<<

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

1

1 Title: In Vivo Mechanisms of -Induced Acute Follicle Loss in the Human Ovary:

2 An Individual-Oocyte Transcriptomic Analysis from Human Ovarian Xenografts

3

4 Running Title: Chemotherapy-Induced Damage to Ovarian Reserve

5

6 One Sentence Summary: Single-cell transcriptomic interrogation of primordial follicles in

7 human ovarian xenografts reveals that chemotherapy causes acute ovarian reserve depletion by

8 inducing a pro-apoptotic state rather than activating pathways that result in follicle growth

9 initiation.

10

11 Keywords: , female infertility, follicle, gene expression, ovary, premature ovarian

12 failure, transcription, transcriptional regulation

13

14 S. Titus1, K.J. Szymanska1, B. Musul1, V. Turan1, E. Taylan1, R. Garcia- Milian2, S.

15 Mehta3, K. Oktay1*

16

17 1 Yale University School of Medicine, Department of Obstetrics, Gynecology and Reproductive

18 Sciences, New Haven, CT, USA

19 2 Bioinformatics Support Program, Yale School of Medicine, New Haven, CT, USA

20 3 Yale Center for Genome Analysis, Yale University, New Haven, CT, USA

21

22

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

2

23 Abstract: Gonadotoxic chemotherapeutics, such as , cause early menopause

24 and infertility in women. Earlier histological studies showed ovarian reserve depletion via

25 severe DNA damage and apoptosis, but others suggested activation of PI3K/PTEN/Akt

26 pathway and follicle ‘burn-out’ as a cause. Using a human ovarian xenograft model, we

27 performed single-cell RNA-sequencing on laser-captured individual primordial follicle oocytes

28 12h after a single cyclophosphamide injection to determine the mechanisms of acute follicle

29 loss after gonadotoxic chemotherapy. RNA-sequencing showed 190 differentially expressed

30 genes between the cyclophosphamide- and vehicle-exposed oocytes. Ingenuity Pathway

31 Analysis predicted a significant decrease in the expression of anti-apoptotic pro-Akt PECAM1

32 (p=2.13E-09), IKBKE (p=0.0001), and ANGPT1 (p=0.003), and reduced activation of

33 PI3K/PTEN/Akt after cyclophosphamide. The qRT-PCR and immunostaining confirmed that

34 in primordial follicle oocytes, cyclophosphamide did not change the expressions of Akt (p=0.9),

35 rpS6 (p=0.3), Foxo3a (p=0.12) and anti-apoptotic Bcl2 (p=0.17), nor affect their

36 status. There was significantly increased DNA damage by γH2AX (p=0.0002)

37 and apoptosis by active-caspase-3 (p=0.0001) staining in the primordial follicles and no change

38 in the growing follicles 12h after chemotherapy. These data suggest that the mechanism of acute

39 follicle loss by cyclophosphamide is via apoptosis, rather than growth activation of primordial

40 follicle oocytes in the human ovary.

41

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

3

42 INTRODUCTION

43 In 2019, approximately 891,480 women were diagnosed with cancer in the U.S. alone, of whom

44 5.5% were in reproductive age [1], and potentially received fertility-damaging cancer

45 treatments. Worldwide, chemotherapy-induced premature menopause and infertility are

46 significant quality of life issues for young women diagnosed with cancer. To preserve fertility

47 in the face of gonadotoxic chemotherapy, successful gamete, and gonadal tissue

48 cryopreservation techniques have been developed [2]. However, these require at least minimally

49 invasive procedures, and there have been no proven targeted medical treatments to block the

50 ovarian damaging side effects of chemotherapy. If the mechanisms of chemotherapy-induced

51 ovarian damage are understood, noninvasive, targeted pharmacological methods of fertility

52 preservation can be developed.

53 Based on our earlier work, we suggest that chemotherapy causes both acute and delayed effects

54 on the primordial follicle reserve. Chemotherapy agents such as , a

55 inhibitor [3], and cyclophosphamide, an alkylating agent [4][5], cause apoptotic primordial

56 follicle death by inducing DNA double-strand breaks (DSBs) in the human ovary. We found

57 that the DNA damage peaks at around 12h and subsequent follicle loss at 48h, accounting for

58 the acute loss of follicle reserve. Besides, we also showed in in vivo human ovarian xenograft

59 models that doxorubicin causes stromal cell death as well as microvascular damage, inducing

60 tissue hypoxia, which may contribute to the delayed loss of ovarian follicles. Others have also

61 shown possible stromal damaging effects of cancer in women [6]. It has also been

62 proposed in rodent models that cyclophosphamide may induce depletion of primordial follicle

63 reserve via inducing a massive entry of follicles into the growth phase [7]. It was surmised that

64 this follicle ‘burn-out’ is the acute result of the direct activation of phosphoinositide 3-kinase/

65 phosphatase and tensin homolog/ protein kinase B (PI3K/PTEN/Akt) pathways, which regulate

66 primordial follicle growth initiation. There could be several reasons for these discrepancies,

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

4

67 including the possibility of the differing mechanism being in play at acute and delayed phases

68 after the chemotherapy exposure [8]. Therefore, mechanistic studies are needed to delineate the

69 acute impact of chemotherapy on ovarian follicle reserve. Utilizing the human ovarian

70 xenografting coupled with laser-capture-microdissection-assisted individual-oocyte RNA-

71 sequencing (RNA-seq) and quantitative real-time PCR (qRT-PCR) approaches perfected by our

72 team, we performed this study to determine the molecular mechanisms of acute chemotherapy-

73 induced loss of primordial follicles in the human ovary [4][5].

