<<

© 2019. Published by The Company of Biologists Ltd | Development (2019) 146, dev180273. doi:10.1242/dev.180273

STEM CELLS AND REGENERATION RESEARCH ARTICLE CDK2 kinase activity is a regulator of male fate Priti Singh1, Ravi K. Patel2, Nathan Palmer3,4, Jennifer K. Grenier1, Darius Paduch5, Philipp Kaldis3,4, Andrew Grimson2 and John C. Schimenti1,*

ABSTRACT About 1 day after birth at postnatal day (P) 1.5, the gonocytes The ability of men to remain fertile throughout their lives depends resume proliferation (Nagano et al., 2000) and complete a process upon establishment of a (SSC) pool from known as the gonocyte-to-spermatogonia transition (GST). The gonocyte progenitors, and thereafter balancing SSC renewal versus GST is not precisely delineated, as there is no clear-cut distinction terminal differentiation. Here, we report that precise regulation of the between gonocytes/prospermatogonia and spermatogonia, although cell cycle is crucial for this balance. Whereas cyclin-dependent marker analysis and single cell sequencing indicate that this kinase 2 (Cdk2) is not necessary for mouse viability or transition begins in late gestation (Law et al., 2019; Pui and Saga, stages prior to meiotic prophase I, mice bearing a 2017). During the GST, some gonocytes establish the permanent deregulated allele (Cdk2Y15S) are severely deficient in pool of SSCs that will seed waves of throughout spermatogonial differentiation. This allele disrupts an inhibitory adult life, whereas others initiate a distinct prepubertal round of phosphorylation site (Tyr15) for the kinase WEE1. Remarkably, spermatogenesis involving several mitotic spermatogonial Cdk2Y15S/Y15S mice possess abnormal clusters of mitotically active divisions, , and postmeiotic development (Bellve et al., SSC-like cells, but these are eventually removed by after 1977). This first round of spermatogonial differentiation is unique failing to differentiate properly. Analyses of lineage markers, germ cell because it originates from neurogenin 3-expressing gonocytes rather proliferation over time, and single cell RNA-seq data revealed delayed than SSC populations (Yoshida et al., 2004, 2006). and defective differentiation of gonocytes into SSCs. Biochemical During adult life, new coordinated waves of spermatogenesis and genetic data demonstrated that Cdk2Y15S is a gain-of-function derive from divisions of an individual Asingle (As) type SSC and its allele causing elevated kinase activity, which underlies these progeny to form Apaired (Apr), and then Aaligned (Aal) type SSCs. Aal differentiation defects. Our results demonstrate that precise SSCs divide without cytokinesis to form clonal chains (Aal4 to regulation of CDK2 kinase activity in male germ cell development is Aal32) linked via their cytoplasm. Further divisions result in A2, A3, crucial for the gonocyte-to-spermatogonia transition and long-term A4, intermediate (In) and B type spermatogonia before ultimately spermatogenic homeostasis. differentiating into preleptotene that enter meiosis. The ‘stemness’ potential of these cells decreases as spermatogonia KEY WORDS: Spermatogonia, Gonocytes, Mouse, Cell cycle chain length increases (reviewed by de Rooij, 2017). The balance between SSC renewal and SSC differentiation is crucial, and failure INTRODUCTION to maintain this balance can cause either insufficient Human and mouse males are capable of reproduction throughout production or exhaustion of the stem cell pool. Genetic or much of their lives owing to a continuously regenerating pool of environmental events that lead to SSC exhaustion, or which spermatogonial stem cells (SSCs) in adult testes. In mice, the compromise the generation or viability of SSCs or their progenitors of SSCs, called primordial germ cells (PGCs), arise as a progenitors, can cause only syndrome (SCOS), a group of ∼45 cells in the of 6- to 6.5-day-old histological phenotype categorizing a subset of patients with non- (Ginsburg et al., 1990). The PGCs migrate to the genital ridges by obstructive azoospermia (NOA). ∼embryonic day (E) 10.5, and proliferate rapidly as the Normal proliferation of cells is dependent on cell cycle regulation. differentiate into either primitive ovaries or testes. These germ cells, Key players in this process are cyclin-dependent kinases (CDKs) now called gonocytes (also called ‘prospermatogonia’ in males), and their activating partner , cyclins, several of which exist reach a population of ∼25,000 by E13.5. Then, female gonocytes in mammals. CDK activity is controlled during the cell cycle in part (‘’) directly enter meiosis, whereas the male gonocytes by the association of CDKs with cyclins. In their activated state, largely cease proliferation for the remainder of gestation (Sasaki and these complexes propel cells through successive stages of the cell Matsui, 2008; Tam and Snow, 1981). cycle, including entry into and through the S and M phases (Satyanarayana and Kaldis, 2009). The transition between active and inactive states is governed by both interaction with CDK-inhibitory proteins (Lim and Kaldis, 2013) and also phosphorylation or 1Cornell University, College of Veterinary Medicine, Department of Biomedical dephosphorylation events at key regulatory sites on CDKs (Cuijpers Sciences, Ithaca, NY 14853, USA. 2Cornell University, Department of Molecular Biology and Genetics, Ithaca, NY 14853, USA. 3Institute of Molecular and Cell and Vertegaal, 2018; Morgan, 1995). Biology (IMCB), Agency for Science, Technology, and Research (A*STAR), Though the activities of cyclins and CDKs have been studied Singapore 138673. 4Department of Biochemistry, National University of Singapore, predominantly in cultured somatic cells and single-celled Singapore 117599, Republic of Singapore. 5Cornell University, Weill Cornell Medicine, Department of Urology, New York, NY 10065, USA . eukaryotes, their roles in the germline have also been investigated (Martinerie et al., 2014; Wolgemuth and Roberts, 2010). Despite its *Author for correspondence ( [email protected]) broad expression in many cell types, Cdk2 is not essential for mouse P.K., 0000-0002-7247-7591; J.C.S., 0000-0002-7294-1876 viability, yet its disruption causes male and female (Berthet et al., 2003; Ortega et al., 2003). Specifically, Cdk2−/−

