bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity.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

2 FOXS1 is a Master Regulator of Pathological Epithelial to Mesenchymal Transition in Human

3 Epithelia

4

5

6 Timothy A Blenkinsop1, Tomasz Swigut 3, Nathan Boles2, Rajini Srinivasan3, Alvaro Rada-

7 Iglesias.5, Qingjie Wang2, Jeffrey H Stern2, Joanna Wysocka3,4, Sally Temple2#

8

9

10 1Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA

11 2Neural Stem Cell Institute, Rensselaer NY, 12144, USA

12 3Stanford University, Stanford, CA, 94305, USA

13 4Howard Hughes Medical Institute, Stanford, CA, 94305, USA

14 5University of Cologne, Cologne, 50931, Germany.

15

16

17 # Corresponding Author:

18 Contact info: [email protected], phone: (518) 694 8188, fax: (518) 694 8187

19

20 Running head: Epigenetics of RPE in EMT

21

22 Word count: main text: 2888 abstract: 148 bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity.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.

23

24 Abstract

25 Epithelial to mesenchymal transition (EMT) is a biological process involved in normal tissue

26 morphogenesis and also in disease pathology. It causes dramatic alterations in cell morphology,

27 migration, proliferation, and phenotype. We captured the global transcriptional and epigenetic

28 programs elicited when polarized, cobblestone human retinal pigment epithelial (RPE) cells

29 were stimulated to undergo EMT, a process associated with several retinal pathologies. The

30 reorganization of chromatin landscapes occurred preferentially at distal enhancers, rather than

31 promoter regions, accompanied by 3136 significantly changing . Of the 95 significantly

32 changing transcription factors, FOXS1 was most upregulated. Loss and gain of function

33 experiments demonstrated that FOXS1 is upstream of canonical EMT regulators, and can

34 stimulate EMT in RPE and other epithelia. Inhibition of p38, stimulated by combined action of

35 the EMT-inducing factors, TGFβ1 and TNFα, dampened FOXS1 expression. An increase of FOXS1

36 in several cancers indicates it has a role in several EMT-involved pathologies.

37

38

39

40

41 Introduction

42 During EMT, epithelial cells change phenotype, lose cell polarity and typically become migratory

43 and proliferative 1. Several pathological conditions involve EMT-related processes resulting in

44 cell invasion and metaplasia. In the retina, the retinal pigment epithelium (RPE) forms a single, bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

45 non-proliferative epithelial cell layer that supports neural retinal functions. RPE cells are

46 polarized and are connected by tight junctions that form the blood-retina barrier. In

47 pathological conditions, the RPE can undergo EMT and delaminate, migrating through the

48 retina into the vitreous. There the cells proliferate and acquire mesenchymal characteristics,

49 leading to epiretinal membrane formation, such as seen in proliferative vitreoretinopathy

50 (PVR), the most damaging source of retinal detachment and vision loss in developed countries

51 2. While many factors have been associated with RPE changes in PVR, the critical factors and the

52 changes they elicit in RPE that lead to its contribution in PVR are unknown.

53

54 We found that TGFβ1 and TNFα pathways synergistically activate an EMT program in adult

55 human RPE, producing changes similar to those observed in PVR. To characterize the molecular

56 mechanism underlying this cellular transition, we mapped epigenomic and transcriptional

57 changes and identified a set of transcription factors that are upregulated upon EMT induction in

58 the adult RPE. Among those, we found that FOXS1, while not expressed in normal RPE, is highly

59 induced in response to TGFβ1 and TNFα and, notably, is present in patient PVR biopsy. We

60 found that FOXS1 is upstream of canonical regulators of the EMT program, SNAIL and SLUG, in

61 RPE cells. Furthermore, consistent with a more general role in promoting EMT in various

62 biological contexts, FOXS1 also induced SNAIL expression in human mammary cells and

63 activated SLUG and TWIST expression in hepatic epithelial cells, driving them towards EMT.

64 Finally, increased copy number of the FOXS1 locus is found in numerous metastatic cancer

65 studies. These observations identify FOXS1 as a master regulator of EMT that could play a

66 driving role in several pathological contexts. bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

67

68 Results

69 TGFβ1 and TNFα have long been known to be present in the vitreous of patients with PVR 3 and

70 both gene loci have SNP risk associations with PVR 4,5. Hence, we hypothesized that when

71 applied to human RPE, their combination may model the PVR disease state.

72

73 RPE cells were isolated and expanded from adult human cadaver globes, then differentiated

74 into a quiescent polarized cobblestone monolayer 6 (Fig. 1A). The resulting cobblestone RPE

75 cells at passage 0 were treated with RPE with TGFβ1 + TNFα (TNT) for 5 days, and gene

76 expression tested by qPCR on RPE lines from three different donors, with measurements

77 reported as mean ± SEM. TNT treatment led to the synergistic upregulation of EMT master

78 transcription factors SNAIL, SLUG and TWIST (Fig. 1B). SNAIL expression increased 112.23 ±

79 27.91 (fold over control, set at one), P < 0.01 (n=3), after TNT treatment, whereas TGFβ1 alone

80 increased by 7.89 ± 2.21, P < 0.05 (n=3), and the TNFα change was not significant. SLUG

81 expression increased 36.08 ± 6.31, P < 0.01 (n=3), in TNT treatment, while TGFβ1 alone and

82 TNFα alone had little effect. TWIST expression increased 11.26 ± 1.05, P < 0.01 (n=3), in TNT

83 compared to control, while TGFβ1 increased TWIST expression by 6.36 ± 1.91, P <0.05 (n=3),

84 and TNFα had little effect.

85

86 In addition to EMT transcription factor expression, TNT treatment induced a profound change

87 in cell morphology, produced three-dimensional (3D) cell masses (Fig. 1C), and activated

88 expression of αSMA and SNAIL (Fig. 1D,E), markers reported in PVR membranes surgically bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

89 dissected from patients 7,8. To corroborate these findings with PVR disease, we next compared

90 5-day TNT treated versus untreated (vehicle control) RPE cells from 4 different donors to PVR

91 membranes removed from four different patients (Fig. 1F). The PVR membranes expressed

92 MITF at significantly higher levels 0.21 ± 0.79, P < 0.01 (mean +/- S.E.M; n=4) compared to

93 control RPE (normalized to one for each sample), and compared to RPE treated with TNT 0.055

94 ± 0.023, P <0.01 (n=4). BEST1 expression in PVR membranes varied widely 19.48 ± 14.47, P >

95 0.05, (n=4), not significantly different from control RPE or TNT treated RPE 2.63 ± 2.63, P > 0.05,

96 (n=4). SNAIL expression was significantly increased in PVR membranes 12.5 ± 2.13, P < 0.01,

97 (n=4), compared to control RPE normalized to one, consistent with SNAIL expression of RPE

98 treated with TNT 22.51 ± 1.46, P < 0.01, (n=4). Taken together, the in vitro TNT model induces

99 changes in RPE similar to those observed in PVR membranes. We therefore used this model to

100 examine the cellular changes TNT evokes in RPE.

