

L

F

7 F

0.5 C T

C 0.5 T

110 108 114 113 5 6 5 6 0.0 0.0 APs mADs APs Endothelial Cells

E F

2.6 2.6

s

s D

P 2.4 A

A 2.4

m

n i

n 2.2

i A

2.2 A

N 2.0

N

R

R m

1.8

2.0 m

2 2

L r = 0.264 r = -0.116 L

7 1.6 7

F 1.8 p = 0.006 p = 0.22

F

C C

T 1.4

T

g

1.6 g o

o 1.2

L L 1.4 1.0 20 30 40 50 20 30 40 50 BMI (kg/m2) BMI (kg/m2) bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

A B Fig. 2

1.5 *** 1.5 ***

### Control ### )

# Low-efficiency KD U ###

)

A

( U

High-efficiency KD

A

n i ( ###

1.0 l 1.0

u

b

S

u

8

1

T

/

2

/

L 2

7 0.5 0.5 #

F

L

7

C

T

F

C T KD 30% KD 70% 68% KD 99% KD 0.0 0.0 shCN sh897 sh843 shCN sh897 sh843 C D shCN *** ### *#* )

2.0 U 4 ### ### A

## ns (

)

sh897 n

o

U

i

t

A a

( 1.5 3

l

u

e

m

m

i u

sh843 c T

2

1.0 c

a

g

n

d

i

i

l

p

i

b

l

u 0.5 1

e

o

v

i

D

t

a l e 0 0.0 R shCN sh897 sh843 shCN sh897 sh843 E F 1.5 ### 1.5 ### ###

#

) )

U ### ###

U

A

A (

( 1.0 1.0

S

S

8

8

1

1

/ /

ns

# 2

A ### ### G

P ### ### R

B 0.5 0.5

A

E

P

C P

0.0 0.0 D0 D7 D14 D0 D7 D14

G H 1.5 ## # ## ### 1.5 ## ### ns

# ) ## ### ns U

p=0.06 A

( )

1.0

U

S

A 8

( 1.0

1

S

/

8

1

Q

/

O

4

P P

I 0.5 B

0.5 D

A A

F *

0.0 0.0 D0 D7 D14 D0 D7 D14 I J shCN ***

) ### *** U 1.5 ###

1.5 ### A ### ( Total

ns 1.5 n o

TCF7L2 i

t

)

)

a U

sh897 l U

## u A

A 1.0

(

(

1.0 m

n

S

i

u l

8 1.0

c

u

1

c

b

/

u a

α-Tubulin

T

2

d

i

L /

7 0.5

p

i

F l 0.5 2 sh843

0.5

L

C

e

7

T

v

F

i

t

C

a T 19% KD l 63% KD 24% KD 99% KD e 0.0 0.0 R 0.0 shCN sh897 sh843 shCN sh897 sh843 shCN sh897 sh843 bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

Fig. 3 EV TCF7L2 A B *** ### Total **

6 ### 6 TCF7L2 #

) U

5 A

) (

Actin

U

n

i

A

t (

4 c 4

S

A

8 /

# 1

p=0.07

2 /

3 L

2 ns

7 L

ns F

7 C

F 2 2

T

C

l

T

a t

1 o 223% 500% T 203% 506% 0 0 Veh 0.01 DOX 0.05 DOX Veh 0.01 DOX 0.05 DOX Veh 0.01 DOX 0.05 DOX Veh 0.01 DOX 0.05 DOX

EV TCF7L2 EV TCF7L2 C D

ns )

1.5 U A

( 1.5 ***

EV TCF7L2

n

) o

i *** t

U * *

a

A

l (

Veh u ***

e 1.0 1.0

m

m

u

i

c

T

c

a

g

n

d

i i

l 0.05 DX p

b 0.5 0.5

i

l

u

o

e

v

D

i

t

a l e 0.0 0.0 R Veh 0.05 DOX Veh 0.05 DOX Veh 0.05 DOX Veh 0.05 DOX EV TCF7L2 EV TCF7L2 E F 2.0 * 1.5 ns ns

* )

) U

U 1.5

A * ( A ns

( 1.0

S

S

8

8

1

1

/

/ 1.0

2

A

G

P R

B 0.5

A

E P

C 0.5 P

0.0 0.0 Veh 0.05 DOX Veh 0.05 DOX Veh 0.05 DOX Veh 0.05 DOX EV TCF7L2 EV TCF7L2 G H 1.5 ns 1.5 ns *

ns )