Received 8 May 2019; Accepted 21 September 2019 meiocytes arrest during the pachytene stage of meiotic prophase I. DEVELOPMENT

1 STEM CELLS AND REGENERATION Development (2019) 146, dev180273. doi:10.1242/dev.180273

This arrest is triggered by defective attachment of telomeres to the cells are present and can divide, but their progeny fail to differentiate nuclear envelope, resulting in failed or incomplete synapsis of and subsequently are lost before entering meiosis. The germ cell homologous chromosomes. In turn, these defects prevent defects are first detectable at P3; GST appears delayed or disrupted homologous recombination repair of meiotic double-strand breaks as determined by analyses of key markers and single cell (sc)RNA- (Viera et al., 2009, 2015). CDK2 is expressed in spermatogonia seq data. We provide evidence that CDK2Y15S-expressing cells (Johnston et al., 2008; Ravnik and Wolgemuth, 1999), but SSCs display altered kinase activity, and that this defect underlies the apparently remain functional because mutant males produce phenotypes observed in such cells. This study highlights the spermatocytes (albeit destined for meiotic arrest) into adulthood. importance of precise regulation of CDK kinase activity in These results suggest that, as in most somatic cells, CDK2 function establishing and maintaining testis homeostasis. is not essential in spermatogonia, but it may provide redundant function in those cells and non-canonical function(s) in meiocytes RESULTS related to recombination in the latter (Berthet et al., 2003; Krasinska Ablation of the Tyr15 inhibitory phosphorylation site in CDK2 et al., 2008). Although a spermatogonia-specific deletion of Cdk1 disrupts gonocyte and spermatogonia differentiation has yet to be described, this kinase is required for metaphase I entry As summarized above, adult Cdk2Y15S/Y15S testes lacked evidence of at the end of the first meiotic prophase (Clement et al., 2015). CDK1 spermatogenesis and were essentially devoid of cells positive for likely acts in concert with the meiosis-specific cyclin A1, which is DDX4 (hereafter MVH, mouse vasa homolog), which is strongly also required at the same stage (Liu et al., 1998). In contrast, expressed in gonocytes and all juvenile germ cells (Toyooka et al., conditional ablation of cyclin B1 (Ccnb1), a CDK1 binding partner, 2000). Our working hypothesis was that most gonocytes blocks proliferation of gonocytes and spermatogonia, but does not differentiated in the initial spermatogenic wave, leaving the adults impact meiosis (Tang et al., 2017). devoid of a renewable SSC pool. To test this hypothesis, we first To identify possible infertility alleles in human populations, we quantified gonocytes in neonatal testes. The number of MVH+ cells modeled a missense variant (SNP rs3087335) altering the Tyr15 in P0 Cdk2Y15S/Y15S testes was no different than in control littermates phosphorylation site of CDK2 in mice (Singh and Schimenti, 2015). (Fig. 1A,B), indicating that the loss of germ cells occurred not Surprisingly, homozygotes for this allele (Cdk2Y15S) caused an during gestation (for example, during PGC expansion), but SCOS-like phenotype. Additionally, Cdk2Y15S heterozygotes during postnatal development. Next, to test the prediction that exhibited age-dependent testis histopathology and reduced sperm all SSCs would be exhausted by adulthood, we performed count, indicating that Cdk2Y15S is a gain-of-function, semidominant, immunohistochemical (IHC) analysis of mutant adult (P180) allele (Singh and Schimenti, 2015). In vitro studies have shown that sections, which lack ongoing spermatogenesis. Tyr15 phosphorylation, typically catalyzed by the WEE1 kinase, Remarkably, Cdk2Y15S/Y15S tubules contained ample numbers of cells negatively regulates CDK activity and, thus, cell cycle progression positive for LIN28, which is expressed in a subset of Type As (Gu et al., 1992; Welburn et al., 2007). We speculated that the spermatogonia, and essentially all Type Apr through Aal spermatogonia Cdk2Y15S allele was hyperactive by virtue of being refractory to (Chakraborty et al., 2014b) (Fig. 1C; Fig. S1B,C), demonstrating that negative regulation by WEE1 (Hughes et al., 2013; Zhao et al., although mutant testes had an SCOS-like appearance, there were indeed 2012), thus driving excessive spermatogonial proliferation and/or undifferentiated A-type spermatogonia present. However, they differentiation over SSC regeneration and maintenance. apparently were not proliferating or differentiating in a normal manner. Here, we report that the apparent SCOS phenotype in Cdk2Y15S/ We next characterized maturation of germ cells using markers of Y15S testes is not due to an absence of germ cells; rather, SSC-like gonocytes, SSCs, and progressively more differentiated spermatogonia.

Fig. 1. Cdk2Y15S/Y15S mice are born with a normal germ cell complement, but exhibit defective spermatogonial differentiation. (A) IHC of newborn testis sections showing germ cells labeled with the germ cell marker MVH. (B) Quantification of the data shown in A. ns, not significant; YS, Cdk2Y15S. (C) IHC on adult testis sections labeled with the spermatogonia marker LIN28 and DNA- binding dye DAPI. Scale bars: 50 μm (A); 100 μm (C). DEVELOPMENT