74 MATERIALS AND METHODS

75 Mice

76 Female severe combined immunodeficiency (NOD-SCID) mice aged 2-3 months were obtained

77 from Taconic farms and housed under pathogen-free conditions. Animals were kept in a 12 h

78 light-dark cycle and provided ad libitum with water and a laboratory diet. All animal

79 experiments were approved and conducted in accordance with the protocol of the Yale

80 University Institutional Animal Care and Use Committee.

81 Ovarian Xenografts

82 Ovarian cortical pieces from cadaveric organ donors aged ≤ 32 years were cryopreserved

83 according to our established protocol [9,10]. Tissue pieces 2-3 mm3 (2-3 × 1 × 1 mm) were

84 xenografted subcutaneously to SCID mice, as described previously [4]. Briefly, after animals

85 were anesthetized, a small incision was made on the dorsal midline of the animal. Human

86 ovarian cortex fragments were transplanted to each animal (n=4/animal). After allowing

87 xenografts to fully vascularize for 10 days, the mice were given a single intraperitoneal (i.p.)

88 dose of cyclophosphamide (75 mg/kg) or the vehicle. The tissues were then recovered 12h later

89 and prepared for either histology and immunohistochemistry or embedded in a medium for

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

5

90 frozen sectioning and isolation of primordial follicle oocytes by laser capture dissection

91 microscopy.

92 Immunohistochemistry

93 Ovarian tissues were fixed in 4% paraformaldehyde (PFA), embedded in paraffin, and serially

94 sectioned at 5-micron intervals. Sections were stained with Hematoxylin and Eosin (H&E) for

95 differential follicle counts, as previously described [4]. Every fifth section of an entire xenograft

96 was chosen for staining and evaluated by observers who were blinded to the intervention and

97 each other's findings. To avoid over-counting, follicles were recorded only when the nucleus

98 was visible. Follicles were classified based on our previously established criteria [11]. A

99 primordial follicle was defined as an oocyte surrounded by a single layer of flattened, squamous

100 follicular cells. A primary follicle was defined as an oocyte surrounded by a single layer of

101 cuboidal granulosa cells. A follicle was classified atretic based on morphological criteria,

102 including nuclear pyknosis, cytoplasm contraction, and nuclear pyknosis of granulosa cells and

103 dissociation of the granulosa cells from the basal membrane [12,13].

104 Randomly selected sections coming from ovarian tissue of least 3 different individuals were

105 used for immunohistochemistry for Anti-active Caspase-3 (AC-3, 1:1000, AF-835, R&D

106 Systems), γH2AX (1:250 IHC-00059, Bethyl Laboratories), rabbit phospho-Akt (S473, 1:400,

107 #4060S, Cell Signaling Technology, Denver, MA), rabbit phospho-rpS6 (S235-236, 1:100;

108 #2211S, Cell Signaling Technology, Denver, MA), and phospho-FOXO3A (S253, 1µg/ml, #

109 ab47285, Abcam, Cambridge, MA, USA). Images were captured on an Olympus IX73

110 microscope. BAD (1:250, A302-384A, Bethyl Laboratories) and Bcl2 (1:200, sc-509, Santa

111 Cruz Biotechnology). Nikon 90i Eclipse microscope was used for imaging. By overlapping both

112 BAD and Bcl2 staining color channels, and selecting the area around primordial follicles, we

113 calculated the mean pixel intensities. We compared the intensity means between primordial

114 follicle oocytes of vehicle and chemo groups after subtracting the background by setting a

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

6

115 constant threshold for both channels across the images. For the calculation, we used 3 slides of

116 each xenografted tissue, and 'n' represents the number of different tissue pieces. The analysis

117 was done using ImageJ software.

118 Laser-capture microdissection (LCM) and isolation of single primordial follicle oocytes

119 After harvesting ovarian xenografts, tissue blocks were immediately prepared in optimum

120 cutting temperature (OCT) compound and stored at -80°C until cryostat sectioning (-20°C).

121 These were later sectioned at 8-micron thickness and stained with H&E according to the

122 manufacturer's protocol. Based on the morphological appearance, i.e., follicle with a single

123 layer of flat granulosa cells around with positively stained oocyte inside, single primordial

124 follicle oocytes were individually isolated using an LCM excluding any pre-granulosa cell from

125 the dissection. To each Eppendorf tube, filled with cell lysis buffer (RNase inhibitor in 0.2%

126 (vol/vol) Triton X-100 solution), the individual primordial oocyte was collected from 2-3

127 adjacent tissue sections to ensure that one full cell was included (Leica Microsystems, Buffalo

128 Grove, IL, USA).

129 RNA-seq analysis on laser captured single primordial follicle oocytes

130 The primordial follicle oocytes were processed for sequencing with modifications of a

131 previously established protocol on non-germline single cells for LCM material [14]. Samples

132 were sequenced to depths of up to 44 million read pairs, 75 nt length reads per sample using the

133 Illumina Rapid v2 kit (75 cycles) on an Illumina HiSeq2500 Sequencing System. Image analysis,

134 base calling, and generation of sequence reads were produced using the HiSeq Control Software

135 v2.0 (NCS) and Real-Time Analysis Software (RTA). Data were converted to FASTQ files

136 using the bcl2fastq2 v1.8.4 software (Illumina Inc.). The reads were trimmed for quality. Those

137 were then aligned to the human reference genome (hg38 gencode for humans) using HiSAT2

138 [15]. The alignments were processed using Ballgown (free online software) [15], and per-gene

139 counts were obtained. The raw counts were processed using DESeq2 and R.

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

7

140 Gene network and pathway analysis

141 Ingenuity Pathway Analysis (IPA) Ingenuity Systems QIAGEN, Content version: 45865156,

142 2018, Redwood City, CA, USA) was used to carry out pathway analysis for differentially

143 expressed genes across samples. Each gene symbol was mapped to its corresponding gene

144 object in the Ingenuity Pathways Knowledge Base. The over-represented pathways are ranked

145 according to the calculated Benjamini-Hochberg FDR P<0.05 [16].

146 qRT-PCR on laser captured primordial follicle oocytes

147 RNA from single primordial follicles was amplified, and cDNA was prepared using established

148 protocol [14], and gene expression analysis was done by qRT-PCR using Syber Green (Applied

149 Biosystems Warrington, UK) on ABI QuantStudio-6 Flex Real-Time PCR machine. PCR

150 cycling conditions were 95°C for 5 min and 45 cycles of 10 s at 95°C, 58°C for 15 s, and 72°C

151 for 15 s, followed by a melting curve analysis to confirm the single, specific products. The

152 expression level of each mRNA was calculated by the comparative Ct method (ΔΔCt), and the

153 fold change (FC) was calculated by the equation 2−ΔΔCt. β-Actin was used as a reference gene

154 for normalization. Primers for qRT-PCR reactions are specified in Supplementary Table 1 and

155 were obtained from UDT (USA). 'n' indicates the number of single follicles. Each experiment

156 contained primordial follicles from at least 3 different tissue donors.