101

102 One of the known signaling pathways downstream of both TGFβ and TNFα, and therefore a

103 candidate for their synergistic effect in RPE is the p38 MAPK pathway. To determine whether

104 this was the case for human RPE, we examined whether TNT treatment induces p38 nuclear

105 localization in RPE cells, testing this on five different donor lines. Indeed, p38 was found to be

106 preferentially localized in RPE nuclei in the TNT conditions 81.69 ± 1.17, P <0.01 (n= 167 cells)),

107 compared to control 65.61 ± 1.02 (n= 144)), TGFβ 62.32 ± .99, (n=161)) or TNFα alone 65.4 ±

108 1.03 (n= 125)) (Fig. 1G). Inhibiting p38 with the small molecule SB202190 (10ng/ml) prevented

109 3D mass formation (Fig. 1H) and suppressed SNAIL 0.04 ± 0.0067, P < 0.01 (n=7) and SLUG 0.34

110 ± 0.09, P < 0.01, (n=7), but not TWIST gene expression compared to the TNT condition (Fig. 1I). bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

111

112 In order to evaluate which additional transcription factors (TFs) may be involved in RPE-EMT,

113 we compared cobblestone RPE and RPE with TGFβ1 alone, TNFα alone or TNT for five days via

114 RNA-sequencing using 2 different donor RPE lines. 3136 genes were found to change

115 significantly between the control and TNT conditions (FDR=0.01), and of those, 95 were known

116 transcription factors. FOXS1 was the most changed TF between these conditions (Fig. 1J).

117

118 We then asked whether p38 activation, induced by TNT was important in inducing FOXS1

119 expression in seven experiments using four different donor lines. When SB202190 (10ng/ml)

120 was added with TNT treatment, FOXS1 expression was suppressed from 60.5 ± 17.19 to 2.57 ±

121 1.29, P < 0.01, (mean+/- S.E.M., n=7) (Fig. 1K), implying FOXS1 is downstream of p38. FOXS1 is

122 expressed in peri-endothelial cells of the testis vasculature 9, in the aorta 10 and during the

123 migration of neural crest derived sensory neurons 11, however it has not previously been linked

124 to EMT. Hence, we have identified key steps along a cascade of changes during RPE

125 transformation from an epithelial to mesenchymal phenotype. To further elucidate the events

126 occurring in RPE EMT, we examined accompanying changes in the epigenome.

127

128 Changes in gene expression patterns during cell fate transitions are associated with changes in

129 the epigenomic status at cis-regulatory elements. In particular, many distal enhancers are

130 activated or decommissioned in a highly cell state-specific manner and this is accompanied by,

131 respectively, gain or loss of certain chromatin marks (reviewed in 12). To systematically

132 characterize epigenomic changes in RPE after EMT, we performed chromatin bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

133 immunoprecipitation and sequencing (ChIP-seq) analysis with antibodies recognizing histone

134 modifications marking active regulatory elements (H3K4me1, H3K4me3, and H3K27ac) using

135 either control cobblestone RPE cells or those treated for 5 days with TNT. First, we examined

136 genome-wide enrichments of H3K27ac, a histone modification associated with active enhancer

137 and promoter states 13,14. Comparisons between untreated or TNT treated RPE revealed a major

138 reorganization of H3K27ac patterns upon EMT, with a large number of sites gaining, and a

139 smaller subset of sites losing H3K27ac in TNT conditions (Fig. 2A). Based on the relative

140 enrichments between the two cellular states, we defined top H3K27ac sites preferentially

141 enriched in TNT conditions (“TNT-specific”, highlighted in red) or untreated RPE (“RPE-specific”,

142 highlighted in blue), and a similar number of sites at which H3K27ac enrichment changed least

143 during treatment (“shared”, shown in orange) (Fig. 2A).

144

145 We then analyzed H3K4me1, a histone mark associated with enhancer elements at the three

146 classes of H3K27ac sites (as defined in Fig. 2D) and observed that changes in H3K4me1

147 generally followed those in H3K27ac: TNT-specific H3K27ac sites had high levels of H3K4me1 in

148 TNT, but not in control RPE and vice versa, whereas shared H3K27ac sites were similarly

149 enriched in both states (Fig. 2B). These observations suggested that major reorganization of

150 H3K27ac patterns might occur at distal enhancer regions rather than proximal promoters. To

151 confirm this, we examined changes in H3K27ac in relation to the H3K4me1/H3K4me3 signal

152 ratio, because relative enrichment of H3K4me1 to H3K4me3 can reliably distinguish putative

153 distal enhancers from promoters (with enhancers having high H3K4me1 and promoters

154 H3K4me3 enrichment) 15. We observed that sites that either gained or lost H3K27ac in TNT RPE bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

155 (highlighted in red or blue, as defined in Fig. 2A) were generally characterized by the high

156 H3K4me1/H3K4me3 ratio, consistent with enhancer identity, whereas unchanged sites (orange)

157 often had low H3K4me1/H3K4me3 ratio, consistent with a large subset of the latter sites having

158 promoter identity (Fig. 2C). In further agreement, dynamically changing H3K27ac sites were

159 preferentially located more than 10 kb from the nearest transcriptional start site (TSS), whereas

160 unchanged sites showed no such preference (and in fact were more enriched in TSS proximity)

161 (Fig. 2D). Functional annotation of these dynamically changing putative enhancer regions

162 revealed association of RPE-specific enhancers with retinal epithelium, visual perception and

163 retinal degeneration categories, whereas enhancers active preferentially in TNT treated cells

164 were enriched for annotations linked to vascular abnormalities (which with the EMT results,

165 may suggest a role in vascular cell migration), cell growth and proliferation (Fig. 2. figure

166 supplements 1-3). Taken together, our data show that EMT in RPE cells is associated with a

167 global reorganization of chromatin landscapes that occurs preferentially at distal enhancer

168 elements.