2 STEM CELLS AND REGENERATION Development (2019) 146, dev180273. doi:10.1242/dev.180273

The transcription factor FOXO1 (forkhead box O1), which in testis henceforth we will refer groups of more than two cells as is expressed only in gonocytes and undifferentiated spermatogonia ‘clusters’). Furthermore, mutants lacked the longer PLZF+LIN28+ (As-Aal) but not more differentiated germ cells, transits from the chains (Aal16-32) that are typical in adult WT testes (Fig. 3A) (Buaas cytoplasm to the nucleus as gonocytes differentiate into et al., 2004; Zheng et al., 2009). This result is consistent with the spermatogonia postnatally (Goertz et al., 2011). At P0, mutant lower overall number of PLZF+ and LIN28+ spermatogonia in testes gonads retained cytoplasmic localization of FOXO1 (Fig. S1A), of young (P5) and old (P90) Cdk2Y15S homozygotes (Fig. S1C). consistent with normal prenatal development of gonocytes. To explore the basis for the defects in spermatogonial However, translocation of FOXO1 to the nucleus, which normally distributions, we examined GFRA1+ progenitor spermatogonia in begins at P3 and is complete by P21 (Goertz et al., 2011), was adolescent (P15, during the first round of spermatogenesis) versus delayed in the mutant. Unlike wild type (WT), in which FOXO1 was adult (P90) tubules, which lack spermatogonial differentiation. localized in the nuclei of all positive cells at P30, >40% of Cdk2Y15S/ Cdk2Y15S/Y15S mutants had about twice as many GFRA1+ cells at Y15S cells exhibited cytoplasmic FOXO1 at this time; this fraction both ages compared with WT (Fig. 3A-C). Strikingly, mutant declined further to ∼10% at P90 (Fig. 2; Fig. S1B). These results tubules contained large clusters of GFRA1+ spermatogonia Y15S/Y15S + suggest that Cdk2 germ cells are delayed in the GST and, (referred to as ‘GFRA1 Acluster>6’) in P15 tubules, which were with the exception of the unique first round of spermatogenesis, are virtually absent in WT (Fig. 3A,D,E). Hypothesizing that these + unable to differentiate properly even after making this transition. GFRA1 Acluster>6 cells might be in an abnormal, delayed state of Studies of other markers by testis IHC confirmed abnormalities in differentiation, we examined them for co-expression of LIN28. the Cdk2Y15S/Y15S germ cell pool. At P3 and P15, there were fewer cells Interestingly, many of the P15 GFRA1+ cells in clusters were also positive for PLZF (zinc finger and BTB domain containing 16; LIN28+ (Fig. S2). Additionally, whereas mutants had ∼2-fold more + + formally ZBTB16) and FOXO1 (Fig. S1B,C), which are expressed in As GFRA1 cells than WT, most were also LIN28 as in WT all undifferentiated spermatogonia (Goertz et al., 2011). The number (Fig. 3F). The clusters of GFRA1+ cells disappeared by P90, yet + of cells positive for LIN28, which is expressed in both undifferentiated mutant tubules had more GFRA1 As and Apr cells compared with (As-Aal) and differentiated (A1-A4) spermatogonia (Chakraborty et al., WT at this age (Fig. 3B-D). The combined data indicate that the 2014a; Gaytan et al., 2013), was also lower in mutants at P3 Cdk2Y15S allele not only causes a delay in the GST, but also impacts + (Fig. S1C). The LIN28 cell shortfall disappeared at P15, possibly the differentiation of progenitor SSCs beyond the Apr reflecting large numbers of differentiated spermatogonia resulting developmental state when GFRA1 should be downregulated in + from the first wave of spermatogenesis, but in adulthood (P90) when chains of LIN28 Aal spermatogonia. the absence of ongoing spermatogenesis in mutants was manifested, LIN28+ cells were again lower in the mutant, as was PLZF (Fig. S1B,C). Regulation of CDK2 activity is crucial for balancing We next performed a series of studies on seminiferous tubule spermatogonia progenitor self-renewal versus whole mounts to determine if there were disruptions to the normal differentiation patterns of Type A spermatogonia subtypes (e.g. As versus Apr Whereas SSC differentiation or maintenance was not obviously −/− versus Aal). A marker for SSCs is GFRA1 [glial cell line-derived impacted in Cdk2 null (Cdk2 ) mice, which demonstrate continued neurotrophic factor family receptor alpha 1, the cell surface receptor rounds of meiotic entry (Berthet et al., 2003; Ortega et al., 2003) (see for glial cell line-derived neurotrophic factor (GDNF)], which mainly also last section of Results and Fig. 7D), Cdk2Y15S heterozygotes labels As (a subset thereof) and Apr spermatogonia. We found that exhibited an age-related decrease in spermatogenesis. This difference + Y15S/Y15S Y15S almost all As and Apr PLZF SSCs from P90 Cdk2 mice also in phenotypes suggests that Cdk2 is a hypermorphic or a gain-of- expressed GFRA1, similar to age-matched WT controls. Mutants also function allele. Given the increase in GFRA1+ spermatogonia in the contained clusters of four to eight PLZF+LIN28+ cells at P90, Cdk2Y15S/Y15S testes (Fig. 3), we hypothesized that Cdk2Y15S either although these clusters did not have the typical appearance of Aal drives abnormal proliferation, and/or it skews these cells towards self- chains (possibly due to disrupted overall tubule architecture; renewal instead of differentiation.

Fig. 2. Cdk2Y15S/Y15S mice have a defective gonocyte-to-spermatogonia transition. (A) Immunolabeling of testis histological sections. Arrows and arrowheads indicate germ cells with cytoplasmic or nuclear FOXO1, respectively. The locations of some seminiferous tubule boundaries are indicated by dashed Y15S/Y15S lines. (B) Percentage of cytoplasmic FOXO1 in P30 and P90 tubule sections. YS, Cdk2 . Scale bar: 50 µm. DEVELOPMENT

3 STEM CELLS AND REGENERATION Development (2019) 146, dev180273. doi:10.1242/dev.180273

Fig. 3. Cdk2Y15S/Y15S seminiferous tubules contain increased undifferentiated spermatogonia. (A,B) Confocal images of P15 seminiferous tubules showing the density of GFRA1+ spermatogonia (A) and of P90 tubules immunolabeled for markers of undifferentiated spermatogonia (B). The outermost two or three optical sections are projected in A and B. Dotted lines indicate the edges of a tubule. Scale bars: 50 μm (A); 200 μm (B). (C,D) Density of total + (C) and As,Apr and Acluster (D) GFRA1 spermatogonia in >8-mm-long tubule segments. n≥3 for each genotype (except n=2 for P90 WT). P-values: a<0.0001, b=0.0001, c=0.0006, d=0.0002, e=0.0039. Data are mean±s.d. (E) Distribution of GFRA1+ subpopulations. cl, cluster.

