follicle dynamics and global organization in the intact ... · follicle dynamics and global...

11
Follicle dynamics and global organization in the intact mouse ovary Mehlika Faire a,1 , Amanda Skillern a,1 , Ripla Arora a , Daniel H. Nguyen a , Jason Wang b , Chester Chamberlain b , Michael S. German b , Jennifer C. Fung a , Diana J. Laird a,n a Department of Ob/Gyn and Reproductive Sciences, Center for Reproductive Sciences, Eli and Edythe Broad, Center for Regeneration Medicine & Stem Cell Research, United States b Diabetes Center UCSF, 35 Medical Center Way, San Francisco, CA 94043, United States article info Article history: Received 24 December 2014 Received in revised form 23 March 2015 Accepted 8 April 2015 Available online 16 April 2015 keywords: Whole tissue labeling Confocal imaging Ovary Primordial follicle Pancreas Islet of Langerhans abstract Quantitative analysis of tissues and organs can reveal large-scale patterning as well as the impact of perturbations and aging on biological architecture. Here we develop tools for imaging of single cells in intact organs and computational approaches to assess spatial relationships in 3D. In the mouse ovary, we use nuclear volume of the oocyte to read out quiescence or growth of oocytesomatic cell units known as follicles. This in-ovary quantication of non-growing follicle dynamics from neonate to adult ts a mathematical function, which corroborates the model of xed oocyte reserve. Mapping approaches show that radial organization of folliculogenesis established in the newborn ovary is preserved through adulthood. By contrast, inter-follicle clustering increases during aging with different dynamics depending on size. These broadly applicable tools can reveal high dimensional phenotypes and age- related architectural changes in other organs. In the adult mouse pancreas, we nd stochastic radial organization of the islets of Langerhans but evidence for localized interactions among the smallest islets. & 2015 Elsevier Inc. All rights reserved. Introduction Recent advances in biological imaging and 3D analysis enable visualization of entire embryos and organs at subcellular resolu- tion. The synthesis of microscopic detail at a macroscopic level is revealing features of tissue architecture such as cell orientation and cell shape as they relate to global properties such as mor- phogenesis (Le Garrec et al., 2013; Veeman and Smith, 2012). Comparison of such phenotypes across development, genetic backgrounds and different environments will begin to link tissue architecture to mechanism. Technical obstacles to wholemount imaging include penetration of the tissue with xative and uor- escent labeling agents, optical clearing, and digital selection of the structures of interest in 3D reconstructed images. Algorithms have been developed for wholemount analysis of branching structures such as the kidney glomeruli (Short et al., 2014), but not for dis- crete units such as ovarian follicles. Study of the ovary has been limited by information that can be obtained from histologic sections and the associated labor-intensive processing. Female reproductive lifespan and ovarian function depend upon the reserve oocyte population. Oocytesomatic cell complexes termed primordial follicles (PFs) are established peri- natally in mice with the encapsulation of a meiotically-arrested oocyte by an epithelial layer of granulosa cells (Pepling and Spradling, 2001). Activation of cohorts of PFs for growth begins immediately following their formation, with the proliferation of granulosa cells and concomitant expansion of the oocyte. Although it remains unclear whether oocyte or granulosa cells initiate growth (Hirsheld, 1992), morphometric analyses of mice and humans have revealed an increase in the diameter of the oocyte as well as the oocyte nucleus accompanying PF activation and early growth (Moore et al., 1974; Westergaard et al., 2007). Folliculogenesis progresses through primary, secondary and pre-antral stages and, before puberty, culminates in follicle demise; thereafter, endocrine signals cyclically rescue further follicle growth and promote meiotic resumption and ovulation (McGee and Hsueh, 2000). When the number of remaining PFs falls below a critical threshold, ovulation ceases and menopause ensues (Gosden et al., 1983; Richardson et al., 1987). Exhaustion of the ovarian reserve depends upon multiple factors: initial endowment of follicles, rate of their loss, and rate of follicle activation (Nelson et al., 2013). Despite the importance of the quiescent PF reserve to fertility and ovarian function, clinical assessment is indirect, using growing antral follicle counts and levels of hormones produced by the pituitary and granulosa cells as proxies (Rosen et al., 2012). Experimentally, the number of PFs is estimated in adults by manual counting in selected Contents lists available at ScienceDirect journal homepage: www.elsevier.com/locate/developmentalbiology Developmental Biology http://dx.doi.org/10.1016/j.ydbio.2015.04.006 0012-1606/& 2015 Elsevier Inc. All rights reserved. n Corresponding author. E-mail address: [email protected] (D.J. Laird). 1 These authors have contributed equally. Developmental Biology 403 (2015) 6979

Upload: others

Post on 09-Aug-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Follicle dynamics and global organization in the intact ... · Follicle dynamics and global organization in the intact mouse ovary Mehlika Fairea,1, Amanda Skillerna,1, Ripla Aroraa,

Developmental Biology 403 (2015) 69–79

Contents lists available at ScienceDirect

Developmental Biology

http://d0012-16

n CorrE-m1 Th

journal homepage: www.elsevier.com/locate/developmentalbiology

Follicle dynamics and global organization in the intact mouse ovary

Mehlika Faire a,1, Amanda Skillern a,1, Ripla Arora a, Daniel H. Nguyen a, Jason Wang b,Chester Chamberlain b, Michael S. German b, Jennifer C. Fung a, Diana J. Laird a,n

a Department of Ob/Gyn and Reproductive Sciences, Center for Reproductive Sciences, Eli and Edythe Broad, Center for Regeneration Medicine & Stem CellResearch, United Statesb Diabetes Center UCSF, 35 Medical Center Way, San Francisco, CA 94043, United States

a r t i c l e i n f o

Article history:Received 24 December 2014Received in revised form23 March 2015Accepted 8 April 2015Available online 16 April 2015

keywords:Whole tissue labelingConfocal imagingOvaryPrimordial folliclePancreasIslet of Langerhans

x.doi.org/10.1016/j.ydbio.2015.04.00606/& 2015 Elsevier Inc. All rights reserved.

esponding author.ail address: [email protected] (D.J. Laird).ese authors have contributed equally.

a b s t r a c t

Quantitative analysis of tissues and organs can reveal large-scale patterning as well as the impact ofperturbations and aging on biological architecture. Here we develop tools for imaging of single cells inintact organs and computational approaches to assess spatial relationships in 3D. In the mouse ovary, weuse nuclear volume of the oocyte to read out quiescence or growth of oocyte–somatic cell units known asfollicles. This in-ovary quantification of non-growing follicle dynamics from neonate to adult fits amathematical function, which corroborates the model of fixed oocyte reserve. Mapping approaches showthat radial organization of folliculogenesis established in the newborn ovary is preserved throughadulthood. By contrast, inter-follicle clustering increases during aging with different dynamicsdepending on size. These broadly applicable tools can reveal high dimensional phenotypes and age-related architectural changes in other organs. In the adult mouse pancreas, we find stochastic radialorganization of the islets of Langerhans but evidence for localized interactions among the smallest islets.