169

170 Next, we associated H3K27ac regions within each class defined in Fig. 2A with nearby genes and

171 investigated the relationship with changes in expression observed during the RPE to TNT

172 transition (Fig. 2E). We observed that genes associated with RPE-specific H3K27ac were

173 commonly downregulated during the transition, whereas genes associated with TNT-specific

174 H3K27ac were commonly upregulated in TNT treated cells as compared to cobblestone RPE

175 (Fig. 2E). Loci encoding TFs that changed expression during the transition (denoted in Fig. 1J),

176 also typically underwent reorganization of enhancer H3K27ac patterns, as exemplified by the bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

177 FOXS1 locus, which gains high levels of H3K27ac and H3K4me1 at nearby putative enhancer

178 regions (Fig. 2F).

179

180 These transcriptomic and epigenomic analyses predict that FOXS1 may be a key TF involved in

181 orchestrating RPE EMT. Consistent with this hypothesis, FOXS1 was expressed much more in

182 the PVR samples, 49.66 ± 20.28 fold, P < 0.01, (mean+/- S.E.M., n=4 donors) compared to

183 cobblestone RPE (Fig. 3A). We then asked whether FOXS1 is upstream or downstream of the

184 canonical master EMT regulators SNAIL, SLUG and TWIST. FOXS1 shRNA inhibited the increase

185 in SNAIL 0.07 ± 0.026, P < 0.01, (n=4) and SLUG 0.24 ± 0.12, P < 0.01 (n=4) during TNT treatment

186 (Fig. 3B) and prevented 3D mass formation (Fig. 3C). Overexpression of FOXS1 for 5 days

187 induced expression of SNAIL 23.88 ± 4.2, P < 0.01 (Mean+/-S.E.M., n=3 donors), SLUG 1.94 ±

188 0.17, P < 0.01 (n=3) and TWIST 2.98 ± 0.96, P < 0.01 (n=3) over RPE control (Fig. 3D), reduced

189 expression of the RPE markers MITF 0.05 ± 0.01, P < 0.01 (n=3) and OTX2 0.26 ± 0.07, P < 0.01

190 (n=3), but increased BEST1 expression to 13.79 ± 4.12, P < 0.01 (n=3) (Fig. 3E), changes

191 consistent with those seen upon TNT treatment and in PVR patient samples. However, FOXS1

192 overexpression alone did not induce 3D mass formation (Fig. 3F). FOXS1 is therefore necessary

193 and sufficient to change SNAIL and SLUG gene expression.

194

195 The observation that FOXS1 regulates canonical EMT transcription factor expression led us to

196 ask whether it plays a role in EMT in other epithelia, including human mammary epithelium

197 (HME) and human hepatic epithelium (HHE). HME cells treated with TNT showed increased

198 expression of both FOXS1 and SNAIL (Fig. 4A), but not SLUG or TWIST (data not shown). p38 bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

199 inhibition blocked the expression of both FOXS1 from 8.44 ± 0.61 to 0.23 ± 0.1, P < 0.01

200 (mean+/-S.E.M., n=3 donors) and SNAIL from 16.23 ± 8.59 to 0.91 ±0.29, P < 0.01 (n=5) in HME

201 under TNT conditions (Fig. 4A). Knocking down FOXS1 during TNT treatment in HME cells

202 decreased SNAIL expression to 0.096 ± 0.16, P < 0.01 (n=3) compared to RPE control (Fig. 4A),

203 while overexpression of FOXS1 induced SNAIL expression by 3.35 ± 0.58 fold, P < 0.01 (n=3),

204 which is also seen at the protein level (Fig. 4B). Therefore, TGFβ1 and TNFα signaling synergizes

205 through p38, leading to an increase in FOXS1, which then induces an increase in SNAIL

206 expression in HME. Similar studies in HHE cells (Fig. 4C) indicate that FOXS1 is sufficient but not

207 necessary to induce expression of SLUG, and necessary and sufficient to change TWIST

208 expression.

209

210 Since EMT is hypothesized to play a role in the metastasis in many cancers, we examined the

211 incidence of FOXS1 changes in several cancer studies 16,17. Out of 91 whole genome sequencing

212 tumor studies analyzed, 47 included at least one case of multiple copies of the FOXS1 gene (Fig.

213 4D), 14 had greater than 5% of cases and in one uterine cancer study, 23% of cases had multiple

214 copies of FOXS1. Based on our observations of a central role in EMT, we hypothesize that

215 FOXS1 may be involved in the metastatic invasion of cancer cells.

216

217

218 Discussion

219 EMT is a complex, multifactorial process resulting in profound changes in cell phenotype and

220 behavior. The molecular mechanism underlying EMT is still not completely resolved. In this bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

221 study we defined the global transcriptomic and epigenomic changes associated with EMT in

222 human RPE cells. Subsequent analysis elucidated factors associated with EMT-linked pathology

223 in several cell types, and identified FOXS1 as a key driver of the process, and a novel upstream

224 factor which regulates known master EMT transcription factors.

225

226 Here we show that TGFβ1 or TNFα treatment alone cause slight alterations in RPE morphology,

227 but together they cause a dramatic induction of EMT, during which the RPE cells lose their

228 normal cobblestone configuration and grow into 3D masses that lose key RPE features and gain

229 mesenchymal characteristics. The synergistic action of these factors in stimulating EMT has

230 been reported for non-neural epithelia, including from the lung 18 and colon 19, and in the latter

231 case, they do so via p38-MAPK signaling, implying a common mechanism is shared by neural

232 epithelia. TNT treatment resulted in numerous changes in the RPE epigenome, with a bias

233 towards gene activation. The changes in active chromatin signatures occurred largely at distal

234 enhancers, while promoters were much less affected, consistent with enhancers serving as key

235 mediators of changes in cell states and being utilized in a highly dynamic manner (reviewed in

236 12). GO analysis of the significantly changing enhancers highlighted those associated with

237 extracellular remodeling, vasculature, wound healing, and cell proliferation, which align with

238 the stimulation of cell proliferation and migration apparent in the cultures. The changes

239 occurred within 5 days of treatment, demonstrating that stable cobblestone RPE can rapidly

240 disassemble and form 3D masses in the presence of stimulating environmental factors.