(F) Quantification of As spermatogonia subtypes. YS, Cdk2Y15S/Y15S. Two or more tubules (6-8 mm length) from three different animals were quantified. Boxes represent minimum and maximum values, and horizontal lines represent median values.

+ To test this, we assayed proliferation of spermatogonia by pulse fewer proliferating LIN28 germ cells, which include As labeling with the DNA analog 5-ethynyl-2′-deoxyuridine (EdU). P2 spermatogonia (Fig. 4F). Collectively, our results support the males were injected with EdU, sacrificed 4 h later, then the testes notion that normal homeostasis of the stem cell niche is disrupted in were immunolabeled for PLZF. In both mutants and WT, >99% of Cdk2Y15S/Y15S mice, causing elevated proliferation of gonocytes and PLZF+ cells were negative for EdU (not shown), consistent with this SSCs without normal differentiation. being the period before gonocytes exit mitotic arrest to begin Despite the evidence for spermatogonial cycling in the absence of establishing the spermatogonial pool (Yang and Oatley, 2014). differentiation, the seminiferous tubules never (up to 20 months of However, in older mutant animals, we noticed severe proliferation age) became replete with undifferentiated cells, a condition that might abnormalities in GFRA1+ and FOXO1+ cells. At P15, there were lead to, or resemble, tumorigenesis. Therefore, we hypothesized that ∼1.6-fold more proliferating (EdU+) GFRA1+ cells in mutant such hyperproliferating progenitor spermatogonia were eliminated by homozygotes than WT, and this disparity persisted through P90 apoptosis. Indeed, there were nearly 6-fold more terminal + (Fig. 4A,C). Furthermore, the fractions of replicating As and Apr deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) GFRA1+ spermatogonia were higher in mutant than WT, and, most seminiferous tubules containing 2- to 4-fold more apoptotic germ cells + dramatically, the Acluster>6 GFRA1 category was unique to the in mutants versus WT at P30 (Fig. S3A-C). Furthermore, double mutant (Fig. 4A,D). Similarly, P90 Cdk2Y15S/Y15S testes, which immunolabeling for FOXO1 and cleaved PARP revealed the presence contained cells with both nuclear (predominantly) and cytoplasmic of spermatogonia undergoing apoptosis in mutants (Fig. S3D). These FOXO1 (nFOXO1+ and cFOXO1+, respectively; Fig. 4B), had combined results suggest that Cdk2Y15S/Y15S undifferentiated more proliferating FOXO1+ cells in both categories (Fig. 4E), in progenitor GFRA1+ spermatogonia enter S phase normally but are aggregate representing ∼50% more in the mutant than WT (22.5% incapable of differentiating; instead, they appear to proliferate, versus 15%, P=0.01). In contrast, P90 mutant testes had ∼40% accumulate, and eventually undergo apoptosis. DEVELOPMENT

4 STEM CELLS AND REGENERATION Development (2019) 146, dev180273. doi:10.1242/dev.180273

Fig. 4. of the CDK2Tyr15 phosphorylation site + impedes differentiation of GFRA1 As spermatogonia. (A) Seminiferous tubule (P15) whole mounts showing abnormally long clusters of GFRA1+ and EdU+ spermatogonia. + Acl,Acluster. (B) DNA replication in FOXO1 germ cells. Arrowheads indicate examples of double-positive cells. FOXO1 is mostly nuclear in WT, and cytoplasmic in mutants (insets). (C) Quantification of the data shown in A (mean±s.d.). (D) Quantification of replicating spermatogonial subtypes (mean±s.d., n>3 for each group). Boxes represent values in the 10-90 percentiles, and horizontal lines represent median values. (E) Replicating FOXO1+ cells at P90. Over 100 and 55 tubules were analyzed from three different animals of each. Error bars represent s.d. (F) Quantification of replicating LIN28+ spermatogonia. Error bars represent s.d. YS, Cdk2Y15S/Y15S. Scale bars: 50 µm (A,B); 25 μm (inset in B).

Single cell transcriptome analysis reveals defects in hypothesized that the neonatal germ cell pool was unable to differentiation of Cdk2Y15S/Y15S gonocytes and SSCs differentiate into SSCs. The germ cell population at birth has substantial functional and To test this at a molecular level, we performed scRNA-seq on molecular heterogeneity (Culty, 2013). Gonocytes in WT P3 testes, unsorted cells from WT and mutant P3 testes. Data were obtained which constitute about half of all germ cells (Ohmura et al., 2004), from WT (5061 cells), Cdk2Y15S/+ (4958 cells) and Cdk2Y15S/Y15S can undergo one of three immediate fates: (1) re-enter the cell cycle, (4403 cells) testes. There was a median of 2239 genes and 5751 (2) migrate towards the basement membrane of the seminiferous mRNA molecules detected per cell. Somatic and germ cells were cords to become SSCs, or (3) differentiate into spermatogonia that evident in the clustering analysis of 14,422 cells from testes seed the first wave of spermatogenesis. Gonocytes decrease from across all genotypes. Following unbiased k-means clustering of 98% to 30% of all germ cells during the first week of life (Ohmura cellsbasedongeneexpressiondifferences,weidentifiedfive et al., 2004), thus lying within the GST interval (although the timing major cell clusters (Fig. 5A). The cell types within each cluster may differ in Swiss outbred mice; Pui and Saga, 2017). Given that were identified by markers diagnostic of particular gonadal Cdk2Y15S mutants are born with a normal number of germ cells lineages (Fig. S4). The percentages and numbers of cells in each (Fig. 1A), and that the first apparent abnormality was a deficit of of the five clusters were as follows: MVH+ germ cells (2.06%, + + + + + PLZF /LIN28 /FOXO1 spermatogonia at P3 (Fig. S1C), we n=299); WT1 Sertoli cells (46.88%, n=6781); CYP11a1 DEVELOPMENT

5 STEM CELLS AND REGENERATION Development (2019) 146, dev180273. doi:10.1242/dev.180273

Fig. 5. See next page for legend.