157 Statistics

158 For RNA-seq analysis, we determined statistical significance for differential gene expression

159 based on the q-value (or the False Discovery Rate), with stringent cut-offs of 0.05 or less. This

160 ensures that the Type II error is kept as low as possible, given the constraints of sample size.

161 Our observations of gene transcript abundances by RNA-seq from a relatively low sample-size

162 are justified as we observe similar trends while interrogating the samples using other more direct

163 biological and biochemical approaches such as qRT-PCR analysis and immunohistochemistry.

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

8

164 Other data from chemotherapy- and vehicle-treated groups were compared using Student's two-

165 tailed t-test. P values less than 0.05 were considered significant.

166 RESULTS

167 Cyclophosphamide treatment does not increase primordial follicle growth initiation; it

168 induces DNA DSBs and apoptotic death

169 First, to morphologically determine if cyclophosphamide exposure increases the entry of

170 primordial follicles into the growth pool, we calculated the ratio of primary to primordial

171 follicles (py/pd = follicle growth initiation index) in xenografts [17]. Based on the growth

172 initiation ratio, we are only able to give an estimate, and not a definite answer of the follicle

173 fate since we use only a small fragment of the cortex and of the unequal distribution of the

174 follicles in each tissue. For H&E and AC-3 staining, we analyzed a total of 70 follicles (51

175 primordial and 19 primaries) from 3 slides of 8 different tissue pieces for the vehicle group and

176 202 follicles (177 primordial and 25 primaries) from 3 slides of 10 pieces of cortex from the

177 chemo-treated group. We found no significant difference in the py/pd ratio between the vehicle

178 and the cyclophosphamide -treated group (34.62 ± 7.94, vs. 19.97 ± 6.79; p = 0.17) (Figure 1A).

179 Because atretic follicles may not initiate growth, we also analyzed the ratio of non-atretic py/pd

180 follicles. We, again, did not observe a significant difference in follicle growth initiation rate

181 between the vehicle- and cyclophosphamide -treated groups (34.59 ± 9.75 vs. 18.58 ± 10.21; p

182 = 0.08) (Figure 1B).

183 Next, we studied whether cyclophosphamide induces apoptotic oocyte death using AC-3

184 immunostaining. We found a significant increase in the percentage of apoptotic primordial

185 (65.03% ± 3.55% vs. 28.27% ± 5.77%; p < 0.0001 cyclophosphamide vs vehicle) and primary

186 follicles (63.67% ± 9.89%, n = 8 vs. 28.57% ± 8.74%; n = 10; p = 0.01 cyclophosphamide vs

187 vehicle) indicating that cyclophosphamide caused massive apoptotic follicle death within 12h

188 of treatment (Figure 1C-F). Moreover, confirming earlier reports, we found by γH2AX staining

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

9

189 that chemotherapy induced DNA DSBs in primordial follicles compared to the vehicle

190 treatment (82 % ± 3.1%, n = 5 vs. 23.82% ± 8.66%, n = 5; p = 0.0002).‘n’ equals the number

191 of xenografted tissue (Figure 1G-H).

192 Individual-oocyte RNA-Seq Analysis of Pathways Activated in Primordial Follicles in

193 Acute Response to Chemotherapy

194 We then performed high throughput RNA-seq on non-atretic, individual primordial follicle

195 oocytes from cyclophosphamide - and vehicle-treated xenografts to have a broader view of the

196 pathways that are altered in primordial follicles in acute response to chemotherapy. For each

197 condition, we used two individual primordial follicles from each of the two different donor

198 ovaries. On average, 29.8 mln reads were obtained per sample with 74% alignment to the human

199 genome. We found that 190 genes were differentially expressed between the chemo- and

200 vehicle-treated groups (fold change ≥ 2, p < 0.05) (Figure 2A and Supplementary Table 2). We

201 then performed IPA enrichment analysis (Qiagen) to determine significantly altered (Figure 2B)

202 canonical pathways (p = 0.05; fold change [fc] = 2). We found that with chemo-exposure, the

203 Fcγ Receptor-mediated Phagocytosis in Macrophages and Monocytes (p = 0.005),

204 Phospholipase C Signaling (p = 0.005), Ephrin Receptor Signaling (p = 0.005) and Interleukin-

205 8 signaling (p = 0.03) were suppressed. We also found significant changes in Axonal Guidance

206 Signaling (p = 0.0003), Gap junction signaling (p = 0.01), and Focal Adhesion Kinase (FAK)

207 Signaling (p = 0.04), but no predictions could be made as to the direction of change. The

208 Phospholipase C Signaling, Ephrin Receptor Signaling, and IL8 signaling pathways are known

209 to be associated with inhibition of apoptosis. We used IPA to generate the network connecting

210 the genes present in these pathways with the apoptosis function (p = 5.37E-19) and to predict if

211 apoptosis is favored based on the expression status of these genes in our data. The IPA

212 suggested activation of apoptotic processes in the chemotherapy-treated samples

213 (Supplementary Figure1). Further, we used IPA to find the overlap between the differentially

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

10

214 expressed genes after chemotherapy exposure and those genes in the IPA knowledge base

215 associated with the functions of apoptosis and activation of primordial follicles. We found in

216 our data set that PTEN, Akt-1 Foxo3, BAD, ANGPT1, Pde and BTG2 overlapped with apoptotic

217 function (p = 3.58E-18), while PTEN and KITLG overlapped with the activation function of

218 ovarian follicles (p = 7.90E-07). The analysis of our dataset showed that the growth activation

219 of the primordial follicle is inhibited while apoptosis is triggered in acute response to

220 chemotherapy (Figure 2C).