241 bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

242 Single nucleotide polymorphisms which confer a significantly increased risk of PVR have been

243 located at both TGFβ15 and TNFα4 genetic loci. Moreover, TGFβ1 and TNFα have both been

244 reported at increased levels in the vitreous of patients with epiretinal membranes 3,20 and

245 TGFβ1 in particular is associated with their maturation into contractile masses that lead to

246 retinal detachment and severe vision loss 21, providing the impetus for use of Rho-kinase

247 inhibitors to prevent the myosin-based contractions. While inhibition of contraction of already

248 formed epiretinal membranes is a valuable therapeutic approach, by determining the causes of

249 RPE EMT, our goal is to identify targets that inhibit the production of these membranes at an

250 earlier stage of the disease.

251

252 Within 5 days of EMT induction, RPE cells increase expression of 95 transcription factors that

253 may play roles in activating the pleiotropic phenotypic changes. Of these, the most dramatic

254 increase occurs in FOXS1, which is not detectable in the control cobblestone RPE. Changes in

255 histone marks surrounding the FOXS1 gene locus were consistent with its activation. We have

256 corroborated expression of FOXS1 in patient PVR samples. Most importantly, knockdown of

257 FOXS1 inhibited production of 3D masses, and was found to be both necessary and sufficient to

258 drive the canonical EMT master regulators SNAIL and SLUG in human RPE cells. These

259 observations implicate FOXS1 induction as an early, causal factor in pathological RPE EMT

260 (modeled in Figure 5), and raise questions about the specific targets of FOXS1, which will be the

261 focus of future studies. Our finding that p38 inhibition can prevent the increase in FOXS1 raises

262 the possibility of an early intervention in this pathological process.

263 bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

264 In addition to its action in RPE cells, we demonstrated that FOXS1 can act as a master

265 transcription factor in EMT in other cells, as it is upstream of the canonical EMT transcription

266 factors SNAIL, SLUG and TWIST in two non-neural epithelial cell types tested. The FOXS1 gene

267 can be hypomethylated in adrenocortical neoplasms 22 and demonstrates increased copy

268 number across a variety of cancers. Given that FOXS1 is not expressed in normal RPE nor in

269 other adult epithelia, its expression and involvement in pathological EMT indicate it may be a

270 valuable therapeutic target across several tissue types.

271

272 Acknowledgements

273 We thank Dr. Barbara Corneo for manuscript review, Carol Charniga and Dr. Marie Fernandes

274 for their invaluable help with eye dissection and cell culture, and the eye donors and their

275 families for the generous donation of retinal tissue. This study was supported by a National Eye

276 Institute (NEI; Bethesda, MD, USA) extramural grant (1R01EY022079; ST) and by NYSTEM

277 through a retinal stem cell consortium grant (N11C012) and by the Icahn School of Medicine at

278 Mount Sinai (TAB) and by a gift from Dr. Heinrich Medicus to the Regenerative Research

279 Foundation (S.T. and J.H.S).

280

281 Author contributions

282 TAB, ST, JHS and JW conceived the experiments, and helped with data analysis, interpretation

283 and manuscript writing. TAB performed the experiments, analyzed data, made figures, and

284 wrote the manuscript. TAB and QW produced shRNA and overexpression FOXS1 lentiviruses, RS

285 prepared RNA and DNA for sequencing, TS and NB analyzed sequencing data and made figures. bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

286 ARI performed 1 ChIP-seq and data analysis. TAB wrote the initial manuscript draft, which was

287 edited primarily by ST and JW and additionally by TS, NB, RS, QW, JHS.

288

289 Author Information

290 Data deposition with URL and accession database numbers

291

292 Competing financial interests

293 TAB, JHS, ST have filed a provisional patent on the method by which FOXS1 is expressed in RPE

294 and RPE related diseases.

295 Correspondence

296 Correspondence and requests for materials should be addressed to

297 [email protected] for questions regarding experimentation,

298 [email protected] for conceptual questions, and [email protected] for

299 bioinformatic analytical questions.

300

301 Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY,

302 10029, USA

303 Timothy A Blenkinsop

304

305 Macular Degeneration Program, Neural Stem Cell Institute, Rensselaer NY, 12144, USA

306 Nathan Boles, Qingjie Wang, Jeffrey H Stern, Sally Temple

307 bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

308 Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA,

309 94305, USA

310 Tomasz Swigut, Rajini Srinivasan, Joanna Wysocka

311

312 Howard Hughes Medical Institute, Stanford, CA, 94305, USA

313 Joanna Wysocka

314

315 University of Cologne, Cologne, 50931, Germany

316 Alvaro Rada-Iglesias

317

318

319 References cited

320 1 Grisanti, S. & Guidry, C. Transdifferentiation of retinal pigment epithelial cells from epithelial to 321 mesenchymal phenotype. Invest Ophthalmol Vis Sci 36, 391-405 (1995). 322 2 Ho, P. C., Yoshida, A., Schepens, C. L., McMeel, J. W. & Duncan, J. E. Severe proliferative 323 vitreoretinopathy and retinal detachment. I. Initial clinical findings. Ophthalmology 91, 1531- 324 1537 (1984). 325 3 Limb, G. A. et al. Cytokines in proliferative vitreoretinopathy. Eye (Lond) 5 ( Pt 6), 686-693, 326 doi:10.1038/eye.1991.126 (1991). 327 4 Rojas, J. et al. A strong genetic association between the tumor necrosis factor locus and 328 proliferative vitreoretinopathy: the retina 4 project. Ophthalmology 117, 2417-2423 e2411- 329 2412, doi:S0161-6420(10)00378-7 [pii] 330 10.1016/j.ophtha.2010.03.059 (2010). 331 5 Sanabria Ruiz-Colmenares, M. R., Pastor Jimeno, J. C., Garrote Adrados, J. A., Telleria Orriols, J. J. 332 & Yugueros Fernandez, M. I. Cytokine gene polymorphisms in retinal detachment patients with 333 and without proliferative vitreoretinopathy: a preliminary study. Acta Ophthalmol Scand 84, 334 309-313, doi:10.1111/j.1600-0420.2005.00600.x (2006). 335 6 Blenkinsop, T. A. et al. Human Adult Retinal Pigment Epithelial Stem Cell-Derived RPE 336 Monolayers Exhibit Key Physiological Characteristics of Native Tissue. Invest Ophthalmol Vis Sci 337 56, 7085-7099, doi:10.1167/iovs.14-16246 (2015). 338 7 Tomasek, J. J., Gabbiani, G., Hinz, B., Chaponnier, C. & Brown, R. A. Myofibroblasts and 339 mechano-regulation of connective tissue remodelling. Nature reviews. Molecular cell biology 3, 340 349-363, doi:10.1038/nrm809 (2002). bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