) *

** U

U

A

A (

( 1.0 1.0

S

S

8

8

1

1

/

/

Q

4

P

O B 0.5 P

I 0.5

A

D

F A

0.0 0.0 Veh 0.05 DOX Veh 0.05 DOX Veh 0.05 DOX Veh 0.05 DOX EV TCF7L2 EV TCF7L2 bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

Fig. 4 A B *** *** 20 ### 3 Control )

*** U ###

A (

Low-efficiency KD ###

y )

r 15 r

U High-efficiency KD

e

A h

( 2 ###

C

S

8

m 1

10

ns / /

###

e

2

s

N I

1 a

r

X e

A 5 ##

f

i

c

u L 0 0 shCN sh897 sh843 Vehicle WNT3a C D *** 6 ###

) *# U

1.5 )

A

U (

# Active A

n 4

(

i l

S ** u

β-catenin 8 ##

b

1

u

/

T 1.0 ns 7

 ns

F /

2 C n ns

i α-Tubulin

T

n

e t

a 0.5

C -

 0 e

v 7 3 7 3 i N 9 4 N 9 4 t C 8 8 C 8 8 c 0.0 h h h h h h A s s s s s s shCN sh897 sh843 DFAT Abdo-APs Primary Abdo-APs

E F ns ns 1.5 1.5 *** # ns ns ) ### **

U ns A

) ns

(

U

y

A r

( 1.0

r e

S 1.0

8

h

1

C

/

2

m

N

/ I

0.5

X

e

A s

a 0.5

r

e

f i

0.0 c u

Veh 0.01 DOX 0.05 DOX Veh 0.01 DOX 0.05 DOX L 0.0 EV TCF7L2 Veh 0.01 DOX 0.05 DOX Veh 0.01 DOX 0.05 DOX

EV TCF7L2 G H Veh ns *** *** ns ) 20 ### 1.5 U *##*#*

A ns

( ##

##

y

r )

r 15

U

e A

h 1.0

(

C

S

m

8

/ 10

1

/

e

s

7

a F

r 0.5

C

e f

5 T

i

c

u L 0 0.0 Veh 0.01 DOX 0.05 DOX Veh 0.01 DOX 0.05 DOX Veh 0.01 DOX 0.05 DOX Veh 0.01 DOX 0.05 DOX EV TCF7L2 EV TCF7L2 WNT3A

) 2.0 U

A ns ns

(

n

I i t 1.5

EV TCF7L2 c

A

/

n i

n 1.0

e t

Active a

C -

β-catenin  0.5

e

v

i t

Actin c A 0.0 Veh 0.01 DOX 0.05 DOX Veh 0.01 DOX 0.05 DOX EV TCF7L2 bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

A B Control vs. High-efficiency KD Control vs. Low-efficiency KD Fig. 5

Control High-efficiency KD Low-efficiency KD

Low vs. High-efficiency KD C Control vs. High-efficiency KD D Control vs. Low-efficiency KD

Fold change >1.5; FDR<0.05 Fold change >1.5; FDR<0.05 Fold change <1.5; FDR>0.05 Fold change <1.5; FDR>0.05

E F Interferon-gamma-mediated signaling pathway Extracellular Matrix Organization Response to type I interferon Cytokine-Mediated Signaling Pathway Extracellular matrix organization Regulation Of Cell Adhesion NES NES Cytokine production Blood Vessel Morphogenesis IRF7 6.501 Regulation of cell adhesion IRF7 5.283 Negative Regulation Of Cellular Component Movement FOXO6 5.234 STAT2 5.478 Negative regulation of cell proliferation Myeloid Leukocyte Activation 4.769 Antigen processing and presentation STAT2 5.036 IRF1 Regulation Of Immune Effector Process of endogenous peptide antigen STAT1 4.384 STAT1 4.553 Negative regulation of cellular component movement Positive Regulation Of Cell Migration JUND 3.832 FOXA1 4.233 Notch signaling pathway Cytokine Production FOSL2 3.656 IRF1 3.915 Regulated exocytosis Cell-Substrate Adhesion ZNF503 3.592 JUNB 3.796 Ameboidal-type cell migration Regulation Of Ossification PURA 3.573 EP300 3.372 Cell-substrate adhesion Chemotaxis NFIC 3.246 CEBPB 3.321 Response to wounding Tissue Morphogenesis JAZF1 3.022 RXRA 3.145 Regulation of protein kinase activity Connective Tissue Development Urogenital system development Positive Regulation Of Response To Cytokine Stimulus Mesenchymal cell differentiation Kidney Development Supramolecular fiber organization Regulation Of Transmembrane Protein Serine/Threonine Kinase Signaling Skeletal system development Regulation Of Inflammatory Response Negative regulation of immune system process Negative Regulation Of Response To External Stimulus Negative regulation of cell differentiation Gland Development

Cell division Monosaccharide Metabolic Process Mitotic cell cycle phase transition Head Development Organelle fission Nephron Development NES Response to decreased oxygen levels NES Response To Hypoxia E2F4 5.808 DNA replication FOSL1 4.876 Cholesterol Metabolic Process TFDP1 5.110 Signal transduction by p53 class mediator PAX4 4.490 Synapse Organization SRF 4.910 Ribonucleotide biosynthetic process E2F4 4.202 Blood Vessel Development SIN3A 4.615 FOXM1 4.442 Muscle structure development HIF1A 3.865 Neuron Projection Morphogenesis MXI1 4.339 Cell part morphogenesis HSF4 3.641 Carbohydrate Biosynthetic Process RPP25 4.159 Actin cytoskeleton organization TWIST2 3.623 Striated Muscle Cell Differentiation 3.551 SMAD1 DNA repair GLIS2 3.559 Signal Transduction By P53 Class Mediator FOXO1 3.224 DMRT1 3.376 TFDP3 3.210 Ribosome biogenesis Regulation Of Neurotransmitter Transport WISP2 3.146 Regulation of cellular response to stress FOXM1 3.297 L- Transport KLF4 3.020 Positive regulation of cell cycle CTBP2 3.259 Free Ubiquitin Chain Polymerization Transmembrane receptor protein Positive Regulation Of Cellular Component Movement tyrosine kinase signaling pathway Positive regulation of transferase activity Mesenchyme Development Meiotic cell cycle process Cognition apoptotic signaling pathway Mammary Gland Development positive regulation of cell migration Circadian Regulation Of Gene Expression extracellular structure organization Positive Regulation Of Neurological System Process

30 20 10 0 10 20 30 30 20 10 0 10 20 30

-Log10 (P-value) -Log10 (P-value) G H

DNER PTGIS WISP2 OSR2 IL6 400 NOV TNXB TAGLN COL11A1 PRKG2 40 3.4E-291 15.7E-82 300 1.11E-37 RNAseq *** 200 30 RNAseq

) qRT-PCR (ImAbdo-KD) U *** ) qRT-PCR (ImAbdo-KD)

A 100 U

( qRT-PCR (Pri-KD)

A l ( **

qRT-PCR (Pri-KD)

o

l r

o 20

t

r

n

t n

o 40

c

o

2.2e-184 c

r

r

e *** e

v * v

o 10

o

20 1.9E-5

e

e g

g 7.8E-4

n n

a ** *** *

a ** h

* h c

0 c

0

d

ns d

l ns

p=0.07 ns l o

*** o F F ns -20 * *** *** -10 2.4E-164 5.44E-61 1.41E-31 -40 1.17E-99 *** -20 bioRxiv preprint doi: https://doi.org/10.1101/854661; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

Fig. 6 A Abdominal AT B Gluteal AT

3 p=0.016 p=0.052

p=0.085 CC [n=54-59] 4 )

TT [n=19-21] ) U

U 3

A

A (

2 (

S

S

8

8

1 1

p=0.083

/

/

2

2

2

L

L

7 7

F 1

F C

C 1

T T

0 0 APs mADs APs mADs C D

) 4 U

A rs7903146

( p=0.02

n i

t 3

c

A

/

2 L

7 2

F

C

T

l a

t 1

o

t

g o L 0 CC TT

E F G

*n*s * *** ns

ns )

2 6000

) # ### ### ###

2.5 2.5 M

U

)

(

A

(

U

e

t

A

i

( z

2.0 2.0

i

n

e

s

U

c

4000

e

n

e

t c

1.5 a 1.5

y

n

n

i

a

c

n

m

o

i

u

p

m i L 1.0

1.0

u d

e 2000

L

a

v

i

e

t

n

v

a i

0.5 l 0.5

a

t

e

i

a

l

R

d

e e

R 0.0 0.0 0 M EV C T EV C T CC TT H ** I