221 Supporting these findings, RNA-seq analysis showed that cyclophosphamide did not cause

222 transcriptomic activation PI3K/PTEN/Akt pathway genes, which are responsible for primordial

223 follicle growth initiation. Specifically, RNA-seq analysis showed a significant decrease in the

224 expressions of PECAM1 (p = 2.13E-09), IKBKE (p = 0.001) and ANGPT1 (p = 0.03) (Figure

225 3A-C) in the cyclophosphamide -treated samples when compared to the vehicle. These genes

226 have anti-apoptotic functions and are also known activators of Akt [18][19][20][21][22].

227 Further analysis of the PI3K/PTEN/Akt pathway in the cyclophosphamide -treated samples

228 showed a trend toward a decline in Akt-1 (p = 0.19) and no significant change in the expressions

229 of Foxo3a (p = 0.8) and rpS6 (p = 0.36) (Figure 3D-F). These findings suggest that

230 cyclophosphamide treatment does not result in the activation of follicle growth or anti-apoptotic

231 pathways but triggers an acute apoptotic response.

232 qRT-PCR and Immunohistochemical Confirmation of Chemotherapy-Induced Acute

233 Pathway Changes in Human Primordial Follicles

234 To further explore and confirm the expression patterns of the PI3K/PTEN/Akt pathway

235 members in primordial follicle oocytes in acute response to chemotherapy, we performed qRT-

236 PCR analysis on LCM, individual primordial follicle oocytes. Based on the relative gene

237 expression analysis, we found no change in the expressions of Akt-1 (n vehicle = 5 and n chemo =

238 6; p = 0.93), rpS6 (n vehicle = 5 and n chemo = 6; p = 0.32) and Foxo3a expression (n vehicle = 5 and

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

11

239 n chemo = 6; p = 0.12) 12h after cyclophosphamide exposure compared to the vehicle (Figure

240 4A). To further confirm our findings, we investigated the changes in the immunohistochemical

241 expression of the key PI3K/PTEN/Akt pathway proteins in response to chemotherapy (Figure

242 4B). We calculated the percentage of positive to the total number of follicles from 3 slides per

243 xenografted tissue, 'n' indicating the number of tissue pieces. We observed no significant

244 differences between the vehicle and cyclophosphamide treatment groups in the expressions of

245 p(phospho)-Akt (7.29 ± 0.22, n = 8 vs. 11.59 ± 0.8, n = 6; p = 0.52), p-Foxo3a (12.03 ± 1.48, n

246 = 9 vs. 18.85 ± 1.43, n = 8 ; p = 0.63), and p-rpS6 (6.06 ± 0.14, n = 11 vs.12.55 ± 0.94, n = 7;

247 p = 0.4). These results further confirm that chemotherapy does not activate the PI3K/PTEN/Akt

248 pathway in primordial follicles within 12h of exposure.

249 Chemotherapy Activates Pro-apoptotic BAD-Bcl2 Action

250 Bcl2 (B-cell 2) is a known anti-apoptotic gene [23]. It is localized to the outer

251 membrane of the mitochondria, and it plays an important role in promoting cell survival via

252 inhibiting the actions of pro-apoptotic genes such as the BAD. By qRT-PCR analysis of laser-

253 captured primordial follicle oocyte we observed a trend towards declining expression of Bcl2

254 (n vehicle = 5 and n chemo = 6; p = 0.17), but together with BAD (n vehicle = 5 and n chemo = 6; p =

255 0.93), it did not reach statistical significance in the cyclophosphamide -treated xenografts vs.

256 the vehicles (Figure 5A-B). Under certain stress conditions, BAD is activated by

257 dephosphorylation and dimerizes with BCL2, preventing it from its anti-apoptotic function, and

258 promote apoptosis. We showed a significantly increased colocalization of BAD and BCL2

259 protein staining in cyclophosphamide -treated samples (n = 6) compared with the vehicle (n =

260 7; p = 0.004) (Figure 5C, D-J), which may suggest increased protein colocalization and potential

261 interactions. These results collectively indicate that within 12h, chemotherapy exposure results

262 in increased liability to apoptotic death in primordial follicle oocytes, possibly via the BAD-

263 Bcl2 pathway.

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

12

264 DISCUSSION

265 Maintenance of primordial follicle reserve is crucial for the reproductive life span of a female

266 individual. This reserve incurs a massive loss when a patient undergoes chemotherapy treatment,

267 later presenting itself as early menopause and infertility. In this study, we, for the first time,

268 identified the molecular mechanisms of chemotherapy-induced acute damage to ovarian

269 primordial follicles using our unique in vivo human ovarian xenograft model and single-cell

270 transcriptomic approaches. Because our earlier work showed that human primordial follicle

271 apoptosis peaks 12 hours post-exposure to chemotherapy [5], and as our focus was to determine

272 the mechanisms of acute follicle loss after cancer treatments, we limited our experiments to 12h

273 time point. We provided multiple lines of evidence converging that cyclophosphamide induces

274 primordial follicle death by prompt activation of apoptotic death pathways, not the

275 PI3K/PTEN/Akt pathway. While we showed that within 12h of treatment, chemotherapy

276 induces DNA DSBs in human oocytes, we found no evidence that it results in increased growth

277 activation of primordial follicles. Likewise, our previous immunohistochemical analyses in

278 rodents and human ovarian xenograft models showed that both cyclophosphamide and

279 doxorubicin induces DSBs and apoptotic oocyte death in primordial follicles [3].

280 In this unique in vivo human ovarian xenograft model, we first showed that chemotherapy

281 exposure did not acutely result in the change of total and non-atretic primary vs. primordial

282 follicle ratio rather, it induced DNA DSBs and AC-3 expression in primordial follicles. This

283 morphological evidence supports that chemotherapy induces primordial follicle apoptotic death

284 rather than growth initiation (Supplementary Figure 2).

285 The morphological data was then backed by LCM-based individual-oocyte RNA-seq and IPA

286 analysis, which we used to interrogate the key pathways that are activated in primordial follicles

287 in response to chemotherapy. The LCM-based cell isolation method enables us to precisely

288 capture individual primordial follicle oocytes from ovarian tissue where they are identified

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

13

289 based on their morphology without the need of unique genetic markers. This initial screening

290 directed us towards pathways related to apoptotic processes rather than those that are involved

291 in follicle growth activation as culprits in acute chemotherapy-induced ovarian reserve loss.