341 8 Abu El-Asrar, A. M., Missotten, L. & Geboes, K. Expression of myofibroblast activation molecules 342 in proliferative vitreoretinopathy epiretinal membranes. Acta Ophthalmol 89, e115-121, 343 doi:10.1111/j.1755-3768.2010.01916.x (2011). 344 9 Sato, Y. et al. Importance of forkhead transcription factor Fkhl18 for development of testicular 345 vasculature. Molecular reproduction and development 75, 1361-1371, doi:10.1002/mrd.20888 346 (2008). 347 10 Cederberg, A. et al. localization, sequence analysis, and expression pattern 348 identify FKHL 18 as a novel human forkhead gene. Genomics 44, 344-346, 349 doi:10.1006/geno.1997.4864 (1997). 350 11 Montelius, A. et al. Emergence of the sensory nervous system as defined by Foxs1 expression. 351 Differentiation 75, 404-417, doi:10.1111/j.1432-0436.2006.00154.x (2007). 352 12 Calo, E. & Wysocka, J. Modification of enhancer chromatin: what, how, and why? Mol Cell 49, 353 825-837, doi:10.1016/j.molcel.2013.01.038 (2013). 354 13 Creyghton, M. P. et al. Histone H3K27ac separates active from poised enhancers and predicts 355 developmental state. Proc Natl Acad Sci U S A, doi:1016071107 [pii] 356 10.1073/pnas.1016071107 (2012). 357 14 Rada-Iglesias, A. et al. A unique chromatin signature uncovers early developmental enhancers in 358 humans. Nature, doi:nature09692 [pii] 359 10.1038/nature09692 (2010). 360 15 Heintzman, N. D. et al. Distinct and predictive chromatin signatures of transcriptional promoters 361 and enhancers in the . Nat Genet 39, 311-318, doi:ng1966 [pii] 362 10.1038/ng1966 (2007). 363 16 Cerami, E. et al. The cBio cancer genomics portal: an open platform for exploring 364 multidimensional cancer genomics data. Cancer discovery 2, 401-404, doi:10.1158/2159- 365 8290.CD-12-0095 (2012). 366 17 Gao, J. et al. Integrative analysis of complex cancer genomics and clinical profiles using the 367 cBioPortal. Science signaling 6, pl1, doi:10.1126/scisignal.2004088 (2013). 368 18 Yamauchi, Y. et al. Tumor necrosis factor-alpha enhances both epithelial-mesenchymal 369 transition and cell contraction induced in A549 human alveolar epithelial cells by transforming 370 growth factor-beta1. Experimental lung research 36, 12-24, doi:10.3109/01902140903042589 371 (2010). 372 19 Bates, R. C. & Mercurio, A. M. Tumor necrosis factor-alpha stimulates the epithelial-to- 373 mesenchymal transition of human colonic organoids. Molecular biology of the cell 14, 1790- 374 1800, doi:10.1091/mbc.E02-09-0583 (2003). 375 20 Bochaton-Piallat, M. L. et al. TGF-beta1, TGF-beta receptor II and ED-A fibronectin expression in 376 myofibroblast of vitreoretinopathy. Invest Ophthalmol Vis Sci 41, 2336-2342 (2000). 377 21 Kita, T. et al. Role of TGF-beta in proliferative vitreoretinal diseases and ROCK as a therapeutic 378 target. Proc Natl Acad Sci U S A 105, 17504-17509, doi:10.1073/pnas.0804054105 (2008). 379 22 Schillebeeckx, M. et al. Novel markers of gonadectomy-induced adrenocortical neoplasia in the 380 mouse and ferret. Molecular and cellular endocrinology 399, 122-130, 381 doi:10.1016/j.mce.2014.09.029 (2015). 382 23 Blenkinsop, T. A., Salero, E., Stern, J. H. & Temple, S. The culture and maintenance of functional 383 retinal pigment epithelial monolayers from adult human eye. Methods Mol Biol 945, 45-65, 384 doi:10.1007/978-1-62703-125-7_4 (2013). 385 24 Phoenix, T. N. & Temple, S. Spred1, a negative regulator of Ras-MAPK-ERK, is enriched in CNS 386 germinal zones, dampens NSC proliferation, and maintains ventricular zone structure. Genes Dev 387 24, 45-56, doi:10.1101/gad.1839510 (2010). bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

388 25 Wang, Q., Yang, L., Alexander, C. & Temple, S. The niche factor syndecan-1 regulates the 389 maintenance and proliferation of neural progenitor cells during mammalian cortical 390 development. PLoS One 7, e42883, doi:10.1371/journal.pone.0042883 (2012). 391 26 Rada-Iglesias, A. et al. A unique chromatin signature uncovers early developmental enhancers in 392 humans. Nature 470, 279-283, doi:nature09692 [pii] 393 10.1038/nature09692 (2011). 394 27 Buecker, C. et al. Reorganization of enhancer patterns in transition from naive to primed 395 pluripotency. Cell Stem Cell 14, 838-853, doi:10.1016/j.stem.2014.04.003 (2014). 396 397 bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

398 Figures

399 Figure 1

A B LOG SNAIL LOG SLUG TWIST Dissect Scrape 10 10 15 100 ** 100 Control Control Control ** ** TGFβ TGFβ ** TGFβ ) A

N 10 R 10 TNFα 10 TNFα TNFα m

Vitreous (

. TNT TNT TNT Plate t uan Q . e 5 R 1 1 Culture 4 months

0 0 0 0 1 3 5 0 1 3 5 0 1 3 5 0 1 3 5 0 1 3 5 0 1 3 5 0 1 3 5 0 1 3 5 0 1 3 5 0 1 3 5 0 1 3 5 0 1 3 5 Day Day Day C D αSMA E SNAIL Control TGFβ

Control TGFβ Control TGFβ Control TGFβ TNFα TNT

TNFα TNT TNFα TNT TNFα TNT

G Control TGFβ TNFα TNT F 9 0 1.5 )

A MITF N ** R 8 0 m (

. 1.0 t NLB

uan 7 0 Q .

e 0.5 R ** ** 6 0 0.0 TGFβ + + + R T TNFα + PE R PV TN H 60 30 BEST1 SNAIL ) ) A A ** N N R R m m ( ( 40 20

. . t t

uan uan ** Q Q . 20 . 10 e e R R

0 0 E R T R T P V PE V R P TN R P TN Control TGFβ TNFα TNT TNT + SB202190

I J K 10 FOXS1 SNAIL SLUG TWIST FOXS1 ) FOXL1 HIC1

500 5 A 600 4 MT3 POU2F2 CREB3L1 ) A N 100 e SOX17 R g 400 POU3F2 TCF4 VDR N R m ** ** 80 ( 3

m . ( t 400 0 ** 300 .