& 2015 Elsevier Inc. All rights reserved.

Introduction

Recent advances in biological imaging and 3D analysis enablevisualization of entire embryos and organs at subcellular resolu-tion. The synthesis of microscopic detail at a macroscopic level isrevealing features of tissue architecture such as cell orientationand cell shape as they relate to global properties such as mor-phogenesis (Le Garrec et al., 2013; Veeman and Smith, 2012).Comparison of such phenotypes across development, geneticbackgrounds and different environments will begin to link tissuearchitecture to mechanism. Technical obstacles to wholemountimaging include penetration of the tissue with fixative and fluor-escent labeling agents, optical clearing, and digital selection of thestructures of interest in 3D reconstructed images. Algorithms havebeen developed for wholemount analysis of branching structuressuch as the kidney glomeruli (Short et al., 2014), but not for dis-crete units such as ovarian follicles.

Study of the ovary has been limited by information that can beobtained from histologic sections and the associated labor-intensiveprocessing. Female reproductive lifespan and ovarian functiondepend upon the reserve oocyte population. Oocyte–somatic cell

complexes termed primordial follicles (PFs) are established peri-natally in mice with the encapsulation of a meiotically-arrestedoocyte by an epithelial layer of granulosa cells (Pepling andSpradling, 2001). Activation of cohorts of PFs for growth beginsimmediately following their formation, with the proliferation ofgranulosa cells and concomitant expansion of the oocyte. Althoughit remains unclear whether oocyte or granulosa cells initiate growth(Hirshfield, 1992), morphometric analyses of mice and humans haverevealed an increase in the diameter of the oocyte as well as theoocyte nucleus accompanying PF activation and early growth(Moore et al., 1974; Westergaard et al., 2007). Folliculogenesisprogresses through primary, secondary and pre-antral stages and,before puberty, culminates in follicle demise; thereafter, endocrinesignals cyclically rescue further follicle growth and promote meioticresumption and ovulation (McGee and Hsueh, 2000). When thenumber of remaining PFs falls below a critical threshold, ovulationceases and menopause ensues (Gosden et al., 1983; Richardsonet al., 1987). Exhaustion of the ovarian reserve depends uponmultiple factors: initial endowment of follicles, rate of their loss,and rate of follicle activation (Nelson et al., 2013). Despite theimportance of the quiescent PF reserve to fertility and ovarianfunction, clinical assessment is indirect, using growing antral folliclecounts and levels of hormones produced by the pituitary andgranulosa cells as proxies (Rosen et al., 2012). Experimentally, thenumber of PFs is estimated in adults by manual counting in selected

Page 2: Follicle dynamics and global organization in the intact ... · Follicle dynamics and global organization in the intact mouse ovary Mehlika Fairea,1, Amanda Skillerna,1, Ripla Aroraa,

M. Faire et al. / Developmental Biology 403 (2015) 69–7970

histologic sections and applying a multiplication constant (Bristol-Gould et al., 2006a, 2006b).

Here we developed methodology to image oocytes in the intactmouse ovary using wholemount immunofluorescence. We useoocyte nuclear volume to distinguish non-growing from growingfollicles and obtain absolute counts as well as early follicle growthdynamics. We develop approaches for spatial analysis and extendprevious observations on the global patterning of quiescent andactivated zones, which is established in neonatal ovaries; althoughthis architecture endures throughout life, we find localized clus-tering of primordial and early growing follicles that increases inmagnitude during aging. These approaches can be extended toother organ systems with a variety of markers, as we demonstratein the adult mouse pancreas by imaging the islets of Langerhans.

Results

Identification of primordial and growing follicles by nuclear volume inintact ovaries

We established permeabilization and optical clearing para-meters in wholemount mouse ovaries using DNA stains. We thenscreened oocyte-specific antibodies using these conditions.Nuclear antigens were preferred for their resolvability in 3Dimaging, and the previously established expansion of the oocytenucleus with early follicle growth suggested that nuclear volumecould be used to distinguish non-growing from growing follicles(Moore et al., 1974; Fig. S1). In wholemount ovaries at postnatalday one (PD1), immunolabeling with the oocyte-specific tran-scription factor Nobox appeared variable, with larger and more

Fig. 1. Oocyte nuclear markers enable spatial resolution of follicles in wholemount neonaovary (A). Optical z stack slices through a PD1 ovary stained with Nobox (red) and GCNAthe core (B). Extended z-stack projection of Nobox staining in an intact PD5 ovary (distribution of Nobox volumes compared with GCNA at PD1 (D) shows cutoff for PFs at 3of largest object sizes from birth to PD5 to PD7 by the lengthening of the right tail, and

robustly stained objects deepest in the ovary (Fig. 1A); this stain-ing pattern was consistent with the reported onset of Noboxexpression in late fetal oocytes (Suzumori et al., 2002; Rajkovicet al., 2004). A different distribution was observed at PD1 with themarker Germ Cell Nuclear Antigen (GCNA), which labels pre-diplotene oocytes at the periphery of the neonatal ovary (Endersand May, 1994; Kerr et al., 2006): a shell of GCNAþ cells sur-rounded a concentration of Noboxþ cells at the center of PD1ovaries (Fig. 1B). By PD5, the majority of oocytes are incorporatedin follicles (Pepling, 2006) and accordingly we observed NOBOXimmunofluorescence more uniformly throughout the ovary, withindividually-labeled cells readily separable in 3D reconstructionsof confocal stacks (Figs. 1C, S2A). Thus wholemount staining of theneonatal ovary recapitulates the temporal and spatial dynamics ofa nuclear marker of meiotic prophase marker (GCNA) and theNobox homeobox transcription factor, which is required for thepostnatal differentiation of oocytes and folliculogenesis (Rajkovicet al., 2004).

Volumes of objects defined by Nobox and GCNA immunos-taining were examined in frequency distribution. At PD1, near theonset of follicle formation (Pepling, 2006), comparison of theseprofiles reveals similar range of sizes but smaller average volumeof Noboxþ objects (Fig. 1D); this difference, together with theshoulder in Nobox distribution above 1500 μm3 may reflect theearliest wave of NOBOX expression. As only oogonial cysts and PFsare present at PD1 (Pepling and Spradling, 2001), we used thistimepoint to set an upper limit for detectable Nobox volumes. Athreshold of 3000 μm3 for PF nuclei was rationalized, as thisincludes 499.5% of both GCNA and Nobox volumes at PD1. Mea-surements of oocyte nuclear volume in PFs from thick histologicsections revealed a similar distribution: 2604 μm3 represents

tal mouse ovaries. Extended projections of Nobox immunostaining in an intact PD1(blue) show surface localization of the GCNAþ cells and exclusive Noboxþ cells inC). Selection of Noboxþ objects is indicated by separate colors, inset. Frequency000 μm3. Comparison of Nobox frequency distributions reveals a stepwise increasea concomitant loss of smallest objects (E). Scale bars represent 150 μm.