292 Considering that the number of the sequenced primordial oocyte follicles was relatively small,

293 we then confirmed these findings by qRT-PCR and immunohistochemistry in a larger number

294 of samples (n=5-6 and n=8-11, where 'n' is the number of single primordial follicle oocytes and

295 number of xenografted ovarian tissues belonging to different donors, respectively).

296 The transcriptomic analysis with our LCM-based individual-oocyte RNA-sequencing approach

297 indicated suppression of several signaling pathways such as Phospholipase-C (PLC), Ephrin

298 (Eph) Receptor, and IL-8 that are anti-apoptotic and involved with Bad-Bcl2-mediated

299 apoptosis [24][25][26]. Activation of IKBKE induces NF-κB nuclear accumulation and DNA-

300 binding activity by phosphorylation, which then increases the transcription of BCL2 [27].

301 Although we did not find a significant decrease in the expression of pro-survival gene BCL-2

302 and pro-apoptotic BAD, we observed an increased colocalization of BAD with BCL2, which

303 indicates an increased tendency for apoptotic death in human primordial follicle oocytes. We,

304 on the other hand, did not find an increase in the phosphorylation of the key PI3K/PTEN/Akt

305 members in acute response to chemotherapy. These converging findings support the common

306 theme that chemotherapy induces human primordial follicle death via apoptotic pathways rather

307 than massive follicle growth activation and resulting ‘burn-out’ in the acute phase. While our

308 data cannot rule out the activation of the primordial follicles at the later time point, it confirms

309 apoptosis as the acute response of the oocyte to chemotherapy.

310 In agreement with our findings in humans, other studies in rodents showed that alkylating agents

311 and cyclophosphamide induce apoptosis and deplete primordial follicle reserve [8,28–

312 31]. Studies in mouse oocytes also showed that cyclophosphamide induces a reduction in

313 mitochondrial transmembrane potential and accumulation of cytochrome-c in the cytosol,

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

14

314 leading to activation of the caspase family and apoptosis [32]. A plethora of factors such as

315 DNA damage, energy stress, loss of growth factor signaling and hypoxia can trigger apoptosis

316 by activation of BCl2 pathway proteins [33]. In our previous in vitro and xenografting studies,

317 we showed that a topoisomerase inhibitor, cancer doxorubicin induces DNA DSBs and

318 apoptosis in primordial follicles [3]. In this study, cyclophosphamide treatment likewise

319 increased DNA DSBs, as shown by increased expression of γH2AX staining in the primordial

320 follicles. Increased DNA damage in primordial follicles is a likely stress signal for the

321 recruitment of the pro-apoptotic BCL2 that causes the activation of the apoptotic cascade in the

322 primordial follicles.

323 Our individual-oocyte transcriptomic analyses also showed significant downregulation of

324 platelet endothelial cell adhesion molecule-1 (PECAM), and Angiopoietin-1(ANGPT1) (Figure

325 3D-F). Previous studies have shown that Jurkat cells with a 70% reduction of PECAM-1

326 expression by siRNA-silencing were significantly more sensitive to chemotherapy-induced

327 apoptosis [34]. Neuronal cell death was caused by the downregulation of Ang-1 (a.k.a.

328 ANGPT1, followed by Akt pathway inhibition [35]. It is also shown that Ang-1 induces

329 phosphorylation of Akt, which is associated with the up-regulation of the apoptosis inhibitor

330 survivin [22]. Inhibition of ANGPT1 in rat follicles increased the levels of AC-3 and decreased

331 Akt phosphorylation [35]. The latter concurs with our finding of increased AC-3 expression

332 after chemo exposure in primordial follicles and brings up the interesting possibility that chemo

333 exposure may indeed result in the suppression of Akt functions, which further favors apoptosis.

334 Interestingly, in addition to PECAM and ANGPT1, IKBKE is also a positive regulator of Akt

335 activation [19][21][22], which we found to have decreased in expression in primordial follicles

336 after chemotherapy.

337

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

15

338 In summary, our in vivo human data show that by suppression of anti-apoptotic mechanisms,

339 gonadotoxic chemotherapy exposure induces a pro-apoptotic state in primordial follicles in the

340 acute phase (Figure 6). Rather than PI3K/PTEN/Akt pathway activation resulting in massive

341 follicle growth and ‘burn-out’, we found contrary evidence that chemotherapy may favor

342 inactivation of the pathway. Nonetheless, our study only focused on the acute effects of

343 chemotherapy. Additional follicle loss may probably occur at later time points as a result of

344 additional mechanisms such as the stromal/microvascular damage and follicle activation.

345 However, given the magnitude of acute damage from DNA damage and apoptotic death, we

346 propose that the research for the development of targeted medical gonadal fertility preservation

347 treatments should focus on preventing DNA damage and/or chemo-induced pro-apoptotic state

348 in primordial follicles.

349 REFERENCES

350 [1] Cancer facts and Figures 2019. 1975.

351 [2] Oktay K, Harvey BE, Partridge AH, Quinn GP, Reinecke J, Taylor HS, Wallace WH,

352 Wang ET, Loren AW. Fertility Preservation in Patients With Cancer: ASCO Clinical

353 Practice Guideline Update. J Clin Oncol 2018; 36:1994–2001.

354 [3] Soleimani R, Heytens E, Darzynkiewicz Z, Oktay K. Mechanisms of chemotherapy-

355 induced human ovarian aging: double strand DNA breaks and microvascular

356 compromise. Aging (Albany NY) 2011; 3:782–793.

357 [4] Li F, Turan V, Lierman S, Cuvelier C, De Sutter P, Oktay K. Sphingosine-1-phosphate

358 prevents chemotherapy-induced human primordial follicle death. Hum Reprod 2014;

359 29:107–113.

360 [5] Oktem O, Oktay K. A novel ovarian xenografting model to characterize the impact of

361 chemotherapy agents on human primordial follicle reserve. Cancer Res 2007;

362 67:10159–10162.