** t 60 2 uan

** old Chan uan 200 F Q HLF FOXQ1SOX8 40 2 . 200 TEF OTX2 Q e . 1 -5 PGR CSRNP3 R

e 20 100 OG DRGX GSX2 MITF R ** L KLF15 HNF4A ** 0 0 0 NR2E1 0 TGFβ + + + + + + + + + -10 TGFβ + + + + + + TNFα + + + + + + + + + TNFα + SB202190 + + + 1e+01 1e+02 1e+03 1e+04 1e+05 SB202190 400 Mean expression bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

401 Figure 1. TGFβ1+TNFα co-treatment of cobblestone RPE induces PVR-like EMT 3D masses. A)

402 Schematic of the method to obtain pure cultures of RPE. B) Time course of SNAIL, SLUG and

403 TWIST expression in control (cobblestone RPE, vehicle treated) and when treated with 10ng/ml

404 of TGFβ1 and/or TNFα. (C-E) Comparison of RPE cells in control conditions or 5 days after

405 treatment with 10ng/ml of TGFβ1 and/or TNFα. C) Phase images D) Immunostaining with anti-

406 SNAIL antibody. E) Immunostaining with anti-αSMA antibody. F) Fundus images of patient with

407 PVR membranes (indicated by dotted line) and gene expression comparison between TNT RPE

408 and PVR samples from two patients. G) Immunofluorescence of p38 localization Inset:

409 DAPI:blue, p38: green (NL=nuclear luminescence. H) Phase contrast images of RPE at day 5 of

410 treatment indicated. I) qPCR of SNAIL, SLUG and TWIST from RPE after 5 days of treatment

411 indicated. J) Scatterplot of RNA-seq data from cobblestone RPE versus TNT RPE with

412 transcription factors highlighted; blue= increased and red= decreased expression. K) qPCR of

413 FOXS1 transcript after treatment indicated. Graph bars indicate SEM. Image scale Bars = 50μm.

414 ** = P-value ≤ 0.01.

415 bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

416 Figure 2 TNT [ChIP signal] TNT [ChIP signal] TNT [ChIP

TNT [ChIP signal] TNT [ChIP signal] H3K4me3 H3K4me1 TNT:log

H3K4me1 RPE:log distance from TSS [bp] log2 fold expression changes of the closest gene H3K4me3

5 kb

417 bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

418 Figure 2. TGFβ1+TNFα co-treatment of cobblestone RPE induces massive epigenetic changes

419 at enhancer elements. A) TGFβ1+TNFα co-treatment induced changes in H3K27 acetylation:

420 ordinate - normalized read density of H3K27ac ChIP-seq at chromatin features (putative

421 enhancers and promoters) in cobblestone RPE cells, abscissa - normalized read density of

422 H3K27ac ChIP-seq in TGFβ1+TNFα treated cells. Regions indicated in red have significantly

423 upregulated ChIP signal upon TGFβ1+TNFα treatment (FDR<0.01), in blue downregulated

424 (FDR<0.01), in orange with no change (FDR<0.1). B) Changes in the H3K4me1 ChIP signal are

425 correlated with H3K27ac changes, regions are color coded according to the H3K27ac classes

426 defined in the previous panel C) TGFβ1+TNFα co-treatment results in relatively few changes in

427 promoter signatures. Plotted is the log ratio of H3K4me1 versus H3K4me3 ChIP density; RPE on

428 the ordinate, treated cells on the abscissa. Negative values are indicative of a promoter-like

429 chromatin signature at interrogated sites. Color-coding as in previous panels. D) Most changes

430 in H3K27 acetylation occur at sites distal from annotated transcription start sites. Plotted is the

431 absolute distance to the closest annotated TSS versus the log2 fold change in the H3K27ac ChIP

432 signal. E) Changes in H3K27ac are correlated with changes in gene expression. Plotted are the

433 distribution of log2 fold expression changes for genes associated with distal elements that have

434 upregulated H3K27ac (red), downregulated H3K27ac (blue) or were unchanged (orange). The

435 differences are significant (Mann-Whitney-Wilcoxon test). F) Visualization of the histone

436 modification changes at the FOXS1 locus after TGFβ1+TNFα treatment in the UCSC browser;

437 H3K27ac density in green, H3K4me1 in blue. Location of two major FOXS1 enhancers is

438 indicated by black bars.

439 bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

440 Figure 3 n A n FOXS1 B 2.0 FOXS1 SNAIL SLUG TWIST o o 2.0 2.0 2.0 t t a a 10000 c c fi fi 1.5 1.5 t t 1.5 1.5 1000 ** quan quan 100 1.0

** 1.0 1.0 1.0 e e v v

t 10 t a a l l 0.5 0.5 0.5 0.5 e e r r 1 **

A A ** 0.1 0.0 ** 0.0 0.0 0.0 RN RN

E D D D m m D + V + V + V + V K K K K RP PVR TNT T T T T 1 +S 1 +S 1 +S 1 +S N N N N T T T T T T T T XS XS XS XS N N N N T T T T FO FO FO FO C D

n FOXS1 SNAIL SLUG TWIST o 30 2.5 5 t ** a c 10000 ** 2.0 ** 4 fi t 1000 20 1.5 3 ** 100 quan 2 e 1.0

v 10 10 t a

l 0.5 1

e 1 r

A 0.1 0 0.0 0

TNT +SV TNT + FOXS1 KD RN SV FOXS1 OE SV FOXS1 OE SV FOXS1 OE SV FOXS1 OE m E F n o MITF OTX2 BEST t 2.0 20 a 2.0 c fi t 1.5 1.5 15 ** quan 1.0 1.0 10 e v t a

l 0.5 0.5 5 e ** r

A ** 0.0 0.0 0 RN SV FOXS1 OE SV FOXS1 OE SV FOXS1 OE m SV FOXS1 OE 441

442 Figure 3. FOXS1 is necessary and sufficient to drive SNAIL and SLUG expression in RPE. A)

443 qPCR gene expression comparison between TNT-RPE and PVR samples from two patients. B)

444 qPCR of RPE after 5 days of TNT and either a scrambled, or FOXS1 shRNA lentivirus treatment

445 and C) corresponding color images. qPCR gene expression comparison of RPE with either a

446 scrambled or FOXS1 overexpression lentivirus of EMT (D) and RPE (E) genes. F) Color images of

447 RPE at day 5 after treatment with either a scrambled or FOXS1 overexpression lentivirus. Image

448 Scale bars= 100mm. ** indicates P-value ≤0.01. Graph bars = SEM. SV=scrambled control

449 vector.