Leydig cells (1.5%, n=217); MYH11+ myoid cells (22.15%, To characterize potential GST defects, we focused on the MVH+ n=3204); and VCAM1+ peritubular/epithelial cells (27.9%, germ cell population. Interestingly, this group of cells consisted n=3963). apparently of two populations that differed dramatically (>2-fold) in DEVELOPMENT

6 STEM CELLS AND REGENERATION Development (2019) 146, dev180273. doi:10.1242/dev.180273

Fig. 5. Single cell RNA sequencing of P3.5 testes reveals abnormal five subgroups of MVH+ germ cells, designated A-E (Fig. 5B), Y15S/Y15S differentiation of Cdk2 gonocytes. (A) A t-SNE (t-distributed using hierarchical clustering based on the SAVER-imputed stochastic neighbor analysis) plot of 14,422 testicular cells with K-means expression of the most divergently expressed genes (n=734; see clustering. Cell types were classified by expression of key marker genes (e.g. Myh11 for myoid cells). Inset shows MVH+ germ cells (263 in total; WT=106, Materials and Methods) (Fig. S6). As depicted in Fig. 5B, cluster A Cdk2Y15S/Y15S=99, Cdk2Y15S/+=58), color coded by genotype. YS, Cdk2Y15S. was exclusive to the WT sample, whereas D and E were exclusive to Y15S/Y15S (B) Identification of germ cell clusters and distribution by genotype. In panels the Cdk2 sample. Clusters B and C were preferentially B-D, the WT and Cdk2Y15S/Y15S germ cells shown in A were filtered for those that enriched in WT and the Cdk2Y15S/Y15S samples, respectively. had >10,000 UMIs/cell, leaving in an expression matrix of 10,451 genes across Clusters A and B express several well-characterized markers of Y15S/Y15S 141 cells (WT=69; Cdk2 =72). 734 highly variably expressed genes undifferentiated germ cells, such as Gfra1 and Id4 (Fig. 5C). Based were used to identify five distinct clusters, called A-E. (C) Violin plots showing log2-transformed reads per million (RPM) of key genes in cells from each of the on the following distinctions between the two groups, we tentatively five clusters (see color-coded legend) defined in panel B. (D) Pseudotime classified cluster A as SSCs (ASSC) and cluster B as gonocytes trajectory analysis plots, showing transitions from one cell state to another, as (BGono). Cluster A was relatively depleted in cell cycle factors such predicted based on expression of the aforementioned 734 most variable genes. as Ccnb1 and Cenpf (Fig. 5C), consistent with the idea that ‘true’ The cells are colored by genotype or clusters A-E. The black circles represent SSCs would have a lower proliferative index. Furthermore, this branch nodes as predicted by Monacle. (E) RNA velocity plotted in tSNE space. cluster was enriched for Txnip (an inhibitor of glucose transport), Shaded circles represent cells. Arrows indicate their estimated differentiation indicating decreased metabolism consistent with lower trends. The lower left area is enriched for cells moving towards a WT cluster A identity, and the upper right towards mutant (clusters C-E). (F) Schematic proliferation. Finally, to validate the cluster identities, we summary of cell states and transitions. The germ cell clusters identified in panel combined previously published scRNA-seq datasets from flow- B can be classified into three states: normal SSCs (cluster A), intermediate sorted OCT4+ (Liao et al., 2017 preprint) and ID4+ cells (Song et al., (clusters B and C) and mutant (clusters D and E). However, based on 2016), and identified 50 and 100 genes uniquely expressed in pseudotime and RNA velocity analyses, the WT cells in clusters B and C (blue) gonocytes and SSCs, respectively, which we then used as diagnostic are predisposed to differentiate towards normal SSCs (cluster A; green), and the markers for cell identities (Fig. S7A,B; Table S1). Gene set mutant cells towards abnormal gonocytes (clusters D and E; red). Ab. gono, abnormal gonocytes; Gono, gonocytes; YS, Cdk2Y15S/Y15S. enrichment analysis (GSEA) revealed that highly expressed genes in the SSC gene sets were upregulated in cluster A compared with the reference cluster B, whereas highly expressed genes in gonocytes the number of unique transcripts (UMIs) per cell (Fig. S5A). This showed upregulation in cluster B (Fig. S7B). The combined data led broad bimodal distribution was not observed in somatic cell us to conclude that cluster A consists of SSCs, and cluster B consists populations. We considered two potential explanations for this of gonocytes. observation. One is that the population with higher UMIs represents cell To better define the cellular defects in mutant germ cells present doublets (Ziegenhain et al., 2017). However, two algorithms used to at P3, we compared key expression patterns of clusters A and D/E, predict doublets from single cell expression data, DoubletFinder which are the populations most specific to WT and Cdk2Y15S/Y15S, (Fig. S5B) (McGinnis et al., 2019) and DoubletDetection (data not respectively. The following features were characteristic of D/E: (1) shown) (https://github.com/JonathanShor/DoubletDetection), indicated cell cycle signature genes and E2F targets were enriched (Figs 5C,F that the cell barcodes with higher UMIs (>10,000) were no more likely and 6F); (2) SSC genes were expressed at relatively low levels to be doublets than those with lower UMIs. A second explanation is (Fig. 5C); and (3) the PGC/gonocyte marker NANOS3 and that neonatal germ cells are transcriptionally more active than differentiating spermatogonia signatures (Stra8, Lmo1, Uchl1, somatic cells, as are embryonic germ and stem cells (Percharde Dmrt1, Sohlh1, Dnmt3b) were co-expressed and enriched et al., 2017). To distinguish between these hypotheses, we (Fig. 5C,F). In summary, mutant germ cells express an unusual quantified total RNA from equal numbers of Oct4-GFP+ cells combination of cell cycle, differentiation and gonocyte signatures. isolated by fluorescence-activated cell sorting from neonatal testes Importantly, when we repeated these analyses using gene of transgenic mice (Szabó et al., 2002). OCT4 (POU5F1) is a expression data without SAVER-based imputation, our results pluripotency marker and its expression is restricted to were consistent (Fig. S8). undifferentiated, prepubertal gonocytes/prospermatogonia (Ohbo We next performed pseudo-time analysis (Trapnell et al., 2014) et al., 2003; Szabó et al., 2002). This revealed 7-fold, 8.3-fold and on the germ cell cluster transcriptomes to explore the implications 2.4-fold higher amounts of RNA in GFP+ germ cells compared with for developmental states and trajectories. In WT, this analysis − lung, brain and GFP testis cells, respectively (Fig. S5C,D), supported a trajectory path bifurcating from cluster B (WTgono)to supporting the hypothesis that neonatal germ cells have more clusters A (WTSSCs) and C (differentiation-primed gonocytes, or transcripts. We next performed knee-plot analysis to identify a UMI WTdiff-Gono) (Fig. 5D), with the latter being defined by virtue of cut-off for reliably profiled cells, revealing that the barcodes with low retaining both gonocyte and differentiation signatures (Fig. 5C,D). UMI counts fell in a low-quality region (under ‘knee’), indicating that This trajectory path in WT is consistent with the known those cells had poor detection (Fig. S5E) (Macosko et al., 2015). developmental progression in the germline. Interestingly, only Therefore, we removed barcodes with low (≤10,000) UMI counts. ∼8% of mutant germ cells fell into cluster B, and these eventually After quality control and data filtering using Seurat (Butler et al., differentiate into two directions: (1) a small subset towards cluster 2018), we identified 10,451 confidently quantified transcripts from C(YSdiff-gono; ∼12%) and (2) ∼80% towards mutant-specific WT and Cdk2Y15S/Y15S germ cells. Because Cdk2Y15S/+ clusters clusters D+E (Fig. 5D). It is worth noting that trajectory analyses closely overlapped with those from WT, we only used the latter in using non-imputed data gave a different topology. Cluster C, subsequent comparisons with homozygous mutants. instead of branching out as a separate cluster, appeared as a Because only a fraction of all transcripts are detected in scRNA- transient stage on a developmental trajectory differentiating from seq, the resulting expression matrices are sparse. We therefore cluster B to E (Fig. S7C,D). Nevertheless, the analyses indicate a employed single cell analysis via expression recovery (SAVER) profound defect in differentiation of mutant germ cells, and are (Huang et al., 2018), which uses information across all genes and consistent with the idea that mutant gonocytes do not undergo a cells in a dataset, to impute gene expression values. We identified normal GST. DEVELOPMENT