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

16

363 [6] Meirow D, Biederman H, Anderson RA, Wallace WHB. Toxicity of chemotherapy and

364 radiation on female reproduction. Clin Obstet Gynecol 2010; 53:727–739.

365 [7] Kalich-Philosoph L, Roness H, Carmely A, Fishel-Bartal M, Ligumsky H, Paglin S,

366 Wolf I, Kanety H, Sredni B, Meirow D. Cyclophosphamide triggers follicle activation

367 and "burnout "; AS101 prevents follicle loss and preserves fertility. Sci Transl Med

368 2013; 5:185ra62.

369 [8] Szymanska KJ, Tan X, Oktay K. Unraveling the mechanisms of chemotherapy-induced

370 damage to human primordial follicle reserve: road to developing therapeutics for

371 fertility preservation and reversing ovarian aging. Mol Hum Reprod 2020.

372 [9] Oktay K, Bedoschi G, Pacheco F, Turan V, Emirdar V. First pregnancies, live birth,

373 and in vitro fertilization outcomes after transplantation of frozen-banked ovarian tissue

374 with a human extracellular matrix scaffold using robot-assisted minimally invasive

375 surgery. Am J Obstet Gynecol 2016; 214:94.e1-94.e9.

376 [10] Oktay K, Karlikaya G. Ovarian Function after Transplantation of Frozen, Banked

377 Autologous Ovarian Tissue. N Engl J Med 2000; 342:1919–1919.

378 [11] Oktay K, Schenken RS, Nelson JF. Proliferating cell nuclear antigen marks the

379 initiation of follicular growth in the rat. Biol Reprod 1995; 53:295–301.

380 [12] Kitajima M, Dolmans MM, Donnez O, Masuzaki H, Soares M, Donnez J. Enhanced

381 follicular recruitment and atresia in cortex derived from ovaries with endometriomas.

382 Fertil Steril 2014; 101:1031–1037.

383 [13] Gougeon A, Chainy GBN. Morphometric studies of small follicles in ovaries of women

384 at different ages. J Reprod Fertil 1987; 81:433–442.

385 [14] Picelli S, Faridani OR, Björklund ÅK, Winberg G, Sagasser S, Sandberg R. Full-length

386 RNA-seq from single cells using Smart-seq2. Nat Protoc 2014; 9:171–181.

387 [15] Pertea M, Kim D, Pertea GM, Leek JT, Salzberg SL. Transcript-level expression

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

17

388 analysis of RNA-seq experiments with HISAT, StringTie and Ballgown. Nat Protoc

389 2016; 11:1650–1667.

390 [16] Yoav Benjamini Yosef Hochberg. Controlling the False Discovery Rate: A Practical

391 and Powerful Approach to Multiple Testing. J R Stat Soc Ser B ( Methodol 1995;

392 57:289–300.

393 [17] Oktay K, Karlikaya G, Akman O, Ojakian GK, Oktay M. Interaction of Extracellular

394 Matrix and Activin-A in the Initiation of Follicle Growth in the Mouse Ovary1. Biol

395 Reprod 2000; 63:457–461.

396 [18] Limaye V, Li X, Hahn C, Xia P, Berndt MC, Vadas MA, Gamble JR. Sphingosine

397 kinase-1 enhances endothelial cell survival through a PECAM-1-dependent activation

398 of PI-3K/Akt and regulation of Bcl-2 family members. Blood 2005; 105:3169–3177.

399 [19] Fleming I, Fisslthaler B, Dixit M, Busse R. Role of PECAM-1 in the shear-stress-

400 induced activation of Akt and the endothelial nitric oxide synthase (eNOS) in

401 endothelial cells. J Cell Sci 2005; 118:4103–4111.

402 [20] Xie X, Zhang D, Zhao B, Lu M-K, You M, Condorelli G, Wang C-Y, Guan K-L.

403 IkappaB kinase epsilon and TANK-binding kinase 1 activate AKT by direct

404 phosphorylation. Proc Natl Acad Sci U S A 2011; 108:6474–9.

405 [21] Mahajan K, Mahajan NP. PI3K-independent AKT activation in cancers: a treasure

406 trove for novel therapeutics. J Cell Physiol 2012; 227:3178–84.

407 [22] Papapetropoulos A, Fulton D, Mahboubi K, Kalb RG, O’Connor DS, Li F, Altieri DC,

408 Sessa WC. Angiopoietin-1 inhibits endothelial cell apoptosis via the Akt/survivin

409 pathway. J Biol Chem 2000; 275:9102–5.

410 [23] Campbell KJ, Tait SWG. Targeting BCL-2 regulated apoptosis in cancer. Open Biol

411 2018; 8.

412 [24] Liu J, Li M, Zeng W-S, Bai X-C, Luo S-Q. [Inhibition of phospholipase C gamma1

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

18

413 signaling pathway promotes apoptosis of human colorectal carcinoma cells]. Di Yi Jun

414 Yi Da Xue Xue Bao 2005; 25:177–80.

415 [25] Maddigan A, Truitt L, Arsenault R, Freywald T, Allonby O, Dean J, Narendran A,

416 Xiang J, Weng A, Napper S, Freywald A. EphB Receptors Trigger Akt Activation and

417 Suppress Fas Receptor-Induced Apoptosis in Malignant T Lymphocytes. J Immunol

418 2011; 187:5983–5994.

419 [26] Stronach EA, Cunnea P, Turner C, Guney T, Aiyappa R, Jeyapalan S, de Sousa CH,

420 Browne A, Magdy N, Studd JB, Sriraksa R, Gabra H, et al. The role of interleukin-8

421 (IL-8) and IL-8 receptors in platinum response in high grade serous ovarian carcinoma.

422 Oncotarget 2015; 6:31593–31603.

423 [27] Heckman CA, Mehew JW, Boxer LM. NF-κB activates Bcl-2 expression in t(14;18)

424 lymphoma cells. Oncogene 2002; 21:3898–3908.