450 bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

451 Figure 4

A B FOXS1 FOXS1 5 FOXS1 Control TGFβ 15 1.5 1.5x10 ** 5 10 ** 1.0 1.0x10 ** 5 5 0.5 .5x10 ** ** 0 0.0 0 l T V V 0 TNT + D S Contro TGFβ TNFα TN TNT + T + S TN FOXS1 OE SB20219 FOXS1 K TNFα TNT 30 SNAIL 1.5 SNAIL 5 SNAIL 4 ** 20 ** 1.0 3 0.5 2 10 ** 1 ** ** 0 0.0 0 l V T 0 TNT + D SV TGFβ TNFα TN TNT + T + S Contro TN FOXS1 OE SB20219 FOXS1 K

C FOXS1 FOXS1 FOXS1 SLUG 1.5 80000 150 10000 ** 60000 8000 1.0 100 ** 6000 * 40000 0.5 50 4000 ** 20000 2000 0.0 0 0 0 l l T 0 V D T 0 SV TGFβ TNFα TN Contro TGFβ TNFα TN TNT + Contro TNT + NT + S SB20219 T FOXS1 K FOXS1 OE SB20219 SLUG SLUG TWIST TWIST TWIST 1.5 15 10 1.5 20 8 15 1.0 10 ** 1.0 6 ** 10 0.5 5 4 0.5 ** 5 2 0.0 0 0 0.0 0 l V D V T 0 V D SV S TN TGFβ TNFα TNT + T + S NT + S Contro N T FOXS1 K FOXS1 OE SB20219 T FOXS1 K FOXS1 OE

D 25 64

FOXS1 NU

32 M

C ASES

N 20 16 IO T

W A

R 8 IT E H T FO L 15 A

4 XS 1 1 XS

A O 2 F L

T H E

T 10 I R

1 A W

TIO N ASES C 5 %

0 l l a a a l r r r e e e t t t r r L L s n n h h h g g g g g g d d 0 T C C e e M E ia ia v v r r rea AM AM AC Li Li Lun Lun Lun Lun Lun Lun a a GB Hea Hea c DLB CCL ladde ladde ladde Breas Breas Breas NCI-6 Uterin Uterin Uterin B B B MPNS Ov Ov Cervica an Prostate Prostate Stomac Stomac Stomac Sarcoma Sarcoma P Colorecta Colorecta Melanom Melanom Melanom Esophagus Esophagus STUDY ABBREVIATION 452 bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

453 Figure 4. FOXS1 in EMT in other epithelia and cancer. A) qPCR of HME cells treated with TGFβ1

454 and/or TNFα with and without p38 inhibitor SB202190; Lentivirus treatment including a

455 scrambled, or FOXS1 shRNA in HME cells with TNT; lentivirus treatment of HME cells with either

456 a scrambled or FOXS1 overexpression construct. B) Immunofluorescence of HME cells treated

457 with TGFβ1 and TNFα stained with anti-SNAIL antibody. C) qPCR of HHE cells treated with

458 TGFβ1 and/or TNFα with and without p38 inhibitor SB202190; Lentivirus treatment including a

459 scrambled, or FOXS1 shRNA in HHE cells with TNT; lentivirus treatment of HHE cells with either

460 a scrambled, or FOXS1 overexpression construct. D) Cross cancer analysis shows FOXS1

461 alterations are present in 47 of 92 studies examined (See Sup Table 1). Graph scale bars = SEM.

462 Image scale Bars = 200μm. * = P-value ≤ 0.05. ** = P-value ≤ 0.01.

463 bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

464 Figure 5

Extracellular space TGFβ

R TNF β α TGF

TNFαR plasma membrane

p38 Cytoplasm

nuclear pore nuclear membrane Epithelial cell

FOXS1 p38

Nucleus FOXS1

SNAIL SLUG TWIST

RPE PVR SNAIL αSMA Collagen

465

466 Figure 5. Model of the pathway of synergistic action of TGFβ and TNFα in transforming RPE

467 into 3D contractile tissue. TGFβ and TNFα bind to their respective receptors leading to a co-

468 activation of p38, which leads to p38 translocation into the nucleus and transcriptional

469 activation of FOXS1. FOXS1 expression leads to SNAIL, SLUG, and TWIST expression, driving an

470 epithelial to mesenchymal transition in RPE.

471 bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

472 Methods

473 Human adult RPESC culture

474 Human globes from donors aged between 51 and 89 yrs were obtained from the National

475 Disease Research Interchange, Philadelphia, PA., the Eye-Bank for Sight Restoration, Inc., New

476 York, NY, and the Lions Eye Bank, Albany, NY. A detailed eye dissection protocol was previously

477 published 23. Globes were obtained within 40 hours of death, RPE sheets were isolated and

478 plated on tissue culture plates coated with 10μg/ml placental ECM (Corning) or Synthemax II

479 (Corning) in RPE medium 23, supplemented with 10% FBS and 10mM Nicotinamide, which was

480 changed 3 times a week. After the first week, FBS was reduced to 2%.

481

482 EMT model

483 Passage 0 adult human RPE cultures (ahRPE) were trypsinized using 0.25% Trypsin for 15

484 minutes, washed and replated at 3 x104 cells per 1.9mm2 per well of a 24-well plate in

485 DMEM/F12 with 5%FBS, L-Glutamine, Na-Pyruvate, NEAA, Pen/Strep. After 24 hours, 10ng/ml

486 TGFβ1 or TNFα, or both (TNT) were added to induce EMT versus vehicle as control, and the

487 cultures were maintained in this medium (with feeding every other day) for 5 days.