7 STEM CELLS AND REGENERATION Development (2019) 146, dev180273. doi:10.1242/dev.180273

Fig. 6. Downstream transcriptional effects of CDK2Y15S. (A) Heat map showing expression of CDK2 activity signature genes (McCurdy et al., 2017) in indicated Y15S/Y15S clusters of WT and Cdk2 germ cells. In A and E, color key shows log2-transformed RPM expression. (B) GSEA enrichment plots for CDK2 activity signature genes. (C) CDK2 activity scores defined as median of normalized expression of CDK2 target genes per cell, across indicated clusters. (Student’s t-test P-values: a=5.0×10−11; b=4.5×10−12; c=5.7×10−19; d=1.2×10−11). YS, Cdk2Y15S/Y15S. (D) Enrichment analysis for GO-slim ‘biological process’ terms in genes upregulated in clusters A versus E. (E) Heat map showing expression of FOXO1 targets in indicated germ cell clusters and genotypes. (F,G) GSEA enrichment score plots for E2F and FOXO1 target-genes (MSigDb) in clusters A versus E. (H) FOXO1 activity defined as median of z-score normalized expression of FOXO1 target genes per cell (Student’s t-test P-values: e=8.9×10−5; f=6.2×10−5; g=1.5×10−13; h=3.7×10−19). In C and H, boxes represent data points between the first and third quartiles of the distribution, whiskers depict minimum and maximum values, with outliers shown as black dots, and black horizontal lines indicate the median value.

To gain additional perspective on developmental defects in the unspliced (nascent) versus spliced (mature) versions of transcripts to mutant, we performed RNAvelocity analysis of the scRNA-seq data predict the future developmental states of cells (La Manno et al.,

(Fig. 5E). This method examines the expression dynamics of 2018). This analysis supports the conclusion that whereas most WT DEVELOPMENT