425 [28] Wandji S-A, Sršeň V, Voss AK, Eppig JJ, Fortune JE. Initiation in Vitro of Growth of

426 Bovine Primordial Follicles1. Biol Reprod 1996; 55:942–948.

427 [29] Luo Q, Yin N, Zhang L, Yuan W, Zhao W, Luan X, Zhang H. Role of SDF-1/CXCR4

428 and cytokines in the development of ovary injury in chemotherapy drug induced

429 premature ovarian failure mice. Life Sci 2017; 179:103–109.

430 [30] Luan Y, Edmonds ME, Woodruff TK, Kim SY. Inhibitors of apoptosis protect the

431 ovarian reserve from cyclophosphamide. J Endocrinol 2019; 240:243–256.

432 [31] Nguyen QN, Zerafa N, Liew SH, Findlay JK, Hickey M, Hutt KJ. - and

433 cyclophosphamide-induced primordial follicle depletion is caused by direct damage to

434 oocytes. MHR Basic Sci Reprod Med 2019:1–12.

435 [32] Perez GI, Knudson CM, Leykin L, Korsmeyer SJ, Tilly JL. Apoptosis-associated

436 signaling pathways are required for chemotherapy-mediated female germ cell

437 destruction. Nat Med 1997; 3:1228–1232.

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

19

438 [33] Hata AN, Engelman JA, Faber AC. The BCL2 family: Key mediators of the apoptotic

439 response to targeted anticancer therapeutics. Cancer Discov 2015; 5:475–487.

440 [34] Gao C, Sun W, Christofidou-Solomidou M, Sawada M, Newman DK, Bergom C,

441 Albelda SM, Matsuyama S, Newman PJ. PECAM-1 functions as a specific and potent

442 inhibitor of mitochondrial-dependent apoptosis. Blood 2003; 102:169–179.

443 [35] Parborell F, Abramovich D, Irusta G, Tesone M. Angiopoietin 1 reduces rat follicular

444 atresia mediated by apoptosis through the PI3K/Akt pathway. Mol Cell Endocrinol

445 2011; 343:79–87.

446 [36] A Vogel CF, Garcia J, Wu D, Mitchell DC, Zhang Y, Kado NY, Wong P, Trujillo DA,

447 Lollies A, Bennet D, Schenker MB, Mitloehner FM. Activation of inflammatory

448 responses in human U937 macrophages by particulate matter collected from dairy

449 farms: An in vitro expression analysis of pro-inflammatory markers. Environ Heal A

450 Glob Access Sci Source 2012; 11:17.

451 [37] Nakatani K, Thompson DA, Barthel A, Sakaue H, Liu W, Weigel RJ, Roth RA. Up-

452 regulation of Akt3 in estrogen receptor-deficient breast cancers and androgen-

453 independent lines. J Biol Chem 1999; 274:21528–21532.

454 [38] W.H. G, M.G. L, S.A. S, I.M. J, D.H. M, S.H. D. Activation of the mTOR pathway by

455 low levels of xenoestrogens in breast epithelial cells from high-risk women.

456 Carcinogenesis 2011; 32:1724–1733.

457 [39] Hui RC-Y, Gomes AR, Constantinidou D, Costa JR, Karadedou CT, Fernandez de

458 Mattos S, Wymann MP, Brosens JJ, Schulze A, Lam EW-F. The Forkhead

459 Transcription Factor FOXO3a Increases Phosphoinositide-3 Kinase/Akt Activity in

460 Drug-Resistant Leukemic Cells through Induction of PIK3CA Expression. Mol Cell

461 Biol 2008; 28:5886–5898.

462 [40] Liang J, Cao R, Wang X, Zhang Y, Wang P, Gao H, Li C, Yang F, Zeng R, Wei P, Li

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

20

463 D, Li W, et al. Mitochondrial PKM2 regulates oxidative stress-induced apoptosis by

464 stabilizing Bcl2. Cell Res 2017; 27:329–351.

465 [41] Borhani N, Manoochehri M, Saleh Gargari S, Ghaffari Novin M, Mansouri A, Omrani

466 MD. Decreased expression of proapoptotic genes caspase-8- and BCL2-associated

467 agonist of cell death (BAD) in ovarian cancer. Clin Ovarian Other Gynecol Cancer

468 2014; 7:18–23.

469 AUTHOR CONTRIBUTIONS

470 K.O. designed the study and conceived the idea, S.T, B.M, V.T, E.T performed the experiments,

471 S.T and K.S analyzed the results, R.G. assisted with bioinformatic analysis, K.O, S.T., and K.S

472 wrote the manuscript

473 ADDITIONAL INFORMATION

474 Grant support: Research was supported by R01 HD053112

475 Competing Interests Statement: The authors declare no competing interests.

476

477

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

21

478 FIGURES

479

480 Figure 1. Cyclophosphamide does not trigger follicle growth initiation but induces DNA

481 DSBs and apoptosis in human primordial follicles 12h after exposure. There was no

482 significant difference observed in (A) total (p = 0.17), (B) non-atretic (p = 0.08) primordial

483 follicle growth initiation rate as calculated by primary to primordial (py/pd) follicle ratios

484 (nvehicle = 8, nchemo = 10). (C) Chemotherapy treatment induced significant DNA damage

485 (γH2AX positive +) in primordial follicles (p = 0.0001). (D) There was a significant increase in

486 the percentage of apoptotic primordial (p = 0.001) and primary (p = 0.01) follicles in

487 cyclophosphamide -treated samples (nvehicle = 8, nchemo = 10). Photomicrographs of γH2AX (E-

488 F) and AC-3 staining (G-H) positive follicles in vehicle- and cyclophosphamide -treated (chemo)

489 groups. Black arrows indicate negative, black arrowheads show follicles positive for AC-3 and

490 γH2AX. The 'n' equals the number of xenografted ovarian tissue, data are presented as mean ±

491 S.E.M.

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

22

492

493 Figure 2. Individual-oocyte RNA-Sequencing and Ingenuity Pathway Analysis (IPA)

494 Results. (A) Heat map of genes that are significantly altered 12h after cyclophosphamide

495 exposure compared to the vehicle treatment. (B) Major pathways that were found to be

496 significantly altered after chemotherapy exposure. The over-represented pathways are ranked

497 according to the calculated Benjamini-Hochberg FDR, p<0.05. (C) IPA was utilized to find the

498 overlap between the differentially expressed genes in our study and those genes in the IPA

499 knowledge base associated with the functions of apoptosis and activation of primordial follicles.