488

489 Immunohistochemistry

490 ahRPE on 24 well-size transwell inserts (Corning) were fixed with 4% paraformaldehyde for 10

491 minutes, rinsed 3 times with phosphate buffered saline (PBS), permeabilized with 0.01%

492 saponin and blocked with normal goat serum (5%) in 1% BSA in PBS for 1 hour. Primary

493 antibody (Table 1) was added overnight at 4oC, then incubated with the corresponding Alexa bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

494 Fluor conjugated secondary antibodies (1:1000) (Life Technologies Alexa Fluor 647 goat anti-

495 mouse IgG (H+L) (Cat#: A-21237), Alexa Fluor 647 goat anti-rabbit IgG (H+L) (Cat#: A-21244), at

496 room temperature for 45 minutes. Cells on transwell inserts were then mounted on glass slides

497 with Prolong gold (Life Technologies) and imaged by confocal microscopy (Leica). DAPI

498 fluorescence was used to demarcate the nucleus in experiments measuring nuclear p38. P38

499 fluorescence intensity was then measured exclusively in the DAPI demarcated region based on

500 a 0-256 scale using ImageJ analysis software. The fluorescence intensity of all nuclei was

501 averaged across experiments and compared between conditions using a student’s t-test.

502

503 qPCR

504 RNA was extracted (Qiagen) and reverse-transcribed (Superscript III Reverse Transcriptase kit)

505 then qPCR was performed according to manufacturer instructions (Power SybrGreen PCR

506 Master Mix) using primers in Table 2. Data was recorded as the cycle by which sybr

507 fluorescence detection threshold is reached during the exponential increase phase. All cycles

508 for queried genes were normalized to the housekeeping gene cyclophilin to control for cDNA

509 quantity variation.

Table 2 List of primers used for Real Time PCR on adult human RPE

Product Human Tann Gene Bank Forward 5'-3' Reverse 3'-5' Size Gene (oC) ID (bp)

RPE65 TACAGAAAGCACTGAGTTGAGC CCATTTAGTAAGTCCACATTCATTTCC 153 55 NM_000329 bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

OTX2 CCATGACCTATACTCAGGCTTCAGG GAAGCTCCATATCCCTGGGTGGAAAG 211 60 NM_000326

MITF TTGTCCATCTGCCTCTGAGTAG CCTATGTATGACCAGGTTGCTTG 87 65 NM_004183

BESTROPHIN CCTTTATGGGCTCCACCTTCAACATC CAGTAGTTTGGTCCTTGAGTTTGCCC 166 65 NM_004183

SNAIL TGTCAGATGAGGACAGTGGGAAAGG CTGAAGTAGAGGAGAAGGACGAAGG 611 53 NM_005985

SLUG AGCGAACTGGACACACATAC TCTAGACTGGGCATCGCAG 410 55 NM_003068

TWIST GTCCGCAGTCTTAGCAGGAG GCTTGAGGGTCTGAATCTTGCT 156 60 NM_000474

FOXS1 AGTGGCATCTACCGCTACATC CACCTTGACAAAGCACTCGT 114 63 NM_004118

CYCLOPHILIN CTTGTCAATGGCCAACAGAGG GCCCATCTAAATGAGGAGTTGGT 82 60 NM_004792 G

510

511 Lentiviral vector production and viral packaging

512 shRNA plasmids: the hairpin oligonucleotides (Scrambled control: TTCTCCGAACGTGTCACGT;

513 FOXS1 3’ UTR: GCCAATAAAGCCATGTGAT) were inserted into the FUGW-H1 lentiviral construct

514 as previously described 24,25. To package the lentivirus, the constructs were co-transfected with

515 pCMV-VSVG and pCMV-dvpr into 293FT cells. Supernatant was harvested 2 and 3 days later and

516 concentrated by ultra-centrifugation. For overexpression constructs, the open reading frame

517 (ORF) of each gene was PCR-amplified, sequence-verified, and cloned into a modified version of

518 FUGW. An IRES-eGFP was included for visualization. Lentiviruses were used at 10 MOI for cell

519 transduction.

520 bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

521 ChIP-seq

522 ChIP assays were performed from 107 RPE cells per experiment, according to a previously

523 described protocol with slight modification 26. Briefly, RPE cells at passage 0 cultures were

524 treated with TGFβ1, TNFα or TNT conditions for 5 days. The cells were then with 1%

525 formaldehyde for 10 minutes at room temperature to crosslink, and the reaction was then

526 quenched by adding glycine at a final concentration of 0.125 M. Cells were dissolved in lysis

527 buffer and chromatin was sonicated to an average size of 0.5–2 kb, using Bioruptor

528 (Diagenode). 5–7.5 μg of antibody was added to the sonicated chromatin and incubated

529 overnight at 4°C. Subsequently, 50μl of protein G Dynal magnetic beads were added to the ChIP

530 reactions and incubated for 4–6 hr at 4°C. Magnetic beads were washed and chromatin eluted,

531 followed by reversal of crosslinks and DNA purification. ChIP-seq and input libraries were

532 prepared according to Illumina protocols and sequenced using the Illumina HiSeq. To identify

533 the approximate positions of regulatory elements from histone modification ChIP-seq profiles

534 alone, we calculated kernel density estimate tracks with bi-modal kernels 27 and identified

535 peaks in the product of the combined H3K27 and H3K4 signals. To analyze changes in histone

536 modifications, we calculated the read coverage for each sample over a combined set of

537 detected peaks and performed a differential analysis with DESeq2.

538

539 RNA-seq

540 RPE cells from two different donors were treated with TGFβ1, TNFα or TNT or vehicle control as

541 described above for 5 days, then the RNA was extracted with Trizol (Invitrogen), following the

542 manufacturer’s recommendations. 10ug of total RNA was subjected to two rounds of bioRxiv preprint doi: https://doi.org/10.1101/154369; this version posted June 26, 2017. 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.

543 purification using Dynaloligo-dT beads (Invitrogen) then fragmented with 10× fragmentation

544 buffer (Ambion) and used for first-strand cDNA synthesis, using random hexamer primers

545 (Invitrogen) and SuperScript II enzyme (Invitrogen). Second strand cDNA was obtained by

546 adding RNaseH (Invitrogen) and DNA Pol I (New England BioLabs). The resulting double-

547 stranded cDNA was used for Illumina library preparation and sequenced with the Illumina

548 HiSeq. Reads were mapped with tophat v2.0.14 to the gencode 23 transcriptome model and

549 the read coverage was calculated with htseq. Differential analysis was performed with DESeq2.

550