Page 3: Follicle dynamics and global organization in the intact ... · Follicle dynamics and global organization in the intact mouse ovary Mehlika Fairea,1, Amanda Skillerna,1, Ripla Aroraa,

M. Faire et al. / Developmental Biology 403 (2015) 69–79 71

3 standard deviations from a mean of 1169 mm3 (Fig. S1C and D).Discrepancies between nuclear volumes obtained in wholemountand section may arise from differences in fixation conditions,partial loss of nuclei in sections, as well as z-axis distortioninherent to confocal imaging (Murray, 2011). When examined at1 week of age, the smoothness of Nobox frequency distributionssupports the notion that immediate and continuous follicle growthfollows PF formation (Fig. 1E). Elongation of the right tail44500 μm3 of the distribution from PD5 to PD7 likely representsan increase in follicles recruited for growth, whereas the dramaticloss of small Noboxþ objects probably reflects more oocyte deaththan recruitment.

Wholemount staining in pre-pubertal and adult ovariesrevealed greater size variation in Noboxþ objects than in neonates(Fig. 2A–C, Fig. S2B and C, Supplemental Movie 1). Nuclearvolumes reached 40,000 μm3, with overlapping frequency dis-tributions between bilateral ovaries of individuals (Fig. S3A).Similar smoothness and shapes of the Nobox distributions at PD21as compared to adults (Fig. S3B) suggest that early stages of folliclegrowth proceed with similar kinetics before and after puberty.Decrease in the tail of the distribution by 24 weeks (Fig. 2D)suggests a slackening in folliculogenesis that accompanies middleand advanced reproductive age in mice (McGee and Hsueh, 2000).Following superovulation at PD21, we observed a dramaticincrease in ovary size and appearance of large antral follicles(Supplementary Movies 2–3). Nobox profiles revealed an increasein the largest objects (30,000–40,000 μm3; Fig. S4), possiblyreflecting the surge in antral follicles (McGee and Hsueh, 2000).One week following treatment with the alkylating chemother-apeutic agent cyclophosphamide, ovaries from 6 week-old mice

Fig. 2. Wholemount imaging of oocyte nuclei in prepubertal and adult ovaries revealsIn 3D reconstruction of a PD21 ovary (A), a range of object sizes is visible, similar to 6-weantibody penetrates the adult ovary, as shown in optical z stack slices at 24 weeks (C).week) adults (D). One week following cyclophosphamide treatment, the most significanalso decreased. Arrows in all graphs indicate size cutoffs at 3000 and 10,000 μm3 (E). S

retained 37% of Noboxþ objects, with the greatest loss in thevolumes over 10,000 μm3 (Fig. 2E, S3C). This upholds a recentdemonstration that cyclophosphamide directly destroys growingfollicles, especially secondary stages, and causes compensatoryrecruitment of PFs (Titus et al., 2013). Together these observationssuggest that oocyte nuclear volume identified by 3D immuno-fluorescence can distinguish primordial from growing folliclesusing a cutoff of �3000 μm3; a threshold of �10,000 μm3 mayseparate primary from secondary follicles (although the com-paratively dimness of larger and deeper objects makes theirdetection less reliable than small Noboxþ nuclei).

Supplementary material related to this article can be foundonline at doi:10.1016/j.ydbio.2015.04.006.

Quantification of PFs in wholemount ovaries across aging

Using the above criteria, we determined PF counts from oocytenuclear volumes in ovaries from PD5 through 6 months (Fig. 3A).The most acute loss and largest variation in small Noboxþ objectsoccur between PD5 and PD7 (Fig. 3B), consistent with reportedneonatal death during the period of follicle formation in themouse (Pepling, 2012). Increased variation in the number ofNoboxþ objects detected in neonates compared to adults possiblyreflects this period of accelerated follicle loss. The absolute num-ber of small Noboxþ objects drops again in early sexual maturity(5–6 weeks), and thereafter decreases more gradually, while theproportion of medium and large objects increases until 3 monthsand declines at 6 months (Fig. 3B inset). To model the PF popu-lation mathematically, small Nobox data were fitted to a curve. Asshown in Fig. 3C, the best fit was a power function in which the

similar growth dynamics.eks (B); nonspecific staining occurs in vasculature of the hylem (arrowhead). NoboxNobox profiles are largely similar between ovaries of young (12 week) and old (24t losses occur in large Noboxþ objects (410,000 μm3), although small objects arecale bars represent 150 μm in panels A–B, and 600 μm in C.

Page 4: Follicle dynamics and global organization in the intact ... · Follicle dynamics and global organization in the intact mouse ovary Mehlika Fairea,1, Amanda Skillerna,1, Ripla Aroraa,

Fig. 3. Quantification of primordial follicles in mouse ovaries is consistent with a finite ovarian reserve. (A) Nobox-positive objects in juvenile and adult ovaries are tabulatedand represented graphically (B), with relative proportion of each size class in the inset. Volume cutoffs for small, medium and large classes of objects are shown. (C) Themean number of small Noboxþ objects, representing PFs, is plotted versus time and fit to a logarithmic (gray) and power function (black). (D) Examination of PD7 ovarieswith the proliferation marker pHH3 (blue) reveals very rare colabeling with Nobox (red; 5/17,972 in n¼6 ovaries). Scale bar, 20 μm.

M. Faire et al. / Developmental Biology 403 (2015) 69–7972

number of PFs decreases indirectly proportionally to the squareroot of time (t�0.5). This function predicts a PF endowment of�9400 at t¼1 and accordingly, we identified 8235 cells at PD1that expressed either Nobox or GCNA (Fig. S5). Alternatively, if t¼1is defined as embryonic day 14 (E14), which corresponds to themaximal number of oocytes at the time of meiotic entry, thisfunction predicts an original endowment of 15,700—close to priorestimates of �15,000 germ cells per ovary (Kerr et al., 2013). Alogarithmic function also fits the data for small Noboxþ objects;however this intercepts 0 before 8 months, which is inconsistentwith observed fertility of female mice until �1 year of age.

Although these results corroborate the fixed ovarian reservemodel, it remains unresolved whether postnatal oocyte renewal canoccur in rare instances (Kerr et al., 2006; White et al., 2012). Aswholemount imaging is ideal for infrequent cellular events, weexamined Nobox in conjunction with markers of proliferation. Fol-lowing pulses at E14.5 and E16.5 with the thymidine analog EdU, wedid not observe incorporation within Noboxþ objects by PD7 (n¼4;data not shown). Immunostaining revealed colocalization of phos-pho-histone H3 (pHH3) with Nobox in 5 cells across 6 ovaries at PD7(Fig. 3D, Fig. S6, Supplemental Movie 4). Reliable EdU and pHH3detection was not achieved with later ovaries. Together these results

most strongly support a static ovarian reserve but raise the possi-bility of rare oocyte proliferation in neonates.