8 STEM CELLS AND REGENERATION Development (2019) 146, dev180273. doi:10.1242/dev.180273

Fig. 7. Phenotypic effects of CDK2 Tyr15 and Thr160 phosphorylation during spermatogenesis. (A,B) Images (A) and weights (B) of P120 testes. (C) Sperm counts of age-matched genotypes at P120. (D) Hematoxylin & Eosin-stained testis cross-sections from adult animals. Abnormal degenerating cells, possibly multinucleate, are indicated by the arrowheads. Scale bar: 100 μm. YS, Cdk2Y15S;TA,Cdk2T160A. −/− corresponds to a putative null allele of Cdk2 generated by CRISPR mutagenesis as described in Materials and Methods. The formal name is Cdk2em3Jcs. gonocytes will give rise to SSCs, the Cdk2Y15S/Y15S mutation causes Next, we assayed the ability of CDK2 isolated from WT and nearly all mutant gonocytes (including those in clusters B and C; mutant P10 spleens to phosphorylate a histone H1 substrate (testis Fig. 5E) to transition to an abnormal gonocyte-like state exemplified was not used as a source because of cellularity differences between by clusters D and E (summarized in Fig. 5F). mutant and WT). Consistent with ablation of the Tyr15 inhibitory phosphorylation site and previous reports examining Cdk2T14AY15F CDK2Y15S has altered kinase activity that impacts activity in mouse tissues and mouse embryonic fibroblasts (MEFs) gonocyte fate (Zhao et al., 2012), CDK2 immunoprecipitated from Cdk2Y15S/+ Although we hypothesized that CDK2Y15S is hypermorphic spleens displayed 1.5-fold more kinase activity than that of WT by virtue of lacking the target (Tyr15) of inhibitory (Fig. S10B-D). Counterintuitively, material immunoprecipitated phosphorylation (Singh and Schimenti, 2015), we considered from Cdk2Y15S/Y15S spleens had >5-fold reduced kinase activity the possibility that Ser15 could be phosphorylated by an compared with WT. This may reflect the consequence of excessive unknown kinase to alter CDK2 activity. To test this, we CDK kinase activity, which can be toxic to cell cycle progression in expressed MYC-tagged WT (CDK2-TYR15), mutant (Ser15) a mechanism involving p21 (CDKN1A)-mediated inhibition of and also Phe15 cDNAs in HEK293T cells, performed mass CDK2/cyclin (see Discussion) (Hughes et al., 2013; Szmyd et al., spectrometry (LC-MS/MS) analysis on immunoprecipitates 2019; Zhao et al., 2012). (Fig. S8A), then analyzed the mass:charge (m/z) spectra for As an orthogonal assessment of CDK2 activity in germ cells, we evidence of phosphorylation of these residues. Phosphorylation was compared expression levels of 97 key CDK2 activity signature detected only at the WT CDK2Y15 residue, indicating that serine at genes in the cell clusters defined earlier (Table S2) (McCurdy et al., this position is not a phosphorylatable substrate, at least in cultured 2017). WTSSCs (cluster A) had much lower expression of CDK2 cells (Fig. S9B). activity signature genes compared with all other clusters, including DEVELOPMENT

9 STEM CELLS AND REGENERATION Development (2019) 146, dev180273. doi:10.1242/dev.180273

WT cells in cluster B and all Cdk2Y15S clusters (Fig. 6A). Cdk2Y15S allele is a result of altered kinase activity from abolition of a Furthermore, CDK2 kinase activity, as inferred by the median of WEE1 phosphorylation site. As a consequence of this defective normalized expression of CDK2 activity signature genes per cell negative regulation, this allele acts semidominantly. across clusters A, B, D and E, was lowest in WTSSCs and highest in mutant-specific clusters D and E (Fig. 6B,C). GSEA analysis DISCUSSION indicated upregulation of CDK2 activity signature genes in clusters The longevity of spermatogenesis in both mice and humans depends B and E compared with cluster A, suggesting that (1) cluster E is more upon proper establishment and homeostasis of the SSC pool. A key like cluster B with respect to CDK2 kinase activity, and (2) that cells in event in male germline establishment is the seeding of prepubertal cluster E have a higher propensity to cycle than those in cluster A testes with a quiescent (G1-arrested) population of gonocytes/ (Fig. 6C). Interestingly, in support of this observation, genes prospermatogonia. About 2-3 days after birth, these cells re-enter the upregulated in cluster E compared with cluster A (q≤0.05; FC≥0.2) cell cycle to expand and differentiate into SSCs that establish a also exhibited enrichment of cell cycle-related gene ontology (GO) permanent, renewable pool of cells that can initiate spermatogenesis terms (Fig. 6D). Overall, these data indicate that removing a layer of throughout life. Once established, there must be a fine balance of SSC negative regulation of CDK2 activity disrupts the normal self-renewal versus differentiation to maintain homeostasis and , differentiation of gonocytes and SSCs into downstream cell types. but this is not well-understood and is the subject of intense research. The molecular basis for this developmental disruption may stem There is some debate as to the character of SSCs and their from alteration of two known functions of active CDK2: (1) behavior with respect to cycling activity (Huckins, 1971b; Sharma phosphorylation of the RB1 transcriptional repressor to inactivate et al., 2019). In some tissues, the ability to continuously produce RB1, enabling timely induction of the E2F transcription factors differentiated cells depends upon proper maintenance of a relatively (TFs) to drive transition to S phase (Morris et al., 2000); and (2) infrequently cycling population of stem cells, as in the case of the inhibition of cytoplasmic-to-nuclear localization of the FOXO1 TF hematopoietic system. Overproliferation of hematopoietic stem cells (nFOXO1 is essential for SSC maintenance) (Huang et al., 2006; (HSCs) caused by deregulated cell cycle control can lead to their Goertz et al., 2011). If CDK2 is indeed controlling the gene exhaustion or transformation (Pietras et al., 2011). It has been regulatory network via E2F and FOXO1, then levels or activity of hypothesized that there is a slow-cycling population of SSCs that their downstream targets might be affected in Cdk2Y15S/Y15S cells. maintains the germline (Huckins, 1971a; Sharma et al., 2019). Indeed, GSEA analysis revealed that FOXO1 targets are However, there is also evidence for rapid turnover of SSCs (Klein upregulated and highest in cluster A versus cluster E, where et al., 2010) and for the ability of chains of differentiating expression is lowest [84 genes; normalized enrichment score (NES): spermatogonia (Apr and Aal) to fragment and de-differentiate to 3.27; family-wise error rate (FWER), p<0.001] (Fig. 6E,G,H). In become As SSCs (Hara et al., 2014; Nakagawa et al., 2010). contrast, E2F target genes are upregulated in cluster E (Fig. 6F; 66 Regardless of various models proposed for the identity and behavior genes; NES: −2.7; FWER, p<0.001). These results indicate that of ‘true’ SSCs (reviewed by de Rooij, 2017), proper regulation of FOXO1 activity is significantly greater in cluster A than E the cell cycle is essential. This is underscored by our studies, which (Student’s t-test, P=3.7×10−19) (Fig. 6H). implicate CDK activity regulation as being crucial for the GST and SSC renewal versus differentiation. Indeed, it is well-recognized Phosphorylation states at Tyr15 and Thr160 residues control that, in general, cell cycle progression and differentiation occur in a CDK2 activity in male germ cells mutually exclusive manner (Dalton, 2015). For example, a decrease If disrupting the ability to negatively regulate CDK2 in adult in CDK activity stimulates differentiation of neural stem cells (Lim GFRA1+ SSCs favors cell cycle progression over differentiation, and Kaldis, 2012), whereas elevated CDK activity decreases then a compensatory alteration that dampens or eliminates CDK2 differentiation (Lange et al., 2009) and stimulates expansion of activity might counteract the aberrant phenotype of Cdk2Y15S neural stem cells in the adult mouse brain (Artegiani et al., 2011). mutant spermatogonia. To test this, we mutated threonine 160 to Phenotypes of certain mouse mutants have provided some insight alanine (T160A) in the Cdk2Y15S allele. Phosphorylation of Thr160 into how regulation of cell cycle impacts spermatogonial is required for activation of CDK2 (Gu et al., 1992; Kaldis, 1999). maintenance and proliferation. Conditional germline knockout of Cdk2T160A is a ‘kinase dead’ allele that causes infertility in both the Rb (Rb1) tumor suppressor, a negative cell cycle regulator, sexes, albeit with less severe meiotic phenotypes than in Cdk2−/− abolished the ability of SSCs to self-renew, causing the entire germ cell mice (Chauhan et al., 2016). Mice homozygous for the doubly pool to undergo a single round of spermatogenesis (Hu et al., 2013). mutated Cdk2 allele (Cdk2Y15S,T160A, abbreviated as Cdk2YS-TA) Cell cycle progression in normal cells requires inactivation of Rb by exhibited three striking phenotypic differences compared with CDK/cyclin-mediated phosphorylation (including by CDK2/cyclinE), Cdk2Y15S. First, whereas Cdk2Y15S adult (P120) heterozygotes had thus allowing expression of genes regulated by E2F transcription small testes and a markedly reduced sperm count, as previously factors (Rubin, 2013). Moreover, ablation of Plzf, which negatively reported (Singh and Schimenti, 2015), Cdk2YS-TA heterozygotes were regulates the cell cycle by both inhibiting key regulators (McConnell indistinguishable from WT in both respects (Fig. 7A-C). Second, as et al., 2003; Yeyati et al., 1999) and also the self-renewal signal of with null but not Cdk2Y15S/Y15S mice, Cdk2YS-TA homozygous females GDNF (Hobbs et al., 2010), causes a less severe phenotype than Rb were sterile (n=3 in matings to WT). Third, although Cdk2YS-TA/YS-TA deficiency. Plzf−/− males are infertile due to a defect in SSC adult males were severely hypogonadal (Fig. 7A,B) and azoospermic, maintenance that leads to progressive germ cell loss and SCOS similar to Cdk2Y15S/Y15S, they exhibited differentiating spermatogonia (Buaas et al., 2004; Costoya et al., 2004). Thus, a cell cycle-centric and meiocytes, which were completely missing from age-matched interpretation of these phenotypes is that unrestrained cycling (as in Cdk2Y15S homozygotes, indicating that the T160A alteration rescued Rb−/−) causes efficient differentiation of all SSCs, but a moderate loss the spermatogonial differentiation defect (Fig. 7D). At these levels of of cell cycle control (as in Plzf−/−) increases the propensity of SSCs to analysis, the Cdk2YS-TA allele resembles the null phenotype (Viera differentiate rather than self-renew. In the context of this model, et al., 2009; Chauhan et al., 2016). Collectively, our results imply that CDK2Y15S might have a lower impact on cell cycle control than Rb and the failed spermatogonial differentiation phenotype caused by the Plzf mutants, possibly mediated by abnormal but partial inactivation of DEVELOPMENT