500 This analysis predicted relationships that favored apoptosis and inhibition of the Akt pathway

501 in primordial follicles 12h after chemotherapy exposure.

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

23

502

503 Figure 3. RNA-Sequencing results indicating lack of activation of the PI3K/PTEN/Akt

504 pathway 12h after chemotherapy exposure. Significant decrease in the expression of (A)

505 PECAM1 (p = 2.13E-09), (B) IKBKE (p = 0.001) and (C) ANGPT1 (p = 0.03) that are reported

506 to be anti-apoptotic with pro effects on the PI3K/PTEN/Akt pathway. In cyclophosphamide -

507 treated samples, there was a trend towards a decline in the expression of PI3K/PTEN/Akt

508 pathway member Akt-1 (p = 0.19) (D) while no changes in the expressions of the Foxo3a (p =

509 0.8) (E) or rpS6 (p = 0.36) (F) were observed.

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

24

510

511 Figure 4. Validation of individual primordial follicle oocyte RNA-seq results by qRT-PCR

512 and immunochemistry. (A) qRT-PCR analysis of PI3K/PTEN/Akt pathway genes shows no

513 significant change in the expression of Akt-1 (n vehicle = 5 and n chemo = 6; p = 0.93 ), rPS6 (n

514 vehicle = 5 and n chemo = 6; p = 0.32) or Foxo3a (n vehicle = 5 and n chemo = 6; p = 0.12) in the

515 cyclophosphamide-treated group (chemo) as compared to the vehicle. The ‘n’ indicates the

516 number of single primordial follicles analyzed, data are presented as mean of the ΔΔCt ± S.E.M.

517 (B) There is no change in the percentage of primordial follicles stained for (phospho)-Akt (7.29

518 ± 0.22, n = 8 vs. 11.59 ± 0.8, n = 6; p = 0.52), p-Foxo3a (12.03 ± 1.48, n = 9 vs. 18.85 ± 1.43,

519 n = 8 ; p = 0.63), and p-rpS6 (6.06 ± 0.14, n = 11 vs.12.55 ± 0.94, n = 7; p = 0.4) after

520 cyclophosphamide treatment. The ‘n’ indicates the number of xenografted ovarian cortex

521 tissues, data are presented as mean ± S.E.M.

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

25

522

523 Figure 5. qRT-PCR and immunostaining analysis of BAD and BCL2 12h after

524 cyclophosphamide exposure. qRT-PCR analysis shows no significant change in the

525 expression of both (A) Bcl-2 (n vehicle = 5 and n chemo = 6; p = 0.17) and (B) BAD (n vehicle = 5

526 and n chemo = 6; p = 0.93) after cyclophosphamide exposure (chemo) compared to the vehicle

527 group. The 'n' indicates the number of single primordial follicle oocytes analyzed, data are

528 presented as ΔΔCt mean of the ± S.E.M. (C) By Image J analysis, the BAD-BCL2

529 colocalization intensity was increased in primordial follicles after cyclophosphamide exposure

530 (n = 6-7; p = 0.004), 'n' indicates the number of different xenografted tissue pieces, data are

531 presented as mean ± S.E.M. (D-E) Immunofluorescence images representative of BAD-BCL2

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

26

532 staining in the vehicle- and chemo-treated samples. White arrowheads point to primordial

533 follicles. Inset (J) exemplifies colocalization (yellow color) of BAD and BCL2 in a primordial

534 follicle.

535

536 Figure 6. Mechanisms of acute chemotherapy-induced damage to ovarian reserve in

537 human. Chemotherapy exposure induces apoptotic death in primordial follicles by direct and

538 indirect mechanisms, and results in the massive depletion of human ovarian reserve. Directly,

539 gonadotoxic chemotherapy agents such as cyclophosphamide induce double-strand DNA

540 breaks in human primordial follicle oocytes which results in the activation of apoptotic cascades

541 and increased co-localization of BAD-BCL2. Indirectly, same chemotherapy exposure also

542 leads to suppression of anti-apoptotic pathways such as the Ephrin Receptor Signaling,

543 Phospholipase C Signaling, and Interleukin-8 Signaling. Likewise, indirect suppression of pro-

544 Akt pathways such as PECAM-1 (Platelet/Endothelial Cell Adhesion Molecule-1), IKBKE

545 (inhibitor of nuclear factor kappa-B kinase subunit epsilon) and ANGPT1 (Angiopoietin-1)

546 likely leads to the inhibition of the pro-survival functions of the Akt pathway. The sum of the

547 latter two indirect mechanisms is increased sensitivity to apoptotic, gonadotoxic agents. While

bioRxiv preprint doi: https://doi.org/10.1101/2020.06.23.167783; this version posted June 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

27

548 this scheme explains the mechanisms of acute primordial follicle losses after chemotherapy,

549 there could be other direct and indirect mechanisms responsible for delayed losses, which are

550 not studied here. For more detailed discussion and an expanded figure, please refer to

551 Szymanska et al.,2020

552 SUPPLEMENTARY DATA LEGENDS

553 Supplementary Figure 1. Detailed representation of altered genes in acute response to

554 chemotherapy exposure in human primordial follicles. IPA predicted activation of apoptotic

555 processes in the cyclophosphamide-treated samples as there is a decrease in the expression of

556 the genes regulating the phospholipase C, Ephrin and IL-8 signaling. A decrease in the

557 expression of the genes of these anti-apoptotic pathways predicted activation of apoptosis in

558 cyclophosphamides-treated primordial follicles.

559 Supplementary Figure 2. Summary of experimental design, methods and results.

560 Supplementary Table 1. qRT-PCR primers for sequencing validation.

561 Supplementary Table 2. Differentially expressed genes between the vehicle- and

562 cyclophosphamide -treated groups. Fold change ≥ 2, p<0.05.