Radial measurements show that global follicle organization is estab-lished in neonatal ovaries and persists in adults

To assess spatial organization of follicle quiescence andmaturation, we measured radial 3D distances from each Noboxþ

object to either the geometric center or to the surface of the ovary;this surface was mapped using the most peripheral objectsdetected. At PD1, 70% of Noboxþ objects (and all of the largestvolumes) localized within 200 μm of the ovary centroid, whereas70% of GCNAþ objects were excluded from this region (Fig. S5),suggesting that oocyte maturation occurs in the interior of theorgan, either deep in the cortex or in the medullary region. Thearchitecture established by PD5 remains through adulthood: mostNoboxþ objects resided within 100–200 μm of the ovary surface(Fig. 4A–H). We generally observed a positive correlation betweensize and depth of objects (Fig. S7; the converse was found whenmeasured from the centroid, except in irregularly-shaped adultovaries). Thus, consistent with observations in histologic sections(Fig. S8A and B; Byskov et al., 1997, Da Silva-Buttkus et al., 2009),

Page 5: Follicle dynamics and global organization in the intact ... · Follicle dynamics and global organization in the intact mouse ovary Mehlika Fairea,1, Amanda Skillerna,1, Ripla Aroraa,

Fig. 4. Follicle spatial dynamics in neonatal ovaries persist through adulthood. Location of small Noboxþ objects (o3000 μm3, gold), medium-sized (3000–10,000 μm3,brown), and large (410,000 μm3, turquoise) are shown in 3D opacity projections of ovaries at PD5 (A), 3 weeks (C), 6 weeks (E), and 24 weeks (G). For each ovary,measurements from the center of each Nobox object to the ovary surface (B, D, F, H) are shown with respect to object volume. At all stages, small oocyte nuclei (gold) skewtoward the surface, whereas medium sized nuclei most frequently reside 50–150 μm from the surface. Median depths for each size class are indicated by gray bars.Exemplary aggregations of objects in 3D are indicated in panels E and G by arrowheads of the corresponding color. Scale bars represent approximately 150 μm.

M. Faire et al. / Developmental Biology 403 (2015) 69–79 73

Page 6: Follicle dynamics and global organization in the intact ... · Follicle dynamics and global organization in the intact mouse ovary Mehlika Fairea,1, Amanda Skillerna,1, Ripla Aroraa,

M. Faire et al. / Developmental Biology 403 (2015) 69–7974

3D measurements show that PFs concentrate near the surface,while follicle growth initiates at greater cortical depths. Interest-ingly, medium-sized Noboxþ objects accumulate in a swathwithin 50–150 μm of the ovary surface from PD5 through 24weeks (brown in Fig. 4B, D, F and H). Centroid measurementsrevealed absence of Nobox from a region o50 μm from the core atPD5 (Fig. S8C) and up to 200 μm in juvenile and adult ovaries (Fig.S8D–F), and this region may correspond to the medulla. These 3Dmodels capture an outside-in organization of follicle developmentthat begins after birth in mice and continues throughout repro-ductive life; they further point to a potential radial zone of initialfollicle growth at 50–150 μm depth.

Point pattern analysis shows increased clustering of follicles with age

Despite continuity in radial distribution of oocytes within theovary from birth through aging, 3D reconstructions revealed size-dependent differences in the spacing of objects. Whereas neonatalovaries appeared homogenous (Fig. 4A) and juveniles regular(Fig. 4C), aggregations of Nobox objects can be seen by 6 weeks ofage (Fig. 4E and G). We measured spatial homogeneity using Rip-ley’s K function, a technique for point pattern analysis, whichquantifies the degree of object clustering or dispersion as comparedto a random distribution. At PD5 and PD7 Nobox object distribu-tions did not deviate from randomness as compared to all nuclei(Fig. S9A). From 3 through 24 weeks, significant reorganization

Fig. 5. Spatial analysis of follicular clustering with age and follicle size. The distributionsecond-order properties. The K(d)–E(K(d)) curves depict deviations of the follicle distribobject centroids. Positive values indicate clustering while negative values indicate disperC) was evaluated as ovaries matured from 1–24 weeks. Objects of all sizes displayed adifferently-sized follicles at a given stage was also investigated at 3, 6 and 24 weeks (D, Esmall through 6 weeks of age, but become similarly aggregated by 24 weeks. Maximarrowheads for comparison.

imposes an increasingly clustered distribution affecting all oocytesdetected regardless of size (Fig. 5A–C); this clustering does not arisedue to displacement by follicle growth (Fig. S9B). During this period,however, the dynamics of aggregation differed by object size. Largeobjects were initially randomized at 3–6 weeks before becoming assimilarly clustered as smaller objects at 24 weeks (Fig. 5C–F). Incontrast, these smaller Noboxþ objects were already aggregated injuvenile ovaries (3–6 weeks). Thus spatial statistics indicate that themagnitude of localized interactions between nongrowing folliclesincreases over reproductive lifespan and furthermore suggest thatprocesses acting on the large class of growing follicles change withage.

Analysis of the pancreas shows random radial organization and size-specific interactions between islets of Langerhans

We extended these imaging and analysis approaches to discretestructures in other organs. In the adult mouse pancreas, weimmunolabeled endocrine β-cells of the islets of Langerhans withthe lineage-specific transcription factor Nkx6.1 (Sander et al.,2000). Low magnification permitted reassembly of the entirepancreas and coalescence of juxtaposed β-cells into a single object(Fig. 6A). Resulting Nkx6.1þ objects were co-labeled with anti-insulin antibody (data not shown) and 3D-selected in the dorsal(n¼1511) and ventral pancreas (n¼565). Volumes obtainedspanned more than 4 orders of magnitude and exhibited similar

of differently-sized Nobox objects was assessed using Ripley’s K-function to identifyution (K(d)) from randomness (E(K(d)) over a given distance (d) between Noboxþ

sion. The change in distribution of small, medium, and large oocyte nuclei (A, B andn increasing trend towards clustering with age. The comparative distributions ofand F). Large Noboxþ objects are comparatively more dispersed than medium andum diameters for primordial, primary and secondary follicles are indicated by

Page 7: Follicle dynamics and global organization in the intact ... · Follicle dynamics and global organization in the intact mouse ovary Mehlika Fairea,1, Amanda Skillerna,1, Ripla Aroraa,

Fig. 6. 3D analysis of islets of Langerhans in the adult mouse pancreas. Islets were visualized and selected in the pancreas by Nkx6.1 wholemount staining, as shown inextended projection (A). Nkx6.1 volume profiles are similar between dorsal and ventral pancreas (B), with breaks between small, medium and large classes shown by arrows.Distribution of Nkx6.1 object volume versus 3D shape factor (a measure of sphericity) shows a similar trend toward elongation with increasing size for both dorsal andventral pancreas (C). Radial distance analysis of Nkx6.1þ objects from a surface defined by the same objects reveals the absence of correlation between volume and depth (D,E). Clustering analysis of Nkx6.1þ objects in the entire pancreas finds no difference between the largest class and random distribution, but a departure of the small andmedium object relationships K(d) from the expected random distribution E(K(d)) (F). The ratio of these distributions reveals a maximal clustering behavior of small objects ata distance of 40–80 μm, which exceeds the maximal inter-centroid distance of 34 μm (gray arrowhead); a smaller magnitude of attraction occurs between medium sizeNkx6.1 objects at a distance between their minimum (gray arrowhead) and maximum (green arrowhead) adjacent distance (G). Scale bar represents 1 mm.

M. Faire et al. / Developmental Biology 403 (2015) 69–79 75

Page 8: Follicle dynamics and global organization in the intact ... · Follicle dynamics and global organization in the intact mouse ovary Mehlika Fairea,1, Amanda Skillerna,1, Ripla Aroraa,

M. Faire et al. / Developmental Biology 403 (2015) 69–7976

frequency distribution profiles between dorsal and ventral pan-creas (Fig. 6B). An additional parameter of shape factor provided ametric of 3D sphericity of Nkx6.1þ objects, and revealed a trendtoward decreasing sphericity with increasing volume (Fig. 6C).Radial distance measurements from a surface defined by themost peripheral Nkx6.1þ objects revealed random depth withrespect to volume (Fig. 6D). Finally, we examined distancesbetween object centroids of small (4000–20,000 μm3), medium(20,000 - 106 μm3), and large volumes (4106 μm3), as defined bybreaks in the frequency distribution in Fig. 6B; by Ripley’s Kfunction, small and medium objects exhibited greater departurefrom spatial randomness as compared to large (Fig. 6E). A ratio ofthe measured K(d) compared to an expected stochastic distribu-tion E[K(d)] revealed maximal deviation at 40– 80 μm for smallobjects, which exceeds the minimum packing distance of this sizeclass (2r¼34 μm; Fig. 6F). Together these point pattern analysessuggest random dispersal of large islets and localized interactionsbetween the smallest detectable aggregations of β-cells, whichmay reflect their proximity to ducts (Pictet et al., 1972). Consistentwith prior observations (Miller et al., 2009) our 3D shape analysissuggests an elongation of islets accompanies their growth.

Discussion

Here we developed tools for wholemount imaging and 3Dspatial analysis of the mouse ovary and adult pancreas. Thisenables more rapid quantification of ovarian follicles and islets ofLangerhans than was previously possible by serial histologic sec-tioning. Furthermore, the visualization and positional analysis ofthese structures globally and with respect to one another providesquantitative bases for previously qualitative observations as wellas revealing new insights.

At two different biological scales, we exploit volume to differ-entiate stage of development. In the ovary, this is predicated uponthe observation that the cross-sectional area of the mouse andhuman oocyte nucleus scales with the growth of the entire folliclefrom primordial to antral stages (Moore et al., 1974; Westergaardet al., 2007). Uridine incorporation studies suggest that RNAsynthesis in the oocyte underlies this nuclear expansion (Mooreet al., 1974); however more recent work raises the possibility ofcontribution from coincident epigenetic changes. Genome-wideDNA methylation occurs postnatally in mouse oocytes, andmethylation marks at imprinted loci are acquired between pri-mary and secondary follicle stages (Tomizawa et al., 2012). Chro-matin remodeling accompanies the burst of transcription in theearly growing oocyte that ceases by the antral follicle stage, andmay contribute to nuclear expansion. Interestingly, in Xenopusoocytes, nuclear size does not scale with ploidy, as previouslythought, but depends upon the rate of import into the nucleus(Levy and Heald, 2010). Linking these observations is the possibi-lity that nuclear expansion in the growing oocyte results from theinflux of transcription factors and epigenetic modifiers as well aschanging chromatin structure associated with meiosis and tran-scription. Despite established synchronization between folliclestructure and expansion of the entire oocyte, it remains to bedetermined whether growth is initiated by oocyte or granulosacells (Hirshfield, 1992). In this sense, the current method of ana-lysis by oocyte nuclear volume does not precisely read out thestate of follicle growth; on the other hand, such wholemountquantitative imaging of oocytes and granulosa cells simulta-neously may resolve this question. At a different scale, in thepancreas we exploit the volume of β-cell clusters to analyze isletgrowth. Our observations that islet elongation increases withvolume and that small islets are increasingly aggregated comparedto large islets do not distinguish between the possibilities of islet

fission and fusion (Miller et al., 2009). This question could beaddressed with a combination of quantitative wholemount ima-ging and inducible lineage tracing.

Using this higher-throughput methodology to quantify non-growing follicles with greater precision, we find adequate endow-ment at birth and maintenance through middle age. A powerfunction fits these data and predicts �500 PFs remaining at 1 year,matching previous estimates (Bristol-Gould et al., 2006b). Given theprevailing belief that reproductive senescence in mice and meno-pause in humans occurs at a critical but non-zero PF thresholdrather than complete exhaustion (Gosden et al., 1983; Richardsonet al., 1987), the power function may be biologically relevant fordescribing the decline of the ovarian reserve. Wholemount ovaryimaging with cell death markers will facilitate identification ofmechanisms of oocyte loss such as atresia that may contribute topremature ovarian insufficiency (Nelson et al., 2013). Although manystudies in mice including this one corroborate a non-renewingovarian reserve (Begum et al., 2008; Zhang et al., 2012; Lei andSpradling, 2013), the origins and fate of rare proliferating postnataloocytes observed (1 per ovary at PD7) and previously described(Kerr et al., 2006; White et al., 2012) can be further explored inwholemount. Understanding the biological underpinnings of suchevents could be informative to regenerative medicine.

Dynamic and spatial information from intact ovary imagingmay yield insights into mechanisms underlying the activation orquiescence of follicles. We observed striking similarity betweensize profiles of oocyte nuclei in prepubertal, young and aging adultovaries. Despite variation across ages in the rate of recruitmentand early growth of follicles (and hence rightward movement onfrequency distributions), identical shapes of these distributionscould imply identical processes, irrespective of hormones presentin adults. Previous work suggests that growing follicles inhibit therecruitment of PFs (Durlinger et al., 1999; Mizunuma et al., 1999).Consistent with this idea, the observed decline in PFs after che-motherapeutic ablation of growing follicles likely representscompensatory recruitment. Alternatively, the release of growth-inhibiting signals by PFs themselves was posited by a recent spa-tial analysis of neonatal ovaries, in addition to the secretion ofgrowth-promoting factors from early growing follicles (Da Silva-Buttkus et al., 2009). Accordingly, we find that the smallest oocytesconcentrate near the ovary surface, whereas larger, growingoocytes avoid the �50 μm outer radius. Perhaps more impor-tantly, we observe aggregations of small oocytes by puberty thatbecome discrete islands in adults. These behaviors conform to thesecreted growth inhibitor and activator predictions of Hardy andcolleagues (Da Silva-Buttkus et al., 2009) and furthermore raisethe possibility that localized interactions maintain a threshold ofPFs through aging, perhaps in a quorum sensing mechanism(Bristol-Gould et al., 2006b; Tingen et al., 2009). Importantly, theextent of early follicle movement within the ovary is poorlyunderstood, and such spatial models which predict the range ofsecreted factors can only assume stasis; however, the study of PFmigration with live imaging of intact or sliced ovaries will makeinroads to this question. By contrast, the absence of radial orga-nization of islets in the pancreas reflects different biological con-straints. Finally, our spatial analyses reveal discrepancies betweenpubertal and adult ovarian follicle activation that may reflectseparate follicle origins. Consistent with the view that follicleformation in the medulla precedes that in the cortex (Byskov et al.,1997), we find Nobox in the largest oocytes at the center of thePD1 ovary. This pattern persists with the largest growing oocyteslying deep within the 3-week ovary; these likely represent the firstwave of medullary follicles (Hirshfield, 1992), recently shown toderive from a separate population of granulosa cells (Mork et al.,2012; Zheng et al., 2014). Although rescue of medullary follicles bysuperovulation and in vitro fertilization confirms their functional