10 STEM CELLS AND REGENERATION Development (2019) 146, dev180273. doi:10.1242/dev.180273

Rb. The result is abnormal SSC proliferation but only partial in Cdk2Y15S/Y15S testes, we propose a model (Fig. 8) in which CDK2- differentiation, ultimately leading to loss of the aberrant cells associated kinase activity must be tightly regulated during specific (clusters D and E) before meiotic entry. The defective differentiation stages of the cell cycle in SSCs, otherwise those cells attempting to may explain the observation that P90 Cdk2Y15S/Y15S mutants had differentiate will eventually die from cell cycle dysregulation. increased proliferating GFRA1+ cells but fewer proliferating cells A caveat to the model is that it is based on cell cycle studies in expressing LIN28, which normally marks a subset of As ‘true’ SSCs. homogeneous cell cultures, as opposed to germ cells developing in a MEFs derived from mice in which both the Thr14 and Tyr15 complex milieu that provides a stem cell niche containing several negative phosphoregulatory sites were mutated exhibited somatic cell types (Hofmann, 2008; Oatley and Brinster, 2012). In accelerated entry into S phase (Zhao et al., 2012). Interestingly, particular, the Sertoli cells, which associate intimately with the germ though no data were presented, the only significant defect reported cells, provide key instructive signals including to in these mice was male infertility due to an apparent absence of induce differentiation (Endo et al., 2015). Furthermore, along with germ cells in homozygotes and also in heterozygotes in one strain endothelial cells, Sertoli cells produce GDNF (which binds background. In that regard, this model appears to resemble our GFRA1), which is essential for spermatogonial maintenance Cdk2Y15S mouse. This might indicate that the Thr14 residue is (Bhang et al., 2018; Meng et al., 2000). It is possible that the redundant or not important for negative regulation of SSCs. phenotypes of Cdk2Y15S mice may not be a manifestation of germ Counterintuitively, although we observed elevated CDK2 kinase cell-intrinsic defects exclusively, but rather to defects in one or more activity in spleens from heterozygous mice, the activity was greatly somatic cell types in addition to those of germ cells. Disruption to reduced in homozygous spleens. However, CDK2 kinase over- individual components of the testis niche could impact the activation has been previously shown to be deleterious to cell cycle transcriptome and behavior of germ cells, and vice versa. progression (Hughes et al., 2013). We postulate that, at least in spleen, Although there were differences between mutant and WT Sertoli cell-autonomous regulation maintains CDK2 activity within an cells transcriptomes, EdU labeling of P30 mice revealed no appropriate range. Mutation of both negative phosphoregulatory sites evidence for active DNA replication in mutant or WