notch signaling governs the recruitment of ovarian ... · research article adam10–notch signaling...

11
RESEARCH ARTICLE ADAM10Notch signaling governs the recruitment of ovarian pregranulosa cells and controls folliculogenesis in mice Lizhao Feng 1 , Yijing Wang 2 , Han Cai 1 , Guanghong Sun 1 , Wanbao Niu 1 , Qiliang Xin 1 , Xiaofang Tang 3 , Jiawei Zhang 1 , Chao Wang 1 , Hua Zhang 1, * and Guoliang Xia 1, * ABSTRACT Ovarian follicles are the basic functional units of female reproduction in the mammalian ovary. We show here that the protein a disintegrin and metalloproteinase domain 10 (ADAM10), a cell surface sheddase, plays an indispensable role in controlling primordial follicle formation by regulating the recruitment of follicle supporting cells in mice. We demonstrate that suppressing ADAM10 in vitro or deletion of Adam10 in vivo disrupts germline cyst breakdown and primordial follicle formation. Using a cell lineage tracing approach, we show that ADAM10 governs the recruitment of ovarian follicle cells by regulating the differentiation and proliferation of LGR5-positive follicle supporting progenitor cells. By detecting the development of FOXL2-positive pregranulosa cells, we found that inhibiting ADAM10 reduced the number of FOXL2-positive cells in perinatal ovaries. Furthermore, inhibiting ADAM10 suppressed the activation of Notch signaling, and blocking Notch signaling also disrupted the recruitment of follicle progenitor cells. Taken together, these results show that ADAM10Notch signaling in ovarian somatic cells governs the primordial follicle formation by controlling the development of ovarian pregranulosa cells. The proper recruitment of ovarian follicle supporting cells is essential for establishment of the ovarian reserve in mice. KEY WORDS: ADAM10, Pregranulosa cell recruitment, Notch signaling, FOXL2, LGR5, Primordial follicle formation INTRODUCTION As the basic functional unit of female reproduction, primordial follicles serve as a source of developing follicles and oocytes over the entire reproductive lifespan of most mammalian species. Meanwhile, the non-renewable pool of primordial follicles, which is established in the perinatal ovary, directly represents the fecundity of each female (Mandl and Zuckerman, 1951; Zhang et al., 2014). Although the formation of primordial follicles has been well studied at a histological level (Pepling, 2012), the inner molecular mechanisms that regulate this process are still unclear. In mice, most germ cells stop dividing, enter into meiosis and form special structures termed germline cysts in the ovary before 16.5 days post coitum (dpc) (Pepling and Spradling, 1998). At same time, progenitor cells of somatic supporting cells start being recruited from the ovarian surface (Maatouk et al., 2012; Mork et al., 2012). At 17.5 dpc, FOXL2-positive (FOXL2 + ) somatic supporting cells, also called pregranulosa cells, begin to invade into cysts and separate germ cells to form primordial follicles (Pepling et al., 2010). At 4 days post partum (dpp), almost all oocytes are surrounded by pregranulosa cells and have formed primordial follicles in the ovary. The origins of pregranulosa cells have been discussed for decades. Previous studies have shown that FOXL2 + cells surround oocytes as pregranulosa cells and form primordial follicles (Mork et al., 2012; Schmidt et al., 2004; Zheng et al., 2014a). Recently, it has been shown that Lgr5, a marker for tissue stem cells in many organs, is expressed in the cortical region of mouse ovary during perinatal stages (Ng et al., 2014; Rastetter et al., 2014). The specific expression pattern of Lgr5 and Foxl2 suggested that LGR5-positive (LGR5 + ) cells in the ovarian surface epithelium are one of the major origins of FOXL2 + pregranulosa cells that contribute to the formation of primordial follicles (Rastetter et al., 2014). However, the molecular mechanisms involved in how such cells are recruited and become differentiated remain unclear. More importantly, the relationship between the process of follicle somatic cell development and primordial follicle formation is totally unknown. The protein a disintegrin and metalloproteinase domain 10 (ADAM10) is a cell surface sheddase that is able to cleave the extracellular domains of membrane-bound proteins and release the soluble ectodomain in a process known as ectodomain shedding(Hayashida et al., 2010; Reiss and Saftig, 2009; Weber and Saftig, 2012). ADAM10 is crucial for the regulation of various physiological processes such as cell proliferation, differentiation and death (Gibb et al., 2011; Glomski et al., 2011; Guo et al., 2015; Jorissen et al., 2010; Tsai et al., 2014; Weber et al., 2011; Zhang et al., 2010). It is well known that the regulatory roles of ADAM10 are closely related to Notch signaling (Weber and Saftig, 2012). ADAM10 functions by releasing the Notch intracellular domain (NICD) from the plasma membrane, which then enters the nucleus to activate Notch target genes such as Hes1 and Hey2 (Borggrefe and Oswald, 2009). Both ADAM10 and Notch signaling regulate the proliferation and differentiation of LGR5 + stem (or progenitor) cells in various mouse organs, such as the intestine, stomach and cochlea (Bramhall et al., 2014; Demitrack et al., 2015; Tsai et al., 2014; VanDussen et al., 2012). More specifically, an increasing body of evidence has shown that Notch signaling is indispensable for the formation of ovarian primordial follicles (Trombly et al., 2009; Vanorny et al., 2014; Xu and Gridley, 2013). However, whether and how ADAM10 plays a role in the establishment of the primordial follicle pool has not been addressed, and the relationship between ADAM10 and Notch signaling in primordial follicle formation remains unknown. In the current study, we investigated the functional role of ADAM10 in regulating the formation of primordial follicles in the perinatal mouse ovary. We found that somatic-cell-expressed ADAM10 plays an indispensable role in the progression of Received 30 November 2015; Accepted 11 April 2016 1 State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China. 2 National Institute of Biological Sciences, Beijing 102206, China. 3 State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China. *Author for correspondence ([email protected]; [email protected]) H.Z., 0000-0002-4700-5971; G.X., 0000-0003-3674-0142 2202 © 2016. Published by The Company of Biologists Ltd | Journal of Cell Science (2016) 129, 2202-2212 doi:10.1242/jcs.184267 Journal of Cell Science

Upload: others

Post on 14-Oct-2020

5 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Notch signaling governs the recruitment of ovarian ... · RESEARCH ARTICLE ADAM10–Notch signaling governs the recruitment of ovarian pregranulosa cells and controls folliculogenesis

RESEARCH ARTICLE

ADAM10–Notch signaling governs the recruitment of ovarianpregranulosa cells and controls folliculogenesis in miceLizhao Feng1, Yijing Wang2, Han Cai1, Guanghong Sun1, Wanbao Niu1, Qiliang Xin1, Xiaofang Tang3,Jiawei Zhang1, Chao Wang1, Hua Zhang1,* and Guoliang Xia1,*

ABSTRACTOvarian follicles are the basic functional units of female reproductionin the mammalian ovary. We show here that the protein a disintegrinand metalloproteinase domain 10 (ADAM10), a cell surfacesheddase, plays an indispensable role in controlling primordialfollicle formation by regulating the recruitment of follicle supportingcells in mice. We demonstrate that suppressing ADAM10 in vitro ordeletion of Adam10 in vivo disrupts germline cyst breakdown andprimordial follicle formation. Using a cell lineage tracing approach,we show that ADAM10 governs the recruitment of ovarian follicle cellsby regulating the differentiation and proliferation of LGR5-positivefollicle supporting progenitor cells. By detecting the developmentof FOXL2-positive pregranulosa cells, we found that inhibitingADAM10 reduced the number of FOXL2-positive cells in perinatalovaries. Furthermore, inhibiting ADAM10 suppressed the activationof Notch signaling, and blocking Notch signaling also disrupted therecruitment of follicle progenitor cells. Taken together, these resultsshow that ADAM10–Notch signaling in ovarian somatic cells governsthe primordial follicle formation by controlling the development ofovarian pregranulosa cells. The proper recruitment of ovarian folliclesupporting cells is essential for establishment of the ovarian reservein mice.

KEY WORDS: ADAM10, Pregranulosa cell recruitment, Notchsignaling, FOXL2, LGR5, Primordial follicle formation

INTRODUCTIONAs the basic functional unit of female reproduction, primordialfollicles serve as a source of developing follicles and oocytes overthe entire reproductive lifespan of most mammalian species.Meanwhile, the non-renewable pool of primordial follicles, whichis established in the perinatal ovary, directly represents the fecundityof each female (Mandl and Zuckerman, 1951; Zhang et al., 2014).Although the formation of primordial follicles has been well studiedat a histological level (Pepling, 2012), the inner molecularmechanisms that regulate this process are still unclear.In mice, most germ cells stop dividing, enter into meiosis and form

special structures termed germline cysts in the ovary before 16.5 dayspost coitum (dpc) (Pepling and Spradling, 1998). At same time,progenitor cells of somatic supporting cells start being recruited fromthe ovarian surface (Maatouk et al., 2012; Mork et al., 2012). At

17.5 dpc, FOXL2-positive (FOXL2+) somatic supporting cells, alsocalled pregranulosa cells, begin to invade into cysts and separategerm cells to form primordial follicles (Pepling et al., 2010). At∼4 days post partum (dpp), almost all oocytes are surrounded bypregranulosa cells and have formed primordial follicles in the ovary.

The origins of pregranulosa cells have been discussed for decades.Previous studies have shown that FOXL2+ cells surround oocytes aspregranulosa cells and form primordial follicles (Mork et al., 2012;Schmidt et al., 2004; Zheng et al., 2014a). Recently, it has beenshown that Lgr5, a marker for tissue stem cells in many organs, isexpressed in the cortical region of mouse ovary during perinatalstages (Ng et al., 2014; Rastetter et al., 2014). The specific expressionpattern of Lgr5 and Foxl2 suggested that LGR5-positive (LGR5+)cells in the ovarian surface epithelium are one of the major origins ofFOXL2+ pregranulosa cells that contribute to the formation ofprimordial follicles (Rastetter et al., 2014). However, the molecularmechanisms involved in how such cells are recruited and becomedifferentiated remain unclear. More importantly, the relationshipbetween the process of follicle somatic cell development andprimordial follicle formation is totally unknown.

The protein a disintegrin and metalloproteinase domain 10(ADAM10) is a cell surface sheddase that is able to cleave theextracellular domains of membrane-bound proteins and release thesoluble ectodomain in a process known as ‘ectodomain shedding’(Hayashida et al., 2010; Reiss and Saftig, 2009; Weber andSaftig, 2012). ADAM10 is crucial for the regulation of variousphysiological processes such as cell proliferation, differentiationand death (Gibb et al., 2011; Glomski et al., 2011; Guo et al., 2015;Jorissen et al., 2010; Tsai et al., 2014; Weber et al., 2011; Zhanget al., 2010). It is well known that the regulatory roles of ADAM10are closely related to Notch signaling (Weber and Saftig, 2012).ADAM10 functions by releasing the Notch intracellular domain(NICD) from the plasma membrane, which then enters the nucleusto activate Notch target genes such as Hes1 and Hey2 (Borggrefeand Oswald, 2009). Both ADAM10 and Notch signaling regulatethe proliferation and differentiation of LGR5+ stem (or progenitor)cells in various mouse organs, such as the intestine, stomach andcochlea (Bramhall et al., 2014; Demitrack et al., 2015; Tsai et al.,2014; VanDussen et al., 2012). More specifically, an increasingbody of evidence has shown that Notch signaling is indispensablefor the formation of ovarian primordial follicles (Trombly et al.,2009; Vanorny et al., 2014; Xu and Gridley, 2013). However,whether and how ADAM10 plays a role in the establishment of theprimordial follicle pool has not been addressed, and the relationshipbetween ADAM10 and Notch signaling in primordial follicleformation remains unknown.

In the current study, we investigated the functional role ofADAM10 in regulating the formation of primordial follicles in theperinatal mouse ovary. We found that somatic-cell-expressedADAM10 plays an indispensable role in the progression ofReceived 30 November 2015; Accepted 11 April 2016

1State Key Laboratory of Agrobiotechnology, College of Biological Science, ChinaAgricultural University, Beijing 100193, China. 2National Institute of BiologicalSciences, Beijing 102206, China. 3State Key Laboratory of Reproductive Biology,Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.

*Author for correspondence ([email protected]; [email protected])

H.Z., 0000-0002-4700-5971; G.X., 0000-0003-3674-0142

2202

© 2016. Published by The Company of Biologists Ltd | Journal of Cell Science (2016) 129, 2202-2212 doi:10.1242/jcs.184267

Journal

ofCe

llScience

Page 2: Notch signaling governs the recruitment of ovarian ... · RESEARCH ARTICLE ADAM10–Notch signaling governs the recruitment of ovarian pregranulosa cells and controls folliculogenesis

germline cyst breakdown and primordial follicle formation in mice.This was achieved by regulating the recruitment of FOXL2+

pregranulosa cells from LGR5+ follicle supporting progenitor cellsthrough Notch signaling. Our findings provide evidence showingthat ADAM10–Notch signaling determines the establishment of theprimordial follicle pool by regulating the development of folliclesomatic cells in the mouse ovary.

RESULTSSuppressing ADAM10 disrupts germline cyst breakdown andprimordial follicle formationTo investigate the potential relationship between ADAM10 andprimordial follicle formation, we first measured the expression ofAdam10 mRNA by quantitative real-time reverse transcription PCR(qRT-PCR) in perinatal ovaries. As shown in Fig. 1A, mRNA levelsof Adam10 changed markedly from 16.5 dpc to 4 dpp as primordialfollicle formation progressed. From 16.5 dpc to 1 dpp, Adam10mRNA expression increased considerably with the breakdown ofgermline cysts; the highest level of Adam10 mRNAwas observed at1 dpp, which was 2.8-fold higher than that at 16.5 dpc. With theestablishment of the primordial follicle pool at 4 dpp, the expressionofAdam10mRNAdecreased to a level comparable to that at 16.5 dpc.To detect the localization of Adam10 in the perinatal ovary, weperformed in situ hybridization and found that Adam10 was mainlyexpressed in somatic cells, especially ovarian surface epithelium cells,in 1 dpp ovary (Fig. 1B, arrowheads). These results indicate thatsomatic-cell-expressed ADAM10 has the potential to be involved inthe regulation of primordial follicle formation in the mouse ovary.To determine the functional role of ADAM10 in primordial

follicle formation, we collected ovaries from 16.5 dpc wild-typemice and cultured them with or without GM6001, an inhibitor ofADAM10 (Lemjabbar and Basbaum, 2002; Saftig and Reiss, 2011;Zhang et al., 2011), for 7 days in vitro. After 7 days of culture,most oocytes (8188.0±1152.3 oocytes per ovary, 86.6±12.2%,mean±s.d.; Fig. 1C) in the control group were surrounded bypregranulosa cells and formed primordial follicles (Fig. 1D,arrowheads). By contrast, only 14.2±1.8% of oocytes weresurrounded by pregranulosa cells to form primordial follicles inGM6001-treated ovaries (1442.0±181.4 per ovary, P<0.001 versuscontrol; Fig. 1C); the majority of oocytes (8744.0±530.7 oocytes,85.8±5.2% per ovary, P<0.001 versus control; Fig. 1C) were stillincluded in cysts without forming primordial follicles (Fig. 1D,broken circle). An identical number of oocytes were observed incontrol (9452.0±999.5 oocytes per ovary; Fig. 1C) and GM6001-treated ovaries (10186.0±355.1 oocytes per ovary; Fig. 1C),indicating that the survival of oocytes was not affected byinhibiting ADAM10. In addition, we cultured ovaries withanother ADAM10 selective inhibitor GI254023X (Hoetteckeet al., 2010; Hundhausen et al., 2003; Lo Sardo et al., 2012). Theresults showed that 7 days of GI254023X treatment alsosignificantly decreased the number of follicles (1138.0±184.0 perovary for GI254023X versus 8188.0±1152.3 per ovary for control;Fig. S1B) and most of the oocytes (5944.0±711.4 per ovary forGI254023X versus 1264.0±273.0 per ovary for control; Fig. S1B)remained included in cysts (Fig. S1A). Taken together, these resultsdemonstrate that ADAM10 plays a crucial role in the formation ofprimordial follicles in the perinatal mouse ovary.

Suppressing ADAM10 inhibits the recruitment of Foxl2+

pregranulosa cells in perinatal ovariesTo find out how somatically expressed ADAM10 controls theformation of primordial follicles, we next assessed the functional

role of ADAM10 in regulating the recruitment of pregranulosa cells.We first investigated the in vivo behavior of FOXL2+ pregranulosacells in the perinatal ovary. Immunostaining confirmed a previousreport (Rastetter et al., 2014) describing the localization of FOXL2+

cells that first emerged in the medulla and then gradually emerged inthe cortical region of the ovary, from 16.5 dpc to 4 dpp, with theformation of primordial follicles (Fig. S2A).

We then cultured 16.5 dpc ovaries with GM6001 to examine therole of ADAM10 in recruiting pregranulosa cells. After 7 days ofculture, we found that 475.4±31.4 FOXL2+ cells per section (mean±s.d.) (Fig. 2B) were detected in the cortical region of control ovaries(Fig. 2A). By contrast, 7 days of GM6001 treatment significantlydecreased the number of FOXL2+ cells (221.2±16.8 cells persection; Fig. 2B; P<0.001) in the cortex of the ovary (Fig. 2A), andonly a few FOXL2+ cells were localized around germline cysts. Toconfirm the role of ADAM10 in recruiting pregranulosa cells, wealso measured the levels of Foxl2 mRNA after culturing in thepresence or absence of GM6001. Foxl2 mRNA expression wasfound to be significantly decreased after 3 or 7 days in culture in thepresence of GM6001 (Fig. 2C; P<0.001 for both). These resultsindicate that ADAM10 determines the recruitment of FOXL2+

pregranulosa cells to the cortex to regulate primordial follicleformation.

Suppressing ADAM10 inhibits ovarian LGR5+ progenitor celldifferentiationGiven that ADAM10 determines the number of FOXL2+

pregranulosa cells, we next examined how ADAM10 disrupts therecruitment of FOXL2+ cells to the ovarian cortex. Recent studieshave reported that FOXL2+ cells are derived from LGR5+ cells inthe ovarian surface epithelium (Ng et al., 2014; Rastetter et al.,2014). Therefore, we used Lgr5-EGFP-ires-CreERT2 (Lgr5-KI)reporter mice to trace the recruitment of LGR5+ cells, andinvestigated the developmental dynamics of LGR5+ cells andFOXL2+ cells in perinatal Lgr5-KI ovaries by co-staining with anti-FOXL2 antibody. LGR5 was located by staining with anti-EGFPantibody.

As shown in Fig. 3A, LGR5+ cells were not only observed in theovarian surface epithelium but also in the cortical region of ovariesat 16.5 dpc and 1 dpp, indicating that the recruitment of LGR5+

cells was activated from 16.5 dpc to 1 dpp, before primordialfollicle formation. At 4 dpp, no LGR5+ cells were found in theovarian cortex, indicating that the recruitment of follicle supportingcells had been terminated in vivo. By staining for FOXL2 in Lgr5-KImice, we found that the transformation of LGR5+ cells to FOXL2+

cells mainly occurred around 1 dpp (Fig. 3A; Fig. S2E) becausemany LGR5 and FOXL2 double-positive cells were observed in1 dpp ovaries (Fig. 3A, arrowheads; Fig. S2D, arrowheads). Bycounting the number of LGR5+ cells and FOXL2+ cells from16.5 dpc to 4 dpp, we assessed the dynamics of cell recruitment, andfound that the number of LGR5+ cells in the cortical regiongradually decreased (from 186.4±17.6 to 4.8±2.3 cells per section),and the transformation of FOXL2+ cells markedly increased (from89.2±19.8 to 571.2±55.3 cells per section) in vivo with primordialfollicle formation (mean±s.d.; Fig. 3B).

We next investigated whether ADAM10 controls the recruitmentand differentiation of LGR5+ cells in ovaries. Ovaries (16.5 dpc)from Lgr5-KI mice were collected and cultured in the presence orabsence of GM6001 for 3 or 7 days, respectively. To validate theculture system, the development of LGR5+ cells and FOXL2+ cellswas first observed in cultured ovaries (Fig. 3C), and similardynamics to that for in vivo developed ovaries follicle were found

2203

RESEARCH ARTICLE Journal of Cell Science (2016) 129, 2202-2212 doi:10.1242/jcs.184267

Journal

ofCe

llScience

Page 3: Notch signaling governs the recruitment of ovarian ... · RESEARCH ARTICLE ADAM10–Notch signaling governs the recruitment of ovarian pregranulosa cells and controls folliculogenesis

(compare Fig. 3C with Fig. 3A). After 7 days of in vitro culture withGM6001, however, LGR5+ cells were not only observed at theovarian surface but also in the cortical region of ovaries (Fig. 3E);the number of LGR5+ cells (105.4±3.6 cells per section; Fig. 3F)was considerably increased in the GM6001-treated group comparedto control (9.8±2.5 cells per section, Fig. 3D). The directcomparison shows that the change is significant (Fig. S2G). Afluorescence-activated cell sorting (FACS) analysis of Lgr5-KI

ovarian cells after 7 days of culture also showed that the proportionof LGR5+ cells was significantly increased in the GM6001-treatedgroup (27.1±1.2%; Fig. S3B) compared to control (14.9±1.5%;Fig. S3A,B). The higher number of LGR5+ cells indicates that eitherthe differentiation of LGR5+ cells is suppressed or the proliferationof LGR5+ cells is increased by GM6001 treatment.

Indeed, our counting results showed that the increase in FOXL2+

cells was significantly suppressed in the cortical region of ovaries by

Fig. 1. Suppressing ADAM10 disrupts germline cyst breakdown and primordial follicle formation. (A) The expression of Adam10mRNAwas assessed byqRT-PCR in perinatal ovaries, and a marked increase in Adam10mRNA expression was observed at 1 dpp. Different letters (a–c) indicate significant differencesbetween all groups (P<0.05, ANOVA and Holm–Sidak test). (B) In situ hybridization of Adam10 showed that this protein was mainly expressed in ovarian somaticcells at 1 dpp, especially in the ovarian surface epithelium (black arrowheads). Scale bar: 50 µm. (C) Although the total number of oocytes was not affected byGM6001, an inhibitor of ADAM10, primordial follicle formation was significantly suppressed and themajority of oocytes still existed in germline cysts after inhibitingADAM10 in ovaries. The graph shows the total numberof follicles, and the numberof oocytes that were in cysts (n=3), together with the total number of oocytes, perovary. ***P<0.001 (t-test), control versus GM6001-treated ovaries. Data are presented as means±s.d. (D) Ovaries at 16.5 dpc were cultured in the presence orabsence ofGM6001 for 7 days in vitro. Histological analysis showed that theADAM10 inhibitor disrupted germline cyst breakdownandprimordial follicle formation.Oocyteswere stained for DDX4 (green color). Nuclei were dyedwith a propidium iodide (PI) counterstain (red color). Yellowarrowheads point to primordial follicles.Yellow dashed lines circle unbroken germline cysts. Sections were re-stained with hematoxylin to show cellular morphology. Scale bars: 20 µm.

2204

RESEARCH ARTICLE Journal of Cell Science (2016) 129, 2202-2212 doi:10.1242/jcs.184267

Journal

ofCe

llScience

Page 4: Notch signaling governs the recruitment of ovarian ... · RESEARCH ARTICLE ADAM10–Notch signaling governs the recruitment of ovarian pregranulosa cells and controls folliculogenesis

GM6001 treatment (Fig. S2E). This indicated that the differentiationof LGR5+ cells was suppressed by GM6001 treatment. Moreover, bycounting the number of LGR5 and FOXL2 double-positive cellsafter 3 or 7 days in GM6001-treated ovaries, it was found that theprogression of LGR5+ cell differentiation was significantly retarded(Fig. 3G,H). In addition, the levels of Lgr5mRNAwere significantlyhigher after 3 or 7 days for the GM6001-treated group comparedto control (Fig. 3I), which is consistent with the results of cellcounting.

ADAM10 controls the recruitment of ovarian surface LGR5+

progenitorsOur previous results demonstrated that the transformation ofLGR5+ cells into FOXL2+ cells was suppressed when ADAM10was inhibited; hence, we next examined whether GM6001treatment increased the proliferation of LGR5+ cells. The ovariansurface cells were labeled by lentivirus-transfected EGFP, whichintegrated into genomic DNA, and the recruitment of LGR5+ cellswas traced in vitro with or without GM6001. The experimentalstrategy is shown in Fig. 4A. After 12 h of infection and 12 h oflentivirus-free culture, surface cells in infected ovaries weresuccessfully labeled by EGFP (white color) and no cells withinthe inner part of ovaries were labeled (Fig. 4B). The statistics ofEGFP+ cells indicated that there was no significant difference inthe infection efficiency of different ovaries (Fig. S2I). Infectedovaries were then cultured in the presence or absence of GM6001for 7 days and the behavior of labeled cells was visualized. Incontrol group, it was found that for untreated control cells, thepopulation of EGFP+ cells had obviously increased, with manyEGFP+ cells invading the cortex and surrounding oocytes to formprimordial follicles after culture (Fig. 4B, control). In sharp

contrast, with GM6001 treatment, EGFP+ cells were mainlyobserved at the ovarian surface and only a few labeled cells hadinvaded into the ovary (Fig. 4B, GM6001). This result indicatesthat the recruitment of ovarian surface cells is suppressed byinhibiting ADAM10. Moreover, the number of EGFP+ cells in theGM6001-treated group (39.7±5.7 cells per section; mean±s.d.;Fig. 4C) was significantly less than the number of cells in thecontrol group (110.0±9.0 cells per section; Fig. 4C; P<0.001),indicating that the proliferation of ovarian surface epithelium wasalso inhibited by GM6001. Given that ovarian surface epitheliumcells are LGR5+ (Fig. 3A), our results show that both therecruitment and proliferation of LGR5+ cells is suppressed byGM6001 treatment.

To confirm our findings, proliferating cells were labeled withBrdU in Lgr5-KI ovaries and the number of dividing LGR5+ cellson the ovarian surfacewasmeasured.We found that the proliferationof ovarian surface LGR5+ cells was significantly suppressedafter 3 days of GM6001 treatment (Fig. 4D, arrowheads; Fig. 4E,control at 22.6±3.4 cells compared to GM6001 at 10.4±1.7 persection). Thus, our results indicate that ADAM10 plays a key role inregulating the recruitment and proliferation of LGR5+ progenitorcells that contributes to primordial follicle formation.

ADAM10 regulates the recruitment and differentiation ofpregranulosa cells through Notch signalingA functional role forNotch signaling in controlling primordial follicleformation has recently been reported (Trombly et al., 2009; Vanornyet al., 2014; Xu and Gridley, 2013). As an upstream regulator ofNotch signaling, ADAM10might play a similar role in regulating theformation of primordial follicles according to our findings. Thus, wenext investigated the relationship between ADAM10 and Notch

Fig. 2. SuppressingADAM10 inhibits the recruitment of pregranulosa cells in perinatal ovaries. (A) Ovaries from 16.5 dpc wild-typemicewere collected andcultured in the presenceorabsenceofGM6001 for 7 days. After in vitro culture, the normal localizationof FOXL2+ cells (red color)was observed in control ovaries. Inthe GM6001 group, numerous oocytes were still in germline cysts. Only a few FOXL2+ cells were observed in the cortical region of ovaries (indicated by thedashed line). (B) Quantification of labeled FOXL2+ cells showed that the number of pregranulosa cells per section was significantly decreased in ovarian corticalregions of GM6001-treated groups compared to control groups (***P<0.001, n=5). Oocytes were labeled with anti-DDX4 antibody (green color), and nuclei stainedwith Hoechst 33342 counterstain (blue color). (C) Compared to the control group, the expression of Foxl2mRNAwas significantly reduced in ovaries after 3 days or7 days in vitro culture with GM6001 (n=6). ***P<0.001 (t-test), control versus GM6001-treated ovaries. Data are presented as means±s.d. Scale bars: 20 µm.

2205

RESEARCH ARTICLE Journal of Cell Science (2016) 129, 2202-2212 doi:10.1242/jcs.184267

Journal

ofCe

llScience

Page 5: Notch signaling governs the recruitment of ovarian ... · RESEARCH ARTICLE ADAM10–Notch signaling governs the recruitment of ovarian pregranulosa cells and controls folliculogenesis

signaling in the advancement of primordial follicle formation,especially the recruitment of follicle supporting cells.To examine whether Notch signaling plays a similar role in

regulating the differentiation and proliferation of LGR5+ cells as

ADAM10, 16.5 dpc ovaries from Lgr5-KI mice were collected andcultured in the presence of DAPT, a Notch signaling inhibitor. Asreported previously (Trombly et al., 2009), 7 days of DAPTtreatment disrupted germline cyst breakdown and primordial follicle

Fig. 3. Suppressing ADAM10 inhibits ovarian LGR5+ progenitor cell differentiation. (A) Using Lgr5-EGFP-ires-creERT2 (Lgr5-KI) reporter mice, the in vivodevelopmental dynamics of LGR5+ progenitor cells (green color) and FOXL2+ pregranulosa cells (red color) were traced in perinatal ovaries. The recruitment ofLGR5+ cells mainly occurred at 16.5 dpc and 1 dpp, and the transformation of FOXL2+ cells from LGR5+ cells was activated around 1 dpp (arrowheads, yellowcolor). At 4 dpp, no LGR5+ cells were observed in the cortical region of ovaries. (B) The developmental dynamics of LGR5+ cells and FOXL2+ cells showed that thenumber of LGR5+ cells gradually decreased and FOXL2+ cells increased with the development of perinatal ovaries. (C,D) Ovaries (16.5 dpc) were culturedin vitro, and a developmental pattern for LGR5+ and FOXL2+ cells was observed in cultured fetal ovaries that was comparable to that in in vivo developed ovaries(A,B). No LGR5+ cells were observed in the cortical region of ovaries after 7 days of in vitro culture. (E) The recruitment of LGR5+ cells was slightly increased after3 days culture in the presence of GM6001. A considerably increased number of LGR5+ cells was observed in the ovary after 7 days of GM6001 treatment, andmany LGR5+ cells were found in the ovarian cortical region. The ovarian cortical region is indicated by the dashed line. (F) Quantification of LGR5+ cells andFOXL2+ cells in ovaries cultured in the presence of GM6001. The decrease in LGR5+ cells and increase in FOXL2+ cells with time, as noted previously, wassuppressed in the GM6001-treated ovaries compared to the in vitro control (D) and in vivo (B). (G) LGR5 and FOXL2 double-positive cells were observed inovaries after 7 days of GM6001 treatment (arrowheads; the area indicated in E is shown). (H) Quantification of LGR5 and FOXL2 double-positive cells revealed amarked retardation of LGR5+ to FOXL2+ cell transformation in GM6001-treated ovaries after 3 days of culture. (I) A significantly high expression of Lgr5 mRNAwas found in GM6001-treated ovaries compared to control (n=6). Graphs in B, D, F and H show cell number per section (n=5). ***P<0.001 (t-test), control versustreated ovaries. Data are presented as means±s.d. Scale bars: 20 µm.

2206

RESEARCH ARTICLE Journal of Cell Science (2016) 129, 2202-2212 doi:10.1242/jcs.184267

Journal

ofCe

llScience

Page 6: Notch signaling governs the recruitment of ovarian ... · RESEARCH ARTICLE ADAM10–Notch signaling governs the recruitment of ovarian pregranulosa cells and controls folliculogenesis

formation (data not shown). For the development of folliclesupporting cells, we found that a comparable phenotype wasobserved with both DAPT and GM6001 treatments. Specifically,the differentiation of LGR5+ cells to FOXL2+ cells was markedlyblocked by 7 days of DAPT treatment (Fig. 5E). In addition, adecrease in FOXL2+ cell numbers (157.6±14.4 for DAPT versus475.4±14.4 cells per section for control; mean±s.d.; Fig. 5B,D) andan increase in LGR5+ cell numbers (133.0±10.2 per section inDAPT versus 9.8±2.5 cells per section in control; Fig. 5B,D) werenoted in the cortical regions of ovaries treated with DAPT for 7 days(Fig. 5A,C). The direct comparison shows that the change wassignificant (Fig. S3C,D). FACS analysis showed that the proportionof LGR5+ cells was significantly increased in the DAPT-treatedgroup (28.8±2.8%; Fig. S3B) compared to control after 7 days ofculture (14.9±1.5%; Fig. S3A,B). Gene expression patternsconfirmed our counting data that significantly decreased levels ofFoxl2 and increased expression of Lgr5were found in ovaries after 3or 7 days of DAPT treatment compared to control (Fig. S3E,F;

P<0.001 for all). Moreover, the proliferation of LGR5+ cells labeledwith BrdU was also significantly suppressed in the DAPT-treatmentgroup (Fig. 5F, arrowheads; 18.2±2.2 for DAPT versus 9.0±1.2cells per section for control; Fig. 5G; P<0.001). Lentivirus EGFPinfection experiments also showed that the recruitment of ovariansurface cells was significantly inhibited by DAPT treatment after7 days of culture (Fig. S3G and H; P<0.001). These results point to asimilar functional role for ADAM10 and Notch signaling incontrolling the recruitment of supporting cells of the ovarian folliclein mice.

To confirm the relationship between ADAM10 and Notchsignaling in primordial follicle formation, we measured gene orprotein expression patterns of Notch signaling in 16.5 dpc ovariesafter 3 days of GM6001 treatment. Gene expression analysisshowed no significant changes of Notch1, Notch2 and Jagged1mRNA levels (data not shown). However, the protein level ofNICD, the Notch intracellular domain, was decreased in ovaries ofthe GM6001-treated group (Fig. 5H) compared with control. In

Fig. 4. Suppressing ADAM10 inhibits ovarianLGR5+ progenitor cell recruitment andproliferation. (A) The strategy used to trace therecruitment of ovarian surface LGR5+

progenitors by lentivirus EGFP is outlined.Ovaries (15.5 dpc) were cultured with lentivirusEGFP for 12 h to label cells on the ovariansurface, followed by culture in virus-freemedium.Labeled ovaries were then cultured in thepresence or absence of GM6001 for 7 days andthe recruitment of the ovarian surface epitheliumwas traced. (B) Ovarian surface epitheliabecame specifically labeled with EGFP (greencolor) by 1 day of in vitro culture with lentivirusEGFP. In the control group, EGFP+ cells(arrowheads) invaded into the ovarian corticalregion and surrounded oocytes (DDX4, redcolor) to form primordial follicles after 7 days offollow-up culture. In the GM6001-treatmentgroup, only a few EGFP+ cells were found toinvade into ovaries, and germline cysts wereobserved in ovaries after culture.(C) Quantification of EGFP+ cells in ovariescultured in the presence or absence ofGM6001. GM6001 significantly suppressedthe number of EGFP cells recruited (n=6).(D,E) Immunostaining of BrdU (blue color) andFOXL2 (red color) in Lgr5-KI mouse ovaries.LGR5 BrdU double-positive cells (arrowheads)were mainly located in the ovarian surfaceepithelium (D). The yellow dashed line shows thesurface LGR5+ cells. Three days of in vitroculture with GM6001 significantly decreased thenumber of LGR5 and BrdU double-positive cellsand inhibited the proliferation of ovarian surfaceLGR5 progenitor cells (n=5) (E). ***P<0.001(t-test), control versus GM6001-treated ovaries.Data are presented as means±s.d. Scale bars:50 µm (B); (D) 20 µm.

2207

RESEARCH ARTICLE Journal of Cell Science (2016) 129, 2202-2212 doi:10.1242/jcs.184267

Journal

ofCe

llScience

Page 7: Notch signaling governs the recruitment of ovarian ... · RESEARCH ARTICLE ADAM10–Notch signaling governs the recruitment of ovarian pregranulosa cells and controls folliculogenesis

addition, the expression of the Notch target genes Hey2 and Hes1was significantly reduced (Fig. 5I,J; P<0.001 for all) by GM6001treatment. We therefore conclude that ADAM10 controls thedifferentiation of LGR5+ cells and recruitment of FOXL2+ cells byregulating Notch signaling in ovarian somatic cells.

Deletion of ADAM10 in vivo disrupts the formation ofprimordial follicles in miceOur data from in vitro experiments outlined above indicate thatADAM10 expressed by ovarian somatic cells plays a key role incontrolling primordial follicle formation. To assess whether

Fig. 5. ADAM10 regulates the recruitment and differentiation of pregranulosa cells through Notch signaling. (A–D) Ovaries were collected from 16.5 dpcwild-type mice and cultured in the presence or absence of the Notch inhibitor DAPT for 3 or 7 days. A considerably increased number of LGR5+ cells wereobserved in the ovary after 7 days of DAPT treatment (n=5). Many LGR5+ cells were found in the ovarian cortical region (dashed line) in the DAPT-treated group(C) compared to control (A). Quantification of LGR5+ cells and FOXL2+ cells showed that the decrease in LGR5+ cells and increase in FOXL2+ cells wassuppressed in DAPT-treated ovaries (D) compared to control (B). (E) Quantification of LGR5 FOXL2 double-positive cells revealed a significant retardation ofLGR5+ to FOXL2+ cell transformation in DAPT-treated ovaries (n=5). The graphs in B, D and E show cell numbers per section. (F,G) Immunostaining andquantitative analysis showed that DAPT inhibited the proliferation of LGR5+ progenitor cells in culture (n=5). LGR5+ (green color); BrdU (blue color); FOXL2+

(red color). Arrowheads show LGR5 BrdU double-positive cells. (H)Western blot analysis of Notch intracellular domain (NICD) protein expression after the in vitroculture of ovaries with vehicle (control), GM6001 or DAPT. Normal expression of NICD was found in control ovaries. As with the Notch inhibitor, DAPT,GM6001 also markedly inhibited the expression of NICD protein in culture. (I,J) The mRNA expression of Notch target genes,Hey2 andHes1,were reduced after3 or 7 days of GM6001 or DAPT treatment. Different letters (A,B, a–c) indicate significant differences between groups (ANOVA and Holm–Sidak test; n=6).**P<0.01, ***P<0.001 (t-test), control versus treated ovaries. Data are presented as means±s.d. Scale bars: 20 µm.

2208

RESEARCH ARTICLE Journal of Cell Science (2016) 129, 2202-2212 doi:10.1242/jcs.184267

Journal

ofCe

llScience

Page 8: Notch signaling governs the recruitment of ovarian ... · RESEARCH ARTICLE ADAM10–Notch signaling governs the recruitment of ovarian pregranulosa cells and controls folliculogenesis

ADAM10 is required in vivo for folliculogenesis, we conditionallydeleted ADAM10 in a somatic lineage of perinatal ovaries using ananti-Müllerian hormone type 2 receptor-Cre (Amhr2-Cre) mouseline (Jamin et al., 2002), which mediates the expression of CRErecombinase in somatic cells of ovaries from 12.5 dpc (Jamin et al.,2002; Jorgez et al., 2004; Vanorny et al., 2014). qRT-PCR analysisof ovaries showed that the levels of Adam10mRNA in Amhr2-Cre;Adam10f/d ovaries was 52.8% that of control littermates (Fig. S4C).This result indicates that Adam10 is partly disrupted in ovaries.Histological analysis of ovaries taken from Amhr2-Cre;Adam10f/d

mice at 7 dpp identified abnormalities in primordial follicleformation (Fig. 6A, yellow, dashed circles). Many germline cystswere observed in the ovaries of Amhr2-Cre;Adam10f/d mice, whichwas in sharp contrast to that observed during normalfolliculogenesis in control ovaries. By counting the number ofgermline cysts and oocyte numbers in cysts, we found that theovaries of Amhr2-Cre;Adam10f/d mice exhibited significantly moreunbroken cysts (114.0±37.6 cysts per ovary; mean±s.d.; Fig. 6B)compared with littermate controls (13.0±9.2 cysts per ovary;Fig. 6B; P<0.001). Many oocytes (892.0±176.9 for mutant versus52.0±36.7 for control oocytes per ovary; Fig. 6C) in Amhr2-Cre;Adam10f/d ovaries remained within the cysts. These data indicatethat ADAM10 in somatic cells of the mouse ovary regulatesprimordial follicle formation under physiological conditions.

DISCUSSIONIn this study, we demonstrate that ADAM10 plays a key role inprimordial follicle formation in the perinatal mouse ovary.Furthermore, we find that ADAM10 controls the recruitment ofFOXL2+ pregranulosa cells by regulating the proliferation anddifferentiation of LGR5+ progenitor cells. In addition, we show thatNotch signaling is the pathway through which ADAM10 regulatesthe recruitment of pregranulosa cells and primordial follicleformation in mice.Recent studies have found that the ovarian surface epithelium,

which is LGR5 positive, appears to be the major origin ofpregranulosa cells (Mork et al., 2012; Ng et al., 2014; Rastetter

et al., 2014). After recruitment from ovarian epithelium, LGR5+

supporting progenitor cells gradually differentiate into pregranulosacells and become Foxl2 positive (Suzuki et al., 2015). Although thedynamics of FOXL2+ cell recruitment have been well established bycell lineage tracing experiments (Mork et al., 2012; Zheng et al.,2014a), the upstream molecular regulation of this process is stillunclear. In the current study, we found that ADAM10, by regulatingNotch signaling, was essential for this process. Protein expressionanalysis of the Notch intracellular domain showed that ADAM10might be responsible for cleavage of the Notch receptor in bothLGR5+ cells and FOXL2+ cells in perinatal ovaries. Moreover, wealso show that the Notch target genes Hey2 and Hes1 (Tromblyet al., 2009) are also indirectly controlled by ADAM10, given thatthe expression ofHey2 andHes1 is mainly restricted to somatic cellsin ovaries.

It is known that Notch signaling influences cell fates, such as celldifferentiation and the cell cycle, particularly in conjunction withstem cell development, in mammals (Chiba, 2006). InhibitingNotch in the stomach reduces the proliferation of LGR5+ gastricstem (progenitor) cells and the differentiation of mucous andendocrine cells (Demitrack et al., 2015). In the intestine, ADAM10–Notch signaling directly targeted the LGR5+ crypt base columnar(CBC) cells and is pivotal to the balance of stemness anddifferentiation of LGR5+ CBC stem cells (Demitrack et al., 2015;Tsai et al., 2014; VanDussen et al., 2012). Our current results showthat ADAM10–Notch signaling is crucial in determining the fate ofLGR5+ cells in the ovary. We hypothesize that the activation ofADAM10–Notch signaling stimulates the differentiation of LGR5+

cells to FOXL2+ cells in the fetal ovary, whereas the quiescence ofthis signal is essential in maintaining the dormancy of the LGR5+

ovarian surface epithelium in the adult ovary.Previous work from our laboratory has demonstrated that the

synchronized development of oocytes and somatic cells is essentialfor primordial follicle formation (Lei et al., 2006). After the deletionof a cell cycle inhibitor gene, p27Kip1 (Cdkn1b), another studyhas reported that the number of somatic pregranulosa cells ispotentially related to the size of the primordial follicle pool

Fig. 6. Deletion of ADAM10 in vivo disruptsthe formation of primordial follicles inmice. (A) Histological analysis of ovaries fromAdam10f/d and Amhr2-Cre;Adam10f/d mice.By 7 dpp, although some normal follicles wereobserved in Amhr2-Cre;Adam10f/d ovaries,many oocytes were still present in germlinecysts within the ovary. This is in sharp contrastto the normal establishment of a primordialfollicle pool in Adam10f/d control ovaries.Yellow dashed lines circle unbroken germlinecysts. (B,C) Quantification of unbroken cystsand oocytes without the surroundingpregranulosa cells in the ovaries of Amhr2-Cre;Adam10f/d (mutant) mice. The ovaries ofAmhr2-Cre;Adam10f/d mice contained largenumbers of oocytes in germline cysts, which isin sharp contrast to the normal follicledevelopment seen in Adam10f/d (control)ovaries (n=6). **P<0.01, ***P<0.001 (t-test),control versus mutant ovaries. Data arepresented as means±s.d. Scale bars: 50 µm.

2209

RESEARCH ARTICLE Journal of Cell Science (2016) 129, 2202-2212 doi:10.1242/jcs.184267

Journal

ofCe

llScience

Page 9: Notch signaling governs the recruitment of ovarian ... · RESEARCH ARTICLE ADAM10–Notch signaling governs the recruitment of ovarian pregranulosa cells and controls folliculogenesis

(Rajareddy et al., 2007). The current study suggests that blockingthe recruitment of either LGR5+ or FOXL2+ cells leads to a failureof primordial follicle formation. Therefore, we conclude that theproper differentiation and proliferation of ovarian supporting cells isessential for the establishment of the primordial follicle pool, andthe recruited number and speed of somatic cells might decide thefinal size of female reproductive reserve. Previous studies havereported there are two waves of primordial follicle formation in theovary. These two primordial follicle populations differ from eachother in terms of their developmental dynamics and their roles inovarian function (Mork et al., 2012; Zheng et al., 2014a,b). Inthe current study, we found that inhibiting ADAM10–Notchsignaling suppressed the formation of the majority of primordialfollicle formation in the ovary. However, we still found that ∼20%of oocytes formed primordial follicles in the medulla whenADAM10–Notch signaling was blocked. Using a cell lineagetracing approach, Zheng et al. have shown that the follicle somaticcells from the first wave of primordial follicles are recruited before16.5 dpc in fetal ovaries (Zheng et al., 2014a). Given thatthe inhibitors of ADAM10–Notch signaling were added intoculture from 16.5 dpc in this study, here, we mainly focused on theadult wave of primordial follicles. It is, however, unknown whetherthe first wave of primordial follicles are controlled by similarmolecular networks in their development, and more exploration isnecessary.In conclusion, we have supplied evidence showing that

ADAM10 plays a key role in determining the recruitment ofpregranulosa cells and early folliculogenesis in mice. This isachieved through ADAM10-mediated Notch signaling thatregulates the proliferation and differentiation of LGR5+ folliclesupporting progenitor cells in fetal ovaries. Our results indicate thatthe proper recruitment of ovarian follicle somatic cells is essentialfor the establishment of the primordial follicle pool in mice. Thesefindings provide important new insights into the mechanisms ofearly ovarian development.

MATERIALS AND METHODSAnimalsAll wild-type mice were obtained from the Laboratory Animal Center of theInstitute of Genetics and Development Biology (Beijing, China). The Lgr5-EGFP-ires-CreERT2 (Lgr5-KI) reporter mice have been described in detailpreviously (Barker et al., 2007). Adam10loxp/loxp (Adam10f/f ) mice weregenerously provided by Dr Xiaohui Wu from the Institute of DevelopmentalBiology and Molecular Medicine at Fudan University, Shanghai, China(Tian et al., 2008). Amhr2-Cremice (Jamin et al., 2002; Jorgez et al., 2004)and Ddx4 (Vasa)-Cre (Gallardo et al., 2007) mice were obtained from theModel Animal Research Center of Nanjing University, Nanjing, China.Ddx4-Cre mice were crossed with Adam10f/f mice to produce Adam10f/d

mice. To generate conditional Adam10 knockouts in somatic cells, Amhr2-Cre mice were then crossed with Adam10f/d mice to produce Amhr2-cre;Adam10f/d mice (Fig. S4A). Genotyping was performed by PCR using theprimer sets detailed in Table S1 (Fig. S4B). Female and male mice werecaged at a ratio of 1:1 overnight and the presence of a vaginal plug wasdesignated 0.5 dpc. The day of birth was considered 0.5 dpp. Birth usuallyoccurred between 19.0 and 19.5 dpc. Protocols and the use of animalsconformed to the guidelines and regulatory standards of the InstitutionalAnimal Care and Use Committee of China Agricultural University.

Ovary isolation and cultureOvaries were dissected from mice and ovarian capsules were removed inpre-cooled PBS (10 mM, pH 7.4) under a stereomicroscope. Ovaries werecultured in six-well culture dishes in 1.1 ml basic Dulbecco’s modifiedEagle’s medium (DMEM) with F12 medium (GIBCO, Life Technologies)at 37°C in a 5% CO2, 95% air atmosphere with saturated humidity. The

ovaries were turned every 12 h to ensure the all of the ovary cells spent halfthe time in the medium and half in the air.

To assess the role of ADAM10 in primordial follicle formation, wecultured 16.5 dpc ovaries (five ovaries/group) for 3 to 7 days in eithermedium alone, or in medium supplemented with the ADAM10 inhibitorGM6001 (sc-203979; Santa Cruz Biotechnology) or GI254023X (A4436;Apexbio). To analyze Notch signaling during primordial follicle formation,16.5 dpc ovaries were cultured in the presence or absence of the Notchinhibitor DAPT (D5942; Sigma-Aldrich) for 3 to 7 days. For measurementsin the bromodeoxyuridine (BrdU) incorporation assay, isolated ovaries fromdifferent days, or cultured ovaries with inhibitors, were cultured with BrdU(B5002; Sigma) for 1 h before ovaries were collected for analysis.

Histology and follicle characterizationOvaries were fixed in 4% paraformaldehyde for over 24 h at 4°C andthen processed to obtain 5-μm paraffin sections. Sections were stainedwith hematoxylin, and the numbers of oocytes and follicles were countedin every fifth section. To estimate the total number of oocytes and folliclesin each ovary, the sum of counted oocytes and follicles was multipliedby five.

Immunofluorescence and labeled cell characterizationOvaries were fixed in 4% paraformaldehyde for over 24 h at 4°C andprocessed to obtain 5-μm paraffin sections. To ensure the comparability ofdifferent ovaries, all ovaries were sliced through the long plane and were cutinto kidney-shaped sections by adjusting the orientation of the ovary whenembedded. Whole-ovary serial sections were made and ten sections in themiddle of the band were selected for subsequent experiments. Sections werede-paraffinized, rehydrated and subjected to microwave antigen retrieval in0.01% sodium citrate buffer (pH 6.0) for 16 min. Ovarian sections werethen blocked with 10% normal donkey serum in PBS for 1 h at roomtemperature and incubated overnight at 4°C with primary antibody. Primaryantibodies were: chicken anti-EGFP (ab13970; Abcam) used at 1:300; goatanti-FOXL2 (IMG-3228; Imgenex/Novus Biologicals) used at 1:400; rabbitanti-DDX4 (ab13840; Abcam) used at 1:300; and mouse anti-BrdU [G3G4;Developmental Studies Hybridoma Bank (DSHB)] used at 1:300antibodies. After washing thoroughly with PBS, slides were incubatedwith Alexa-Fluor-350-, -488- or -555-conjugated donkey secondaryantibodies against mouse, rabbit, goat or chicken IgG (1:100, Invitrogen/Life Technologies) for 1 h at 37°C. Slides were subsequently washed withPBS again and stained with Hoechst 33342 (B2261; Sigma) or propidiumiodide (421301; BioLegend) as a nuclear counterstain. Sections wereexamined and photographed using a Nikon Eclipse 80i digital fluorescencemicroscope.

To count labeled cells, whole ovarian sections were photographed. Thecortical region was defined as 25% of the whole ovary from the ovariansurface to the ovarian center in this study, which was based on thelocalization of LGR5+ cells, FOXL2+ cells and germline cysts (arrowheads)in fetal ovaries at 16.5 dpc (Fig. S2H). Other days of ovarian sections in vivoor in vitro also used this method to define regions for counting. All positivecells in the cortical region of each section were counted and analyzed. Theimages were merged to analyze double-labeled cells. At least three differentsections were analyzed for each ovary.

ImmunoblottingWestern blot analyses were conducted as described previously (Wang et al.,2015). Cultured ovaries were collected and lysed in WIP (BeijingCellChip Biotechnology Co.), according to the manufacturer’s protocol.Electrophoresis was performed using 50 μg of total protein per sampleseparated by 15% SDS-PAGE and transferred to polyvinylidene fluoridemembranes (IPVH00010, Millipore). Membranes were incubated overnightat 4°C with anti-Notch2 antibody (Abcam) diluted 1:200, which detects theNotch2 full-length band at 120 kDa and the Notch2 intracellular domain at85 kDa. The secondary antibodies (ZB-2301, ZB-2305 from ZSGB-BIO)were diluted 1:5000 in TBST (TBS plus 0.5% Tween 20). The membraneswere visualized using a Super Signal Chemiluminescent Detection System(34080; Thermo Scientific). The level of GAPDH (anti-GAPDH AM4300;Life Technologies; 1:4000) was used as an internal control.

2210

RESEARCH ARTICLE Journal of Cell Science (2016) 129, 2202-2212 doi:10.1242/jcs.184267

Journal

ofCe

llScience

Page 10: Notch signaling governs the recruitment of ovarian ... · RESEARCH ARTICLE ADAM10–Notch signaling governs the recruitment of ovarian pregranulosa cells and controls folliculogenesis

Quantitative real-time RT-PCRRNA was isolated from eight ovaries for each sample using TRIZOL(Invitrogen/Life Technologies) according to the manufacturer’s protocol.Reverse transcription reactions were carried out using 1 μg RNA and aPromega Reverse Transcription System according to the manufacturer’sinstructions. qRT-PCR reactions were undertaken and analyzed using anABI 7500 Sequence Detection System (Applied Biosystems). Data werenormalized to results with Gapdh. The primers used for testing genes arelisted in Table S2.

Infection of ovariesLentivirus EGFP vector was kindly provided by Professor Haibin Wangfrom the Institute of Zoology, Chinese Academy of Sciences, Beijing,China. Packaged virus (≥1×108 transducing units/ml) was diluted inDMEM/F12 (1:1) and 15.5 dpc ovaries were incubated in drops of 20 μlDMEM/F12 medium with lentiviral vectors for 12 h. Infected ovaries werethen transferred into six-well culture dishes with 1.1 ml basic DMEM/F12medium for 12 h to ensure full virus infection. Finally, the infected ovariesfrom one drop were, respectively, cultured in the presence and absence ofinhibitors to examine the behavior of labeled cells.

FACS analysisLGR5–EGFP-positive ovaries from Lgr5-EGFP-ires-CreERT2 (Lgr5-KI)reporter mice were used in FACS analysis. After 7 days of culture in thecontrol or inhibitor treatment group, ovaries were incubated with pre-warmed 0.25% trypsin (Invitrogen) at 37°C and 5% CO2 for 15 min. Thenovaries were dissociated into single-cell suspensions. Following trypsininactivation with PBS supplemented with 10% fetal bovine serum (FBS),ovarian cells were pelleted at 1500 g for 5 min at 4°C, re-suspended in PBSwith 5% FBS and were immediately analyzed in a FACS Calibur (BDBiosciences) machine.

In situ hybridizationIn situ hybridization was performed as described previously (Zhang et al.,2015). Briefly, ovaries with ovarian capsules were dissected from mice andwere embedded in optimal cutting temperature (OCT) compound and frozenin liquid nitrogen until analysis. Frozen sections (10 μm) of ovarieswere hybridized with a mouse-specific DIG-labeled Adam10 probeaccording to the manufacturer’s protocol. Primers used to generate theAdam10 amplicon were: forward 5′-TATGGTGGAGTTGGTTCTTA-3′and reverse 5′-GCAGGCTTGAAGTATATGG-3′. Sections hybridizedwith sense probes served as negative controls and did not show positivesignals.

Statistical analysisAll culture, immunochemical and immunofluorescence analyses wererepeated at least three times using ovaries from different fetuses. Data areexpressed as means±s.d. with each experiment performed in triplicate. Datawere analyzed by t-test. Values of P<0.05 were considered statisticallysignificant.

AcknowledgementsWe are grateful to Professor Haibin Wang’s laboratory (State Key Laboratory ofReproductive Biology, Institute of Zoology, Chinese Academy of Sciences, China)for help in experimental techniques.We are also grateful to Dr. Xiaohui Wu (Instituteof Developmental Biology and Molecular Medicine, Fudan University, Shanghai,China), Professor Enkui Duan (State Key Laboratory of Reproductive Biology,Institute of Zoology, Chinese Academy of Sciences, China) and Dr. Ting Chen(National Institute of Biological Sciences, Beijing, China) for providing transgenicmice.

Competing interestsThe authors declare no competing or financial interests.

Author contributionsL.F., H.Z. andG.X. designed thework, with input from other authors. L.F., Y.W., H.C.,G.S., X.T. and J.Z. performed the experiments. L.F, Y.W., W.N., Q.X. and H.Z.analyzed the data and contributed reagents, materials and/or analytical tools. Themanuscript was written by L.F. and revised by C.W., H.Z. and G.X.

FundingThis study was supported by grants from the National Natural Science Foundation ofChina [grant numbers 31571542 to H.Z., 31571540 to G.X.]; and the National BasicResearchProgramofChina (973, part of theMinistry of Science andTechnologyof thePeople’sRepublic ofChina) [grantnumbers2013CB945501, 2012CB944701 toG.X.].

Supplementary informationSupplementary information available online athttp://jcs.biologists.org/lookup/suppl/doi:10.1242/jcs.184267/-/DC1

ReferencesBarker, N., van Es, J. H., Kuipers, J., Kujala, P., van denBorn, M., Cozijnsen, M.,

Haegebarth, A., Korving, J., Begthel, H., Peters, P. J. et al. (2007).Identification of stem cells in small intestine and colon by marker gene Lgr5.Nature 449, 1003-1007.

Borggrefe, T. and Oswald, F. (2009). The Notch signaling pathway: transcriptionalregulation at Notch target genes. Cell. Mol. Life Sci. 66, 1631-1646.

Bramhall, N. F., Shi, F., Arnold, K., Hochedlinger, K. and Edge, A. S. B. (2014).Lgr5-positive supporting cells generate new hair cells in the postnatal cochlea.Stem Cell Rep. 2, 311-322.

Chiba, S. (2006). Notch signaling in stem cell systems. Stem Cells 24, 2437-2447.Demitrack, E. S., Gifford, G. B., Keeley, T. M., Carulli, A. J., VanDussen, K. L.,

Thomas, D., Giordano, T. J., Liu, Z., Kopan, R. and Samuelson, L. C. (2015).Notch signaling regulates gastric antral LGR5 stem cell function. EMBO J. 34,2522-2536.

Gallardo, T., Shirley, L., John, G. B. and Castrillon, D. H. (2007). Generation of agerm cell-specific mouse transgenic Cre line, Vasa-Cre. Genesis 45, 413-417.

Gibb, D. R., Saleem, S. J., Chaimowitz, N. S., Mathews, J. and Conrad, D. H.(2011). The emergence of ADAM10 as a regulator of lymphocyte developmentand autoimmunity. Mol. Immunol. 48, 1319-1327.

Glomski, K., Monette, S., Manova, K., De Strooper, B., Saftig, P. and Blobel,C. P. (2011). Deletion of Adam10 in endothelial cells leads to defects in organ-specific vascular structures. Blood 118, 1163-1174.

Guo, Q., Wang, Y., Tripathi, P., Manda, K. R., Mukherjee, M., Chaklader, M.,Austin, P. F., Surendran, K. and Chen, F. (2015). Adam10 mediates the choicebetween principal cells and intercalated cells in the kidney. J. Am. Soc. Nephrol.26, 149-159.

Hayashida, K., Bartlett, A. H., Chen, Y. and Park, P. W. (2010). Molecular andcellular mechanisms of ectodomain shedding. Anat. Rec. 293, 925-937.

Hoettecke, N., Ludwig, A., Foro, S. and Schmidt, B. (2010). Improved synthesis ofADAM10 inhibitor GI254023X. Neurodegener. Dis. 7, 232-238.

Hundhausen, C., Misztela, D., Berkhout, T. A., Broadway, N., Saftig, P., Reiss,K., Hartmann, D., Fahrenholz, F., Postina, R., Matthews, V. et al. (2003). Thedisintegrin-like metalloproteinase ADAM10 is involved in constitutive cleavage ofCX3CL1 (fractalkine) and regulates CX3CL1-mediated cell-cell adhesion. Blood102, 1186-1195.

Jamin, S. P., Arango, N. A., Mishina, Y., Hanks, M. C. and Behringer, R. R.(2002). Requirement of Bmpr1a for Mullerian duct regression during male sexualdevelopment. Nat. Genet. 32, 408-410.

Jorgez, C. J., Klysik, M., Jamin, S. P., Behringer, R. R. andMatzuk, M. M. (2004).Granulosa cell-specific inactivation of follistatin causes female fertility defects.Mol. Endocrinol. 18, 953-967.

Jorissen, E., Prox, J., Bernreuther, C., Weber, S., Schwanbeck, R., Serneels, L.,Snellinx, A., Craessaerts, K., Thathiah, A., Tesseur, I. et al. (2010). Thedisintegrin/metalloproteinase ADAM10 is essential for the establishment of thebrain cortex. J. Neurosci. 30, 4833-4844.

Lei, L., Zhang, H., Jin, S., Wang, F., Fu, M., Wang, H. and Xia, G. (2006). Stage-specific germ-somatic cell interaction directs the primordial folliculogenesis inmouse fetal ovaries. J. Cell. Physiol. 208, 640-647.

Lemjabbar, H. and Basbaum, C. (2002). Platelet-activating factor receptor andADAM10 mediate responses to Staphylococcus aureus in epithelial cells. Nat.Med. 8, 41-46.

Lo Sardo, V., Zuccato, C., Gaudenzi, G., Vitali, B., Ramos, C., Tartari, M., Myre,M. A., Walker, J. A., Pistocchi, A., Conti, L. et al. (2012). An evolutionary recentneuroepithelial cell adhesion function of huntingtin implicates ADAM10-Ncadherin. Nat. Neurosci. 15, 713-721.

Maatouk, D. M., Mork, L., Hinson, A., Kobayashi, A., McMahon, A. P. and Capel,B. (2012). Germ cells are not required to establish the female pathway in mousefetal gonads. PLoS ONE 7, e47238.

Mandl, A. M. and Zuckerman, S. (1951). The relation of age to numbers of oocytes.J. Endocrinol. 7, 190-193.

Mork, L., Maatouk, D. M., McMahon, J. A., Guo, J. J., Zhang, P., McMahon, A. P.and Capel, B. (2012). Temporal differences in granulosa cell specification in theovary reflect distinct follicle fates in mice. Biol. Reprod. 86, 37.

Ng, A., Tan, S., Singh, G., Rizk, P., Swathi, Y., Tan, T. Z., Huang, R. Y-J.,Leushacke, M. and Barker, N. (2014). Lgr5 marks stem/progenitor cells in ovaryand tubal epithelia. Nat. Cell Biol. 16, 745-757.

Pepling, M. E. (2012). Follicular assembly: mechanisms of action. Reproduction143, 139-149.

2211

RESEARCH ARTICLE Journal of Cell Science (2016) 129, 2202-2212 doi:10.1242/jcs.184267

Journal

ofCe

llScience

Page 11: Notch signaling governs the recruitment of ovarian ... · RESEARCH ARTICLE ADAM10–Notch signaling governs the recruitment of ovarian pregranulosa cells and controls folliculogenesis

Pepling, M. E. and Spradling, A. C. (1998). Female mouse germ cells formsynchronously dividing cysts. Development 125, 3323-3328.

Pepling, M. E., Sundman, E. A., Patterson, N. L., Gephardt, G. W., Medico, L., Jrand Wilson, K. I. (2010). Differences in oocyte development and estradiolsensitivity among mouse strains. Reproduction 139, 349-357.

Rajareddy, S., Reddy, P., Du, C., Liu, L., Jagarlamudi, K., Tang, W., Shen, Y.,Berthet, C., Peng, S. L., Kaldis, P. et al. (2007). p27kip1 (cyclin-dependentkinase inhibitor 1B) controls ovarian development by suppressing follicleendowment and activation and promoting follicle atresia in mice. Mol.Endocrinol. 21, 2189-2202.

Rastetter, R. H., Bernard, P., Palmer, J. S., Chassot, A.-A., Chen, H., Western,P. S., Ramsay, R. G., Chaboissier, M.-C. andWilhelm, D. (2014). Marker genesidentify three somatic cell types in the fetal mouse ovary. Dev. Biol. 394, 242-252.

Reiss, K. and Saftig, P. (2009). The “a disintegrin and metalloprotease” (ADAM)family of sheddases: physiological and cellular functions. Semin. Cell Dev. Biol.20, 126-137.

Saftig, P. and Reiss, K. (2011). The “a disintegrin and metalloproteases” ADAM10and ADAM17: novel drug targets with therapeutic potential? Eur. J. Cell Biol. 90,527-535.

Schmidt, D., Ovitt, C. E., Anlag, K., Fehsenfeld, S., Gredsted, L., Treier, A.-C.and Treier, M. (2004). The murine winged-helix transcription factor Foxl2 isrequired for granulosa cell differentiation and ovary maintenance. Development131, 933-942.

Suzuki, H., Kanai-Azuma, M. and Kanai, Y. (2015). From sex determination toinitial folliculogenesis in mammalian ovaries: morphogenetic waves along theanteroposterior and dorsoventral axes. Sex. Dev. 9, 190-204.

Tian, L., Wu, X., Chi, C., Han, M., Xu, T. and Zhuang, Y. (2008). ADAM10 isessential for proteolytic activation of Notch during thymocyte development. Int.Immunol. 20, 1181-1187.

Trombly, D. J., Woodruff, T. K. and Mayo, K. E. (2009). Suppression of Notchsignaling in the neonatal mouse ovary decreases primordial follicle formation.Endocrinology 150, 1014-1024.

Tsai, Y.-H., VanDussen, K. L., Sawey, E. T., Wade, A. W., Kasper, C., Rakshit, S.,Bhatt, R. G., Stoeck, A., Maillard, I., Crawford, H. C. et al. (2014). ADAM10regulates Notch function in intestinal stem cells of mice. Gastroenterology 147,822-834.e13.

VanDussen, K. L., Carulli, A. J., Keeley, T. M., Patel, S. R., Puthoff, B. J.,Magness, S. T., Tran, I. T., Maillard, I., Siebel, C., Kolterud, A. et al. (2012).

Notch signaling modulates proliferation and differentiation of intestinal crypt basecolumnar stem cells. Development 139, 488-497.

Vanorny, D. A., Prasasya, R. D., Chalpe, A. J., Kilen, S. M. and Mayo, K. E.(2014). Notch signaling regulates ovarian follicle formation and coordinatesfollicular growth. Mol. Endocrinol. 28, 499-511.

Wang, Y., Teng, Z., Li, G., Mu, X., Wang, Z., Feng, L., Niu, W., Huang, K., Xiang,X., Wang, C. et al. (2015). Cyclic AMP in oocytes controls meiotic prophase I andprimordial folliculogenesis in the perinatal mouse ovary. Development 142,343-351.

Weber, S. andSaftig, P. (2012). Ectodomain shedding and ADAMs in development.Development 139, 3693-3709.

Weber, S., Niessen, M. T., Prox, J., Lullmann-Rauch, R., Schmitz, A.,Schwanbeck, R., Blobel, C. P., Jorissen, E., de Strooper, B., Niessen, C. M.et al. (2011). The disintegrin/metalloproteinase Adam10 is essential for epidermalintegrity and Notch-mediated signaling. Development 138, 495-505.

Xu, J. and Gridley, T. (2013). Notch2 is required in somatic cells for breakdown ofovarian germ-cell nests and formation of primordial follicles. BMC Biol. 11, 13.

Zhang, C., Tian, L., Chi, C., Wu, X., Yang, X., Han, M., Xu, T., Zhuang, Y. andDeng, K. (2010). Adam10 is essential for early embryonic cardiovasculardevelopment. Dev. Dyn. 239, 2594-2602.

Zhang, H., Chang, M., Hansen, C. N., Basso, D. M. and Noble-Haeusslein, L. J.(2011). Role of matrix metalloproteinases and therapeutic benefits of theirinhibition in spinal cord injury. Neurotherapeutics 8, 206-220.

Zhang, H., Liu, L., Li, X., Busayavalasa, K., Shen, Y., Hovatta, O., Gustafsson,J.-A. and Liu, K. (2014). Life-long in vivo cell-lineage tracing shows that nooogenesis originates from putative germline stem cells in adult mice. Proc. Natl.Acad. Sci. USA 111, 17983-17988.

Zhang, W., Yang, Y., Liu, W., Chen, Q., Wang, H., Wang, X., Zhang, Y., Zhang, M.and Xia, G. (2015). Brain natriuretic peptide and C-type natriuretic peptidemaintain porcine oocyte meiotic arrest. J. Cell. Physiol. 230, 71-81.

Zheng, W., Zhang, H., Gorre, N., Risal, S., Shen, Y. and Liu, K. (2014a). Twoclasses of ovarian primordial follicles exhibit distinct developmental dynamics andphysiological functions. Hum. Mol. Genet. 23, 920-928.

Zheng, W., Zhang, H. and Liu, K. (2014b). The two classes of primordial follicles inthe mouse ovary: their development, physiological functions and implications forfuture research. Mol. Hum. Reprod. 20, 286-292.

2212

RESEARCH ARTICLE Journal of Cell Science (2016) 129, 2202-2212 doi:10.1242/jcs.184267

Journal

ofCe

llScience