Page 9: Follicle dynamics and global organization in the intact ... · Follicle dynamics and global organization in the intact mouse ovary Mehlika Fairea,1, Amanda Skillerna,1, Ripla Aroraa,

M. Faire et al. / Developmental Biology 403 (2015) 69–79 77

capability, the biological significance of this first wave of folliclesdestined to die before adulthood remains to be determined.

In conclusion, wholemount imaging increases the throughputand accuracy of counting PFs in the ovary and can be applied todiscrete structures across a range of sizes and organs. The resultingcapability for quantitative phenotyping will advance character-ization of mutants and enhance understanding of follicle pooldynamics over lifetime as related to genetic background, ovarianaging, and exogenous agents.

Materials and methods

Mice

All animal protocols were reviewed and approved by IACUCpolicies of University of California, San Francisco. Mice were main-tained on a mixed genetic background comprising 50% C57BL/6 and50% CD1 in an AAALAC approved facility. Chemotherapy consistedof a single intraperitoneal injection of 75 mg/kg cyclophosphamidegiven at 5 weeks of age, followed by euthanasia 1 week later.Superovulation was carried out at PD21 by intraperitoneal injectionof 50 units of PMSG (Harbor-UCLA Research and Education Insti-tute), followed by euthanasia 2 days later.

Immunofluorescence

Ovaries for sectioning were fixed in 4% paraformaldehydeovernight, cryoprotected in 30% sucrose, embedded and cryosec-tioned in OCT at 5 μm or 25 μm. Sections were washed, blockedwith 1X phosphate buffered saline (PBS)/0.1% Triton X-100/5% goatserum, and incubated overnight in primary antibody. Followingprimary incubation, slides were washed with 1X PBS/0.1% Triton,incubated with secondary antibody, and mounted on slides inVectashield (Vector Labs).

Ovaries for wholemount immunostaining were carefullyremoved from the animal and surrounding bursa under the dis-secting microscope, fixed in 4:1 Methanol:dimethylsulfoxide(DMSO) and stored at �20 °C overnight or longer. Subsequent processing steps were carried out in 0.5 or 1.5 mL eppendorf tubeswhile rocking. Prior to staining, ovaries were washed with 50%Methanol/1X PBS for 30 min at room temperature, then blockedwith 3 one-hour consecutive washes of blocking solution (1X PBS/1%Triton/10% goat serum) at room temperature. Ovaries were incu-bated with primary antibody overnight at 4 °C, washed with block-ing buffer 3�1 h and incubated with secondary antibody overnightat 4 °C. Secondary antibody was removed and ovaries were seriallydehydrated through increasing methanol concentrations (25%, 50%,75% and 100%) for 1 h at each. For PD1s, PD5s, and PD7s, tissueswere cleared in Benzyl Alcohol:Benzoyl Benzoate (BABB,1:2) forminimum of 6 h at room temperature. Ovaries PD21 and older wereincubated in 3% H2O2 in methanol overnight at 4 °C, followed by2�1 h 100% methanol washes, and stored in BABB overnight atroom temperature. All tissues were cleared in 10 mm long glasscylinders (ACE Glass 3865-10) mounted onto coverslips (FisherfinestPremium coverglass cat: 12-548-5P). Primary antibodies used wereas follows: GCNA, a gift from George Enders (undiluted), pHH3(1:100, Sigma H9908), Nobox (1:50 Abcam 41521), Vasa (RnDAF2030) and stains included DAPI (1:500, Roche 236276), Hoechst(1:100 Life Technologies H3569), and Topro (1:100, InvitrogenT3605). Alexa-555 and 633 secondary antibodies were purchasedfrom Invitrogen and used at 1:200.

Pancreas was harvested from 12 week old adult mice whichhad been heart perfused with 4% paraformaldehyde (PFA). Pan-creas was then incubated in 4% PFA over night, washed anddehydrated in methanol before processing as described above. To

allow full antibody penetration, the membrane surrounding thepancreas was removed and the pancreas was vigorously shaken inlarge volumes throughout antibody incubations (20 mL/pancreas)and washes (45 mL/pancreas). Blocking solution was 1X PBS/0.5%Triton/5% donkey and Nkx6.1 primary antibody was used at 1:250(Sigma HPA036774).

Image acquisition

Optical stacks were collected through tissues using an invertedLeica TCS SP5 confocal microscope with a pulsed white light laser.A 10� objective was used for PD21 and older ovaries and pan-creas, while 20� objective was used for PD1, PD5, and PD7ovaries, both at 1024�1024 pixel resolution. Stacks were acquiredat system optimized z steps between optical sections (0.33 μm at20� and 1.98 μm at 10� ). Adult ovaries were tile-scanned in 3–4stacks of approximately 300 images each and pancreas wasreconstructed from 7 x 8 stacks. Gain and offset were manuallyadjusted during the scan to maximize the contrast between thestained objects and surrounding tissue.

Image processing and object selection

Image stacks were visualized in Volocity 6.2 (Improvision) byextended projection of the z-axis and 3D opacity projections,which renders dark voxels transparent. Processing included noiseremoval (fine filter) and contrast adjustment. Objects of interestwere selected by signal intensity threshold, determined as thedimmest visible oocyte using the voxel spy function, typically Z3standard deviations above the mean intensity in the entire stack.Noise and fragmented objects were excluded by volume cutoff; forovaries imaged with the 10� objective, minimum volume was180–300 μm3, while 50 μm3 was used at PD1–7 owing to thehigher resolution of the 20� objective. Volume thresholds werevetted in control tissues stained with secondary antibody to con-trol for debris. Selected objects were manually inspected byscrolling through the stack. Object data and xyz coordinates wereexported to Excel for analysis. For double immunostaining, theintersection of objects was determined by (1) selecting the pri-mary marked population (i.e. Nobox); (2) establishing intensitythreshold for the dimmest objects identified by the second marker,as the minimum number of standard deviations from the meanintensity value (e.g. 3 for GCNA, 63 for pHH3); (3) excludingNoboxþ objects with signal in the second channel below thisthreshold; (4) applying a second size cutoff to the double positiveobjects (50 μm3 for pHH3); (5) manual inspection by scrollingthrough z stacks to verify overlap of both channels. In the pan-creas, Nkx6.1þ objects were selected by automatic threshold usingan offset of �75% and minimum object size 4952 μm3. Subsequentoperations included separate touching objects (size guide of15,000,000 μm3), fill holes, and separate touching objects (sizeguide of 2Eþ08 μm3).

Oocyte spatial analysis

Analysis of follicle distances from the centroid and ovary sur-face was performed using a custom MATLAB program (script isprovided in the Supplemental Methods). For each dataset con-sisting of xyz coordinates and volumes of every Noboxþ object, thecentroid of the entire object set was calculated by averaging x, yand z coordinate values for all oocytes. This reference point wasthen used to calculate the distance from the centroid of eachoocyte to the ovary centroid. To probe the surface of the ovary, thesmallest polygonal surface containing all the oocytes was gener-ated by first calculating a 3D Delaunay triangulation of the com-plete set of follicle coordinates and then extracting the convex hull

Page 10: Follicle dynamics and global organization in the intact ... · Follicle dynamics and global organization in the intact mouse ovary Mehlika Fairea,1, Amanda Skillerna,1, Ripla Aroraa,

M. Faire et al. / Developmental Biology 403 (2015) 69–7978

surface using the built-in convexHull() function. The distance ofeach oocyte from the surface was calculated by determining thedistance from the Nobox object centroid to each facet of thepolygonal convex hull and taking the minimum distance. Corre-lation coefficients were then calculated between the distances toeither the surface or centroid and the follicle volume usingMATLAB’s corrcoef() function.

Clustering analysis was performed on Nobox object coordinatesusing RipleyGUI (Hansson et al., 2013) to calculate Ripley’s Kfunction in three dimensions. The K function describes second-order properties of a spatial point process to evaluate how eventsare distributed in relation to each other. Briefly, the K function isexpressed as the number of objects within a distance d of anarbitrary object divided by the object density of the correspondingsampled volume with radius d. To assess the degree of clusteringfor a follicle distribution, we calculated the difference between theexperimental K score (K(d)) and an expected K score (E(K(d)) for aPoisson distribution following complete spatial randomness.Clustering is indicated by a K(d)4E(K(d)). K functions weredetermined at one-micron intervals (d¼1) to a maximum of200 μm. Comparisons between groups were performed withbootstrapped K(d)–E(K(d)) values.

Statistics

Frequency distributions and graphs were generated in Excel.Statistical tests including Student’s T test, ANOVA, and correlationcoefficients were performed in Excel, MATLAB and Prism; com-parison of Ripley K functions using the between-treatments sumof squares was performed in RipleyGUI.

Author contributions

A.S., D.J.L., and M.F. designed the experiments, A.S., M.F., R.A. C.C.and J.W. carried out the staining and imaging, D.H.N. carried out theclustering analysis, J.C.F. carried out surface and centroid modeling,D.J.L. and M.F. analyzed the data, made the figures and wrote themanuscript, all authors contributed to data interpretation.

Competing interests

None.

Acknowledgments

We thank Matthew Gormley, Paul Miller and David Castanedafor technical help and Marcelle Cedars, Marco Conti, Amy Laird,Walter Miller, Valerie Weaver and members of the Laird lab foruseful discussions and critical feedback. Funding for this work wasprovided by NIH 1DP2OD007420 and the UCSF Research AllocationProgram to D.J.L., NIH R01 GM097213 to J.C.F, California Institute ofRegenerative Medicine Training Grants TB1-01194 to M.F. andTG2-01153 to R.A., NIH Training Grant T32 HD007263 to A.S. andNSF Graduate Research Fellowship 1144247 to D.N.

References

Begum, S., Papaioannou, V.E., Gosden, R.G., 2008. The oocyte population is notrenewed in transplanted or irradiated adult ovaries. Hum. Reprod. 23 (10),2326–2330.

Bristol-Gould, S.K., Kreeger, P.K., Selkirk, C.G., Kilen, S.M., Cook, R.W., Kipp, J.L., Shea,L.D., Mayo, K.E., Woodruff, T.K., 2006a. Postnatal regulation of germ cells byactivin: the establishment of the initial follicle pool. Dev. Biol. 298 (1), 132–148.

Bristol-Gould, S.K., Kreeger, P.K., Selkirk, C.G., Kilen, S.M., Mayo, K.E., Shea, L.D.,Woodruff, T.K., 2006b. Fate of the initial follicle pool: empirical and mathe-matical evidence supporting its sufficiency for adult fertility. Dev. Biol. 298 (1),149–154.

Byskov, A.G., Guoliang, X., Andersen, C.Y., 1997. The cortex–medulla oocyte growthpattern is organized during fetal life: an in-vitro study of the mouse ovary. Mol.Hum. Reprod. 3 (9), 795–800.

Da Silva-Buttkus, P., Marcelli, G., Franks, S., Stark, J., Hardy, K., 2009. Inferringbiological mechanisms from spatial analysis: prediction of a local inhibitor inthe ovary. Proc. Natl. Acad. Sci. USA 106 (2), 456–461.

Durlinger, A.L., Kramer, P., Karels, B., de Jong, F.H., Uilenbroek, J.T., Grootegoed, J.A.,Themmen, A.P., 1999. Control of primordial follicle recruitment by anti-Mul-lerian hormone in the mouse ovary. Endocrinology 140 (12), 5789–5796.

Enders, G.C., May 2nd, J.J., 1994. Developmentally regulated expression of a mousegerm cell nuclear antigen examined from embryonic day 11 to adult in maleand female mice. Dev. Biol. 163 (2), 331–340.

Gosden, R.G., Laing, S.C., Felicio, L.S., Nelson, J.F., Finch, C.E., 1983. Imminent oocyteexhaustion and reduced follicular recruitment mark the transition to acyclicityin aging C57BL/6J mice. Biol. Reprod. 28 (2), 255–260.

Hansson, K., Jafari-Mamaghani, M., Kreiger, P., 2013. RipleyGUI: software for ana-lyzing spatial patterns in 3D cell distributions. Front. Neuroinform. 7, 5.

Hirshfield, A.N.A., 1992. Heterogeneity of cell populations that contribute to theformation of primordial follicles in rats. Biol. Reprod. 47 (3), 466–472.

Kerr, J.B., Duckett, R., Myers, M., Britt, K.L., Mladenovska, T., Findlay, J.K., 2006.Quantification of healthy follicles in the neonatal and adult mouse ovary: evi-dence for maintenance of primordial follicle supply. Reproduction 132 (1),95–109.

Kerr, J.B., Myers, M., Anderson, R.A., 2013. The dynamics of the primordial folliclereserve. Reproduction 146 (6), R205–R215.

Le Garrec, J.F., Ragni, C.V., Pop, S., Dufour, A., Olivo-Marin, J.C., Buckingham, M.E.,Meilhac, S.M., 2013. Quantitative analysis of polarity in 3D reveals local cellcoordination in the embryonic mouse heart. Development 140, 395–404.

Lei, L., Spradling, A.C., 2013. Female mice lack adult germ-line stem cells but sustainoogenesis using stable primordial follicles. Proc. Natl. Acad. Sci. USA 110 (21),8585–8590.

Levy, D.L., Heald, R., 2010. Nuclear size is regulated by importin alpha and Ntf2 inXenopus. Cell 143 (2), 288–298.

McGee, E.A.E., Hsueh, A.J.A., 2000. Initial and cyclic recruitment of ovarian follicles.Endocr. Rev. 21 (2), 200–214.

Miller, K., Kim, A., Kilmnik, G., Jo, J., Moka, U., Periwal, V., Hara, M., 2009. Isletformation during neonatal development in mice. PLoS One 4 (11), e7739.

Mizunuma, H., Liu, X., Andoh, K., Abe, Y., Kobayashi, J., Yamada, K., Yokota, H., Ibuki,Y., Hasegawa, Y., 1999. Activin from secondary follicles causes small preantralfollicles to remain dormant at the resting stage. Endocrinology 140 (1), 37–42.

Moore, G.P., Lintern-Moore, S., Peters, H., Faber, M., 1974. RNA synthesis in themouse oocyte. J. Cell Biol. 60 (2), 416–422.

Mork, L., Maatouk, D.M., McMahon, J.A., Guo, J.J., Zhang, P., McMahon, A.P., Capel, B.,2012. Temporal differences in granulosa cell specification in the ovary reflectdistinct follicle fates in mice. Biol. Reprod. 86 (2), 37–42.

Murray, J.M., 2011. Methods for imaging thick specimens: confocal microscopy,deconvolution, and structured illumination. Cold Spring Harb. Protoc. 2011 (12),1399–1437.

Nelson, S.M., Telfer, E.E., Anderson, R.A., 2013. The ageing ovary and uterus: newbiological insights. Hum. Reprod. Update 19 (1), 67–83.

Pepling, M.E., Spradling, A.C., 2001. Mouse ovarian germ cell cysts undergo pro-grammed breakdown to form primordial follicles. Dev. Biol. 234 (2), 339–351.

Pepling, M.E., 2006. From primordial germ cell to primordial follicle: mammalianfemale germ cell development. Genesis 44 (12), 622–632.

Pepling, M.E., 2012. Follicular assembly: mechanisms of action. Reproduction 143(2), 139–149.

Pictet, R.L., Clark, W.R., Williams, R.H., Rutter, W.J., 1972. An ultrastructural analysisof the developing embryonic pancreas. Dev. Biol. 29 (4), 436–467.

Rajkovic, A., Pangas, S.A., Ballow, D., Suzumori, N., Matzuk, M.M., 2004. NOBOXdeficiency disrupts early folliculogenesis and oocyte-specific gene expression.Science 305 (5687), 1157–1159.

Richardson, S.J., Senikas, V., Nelson, J.F., 1987. Follicular depletion during themenopausal transition: evidence for accelerated loss and ultimate exhaustion.J. Clin. Endocrinol. Metab. 65 (6), 1231–1237.

Rosen, M.P., Johnstone, E., McCulloch, C.E., Schuh-Huerta, S.M., Sternfeld, B., Reijo-Pera, R.A., Cedars, M.I., 2012. A characterization of the relationship of ovarianreserve markers with age. Fertil. Steril. 97 (1), 238–243.

Sander, M., Sussel, L., Conners, J., Scheel, D., Kalamaras, J., Dela Cruz, F., Schwitz-gebel, V., Hayes-Jordan, A., German, M., 2000. Homeobox gene Nkx6.1 liesdownstream of Nkx2.2 in the major pathway of beta-cell formation in thepancreas. Development 127 (24), 5533–5540.

Short, K.M., Combes, A.N., Lefevre, J., Adler, L., Ju, K.M., Georgas, T.M., Lamberton, T.,Cairncross, O., Rumballe, B.A., McMahon, A.P., Hamilton, N.A., Little, M.H., 2014.Global quantification of tissue dynamics in the developing mouse kidney. Dev.Cell 29 (2), 188–202.

Suzumori, N., Yan, C., Matzuk, M.M., Rajkovic, A., 2002. Nobox is a homeobox-encoding gene preferentially expressed in primordial and growing oocytes.Mech. Dev. 111 (1–2), 137–141.

Tingen, C., Kim, A., Woodruff, T.K., 2009. The primordial pool of follicles and nestbreakdown in mammalian ovaries. Mol. Hum. Reprod. 15 (12), 795–803.

Titus, S., Li, F., Stobezki, R., Akula, K., Unsal, E., Jeong, K., Dickler, M., Robson, M.,Moy, K., Goswami, S., Oktay, K., 2013. Impairment of BRCA1-related DNA

Page 11: Follicle dynamics and global organization in the intact ... · Follicle dynamics and global organization in the intact mouse ovary Mehlika Fairea,1, Amanda Skillerna,1, Ripla Aroraa,

M. Faire et al. / Developmental Biology 403 (2015) 69–79 79

double-strand break repair leads to ovarian aging in mice and humans. Sci.Transl. Med. 5 (172) 172ra21-172ra21.

Tomizawa, S., Nowacka-Woszuk, J., Kelsey, G., 2012. DNA methylation establish-ment during oocyte growth: mechanisms and significance. Int. J. Dev. Biol. 56(10–12), 867–875.

Veeman, M.T., Smith, W.C., 2012. Whole-organ cell shape analysis reveals thedevelopmental basis of ascidian notochord taper. Dev. Biol. 373, 281–289.

Westergaard., C.G., Byskov., A.G., Andersen, C.Y., 2007. Morphometric character-istics of the primordial to primary follicle transition in the human ovary inrelation to age. Hum. Reprod. 22 (5), 2225–2231.

White, Y.A.R., Woods, D.C., Takai, Y., Ishihara, O., Seki, H., Tilly, J.L., 2012. Oocyteformation by mitotically active germ cells purified from ovaries of reproduc-tive-age women. Nat. Med. 18 (3), 413–421.

Zhang, H.H., Zheng, W.W., Shen, Y.Y., Adhikari, D.D., Ueno, H.H., Liu, K.K., 2012.Experimental evidence showing that no mitotically active female germlineprogenitors exist in postnatal mouse ovaries. Proc. Natl. Acad. Sci. USA 109 (31),12580–12585.

Zheng, W., Zhang, H., Gorre, N., Risal, S., Shen, Y., Liu, K., 2014. Two classes ofovarian primordial follicles exhibit distinct developmental dynamics and phy-siological functions. Hum. Mol. Genet. 23 (4), 920–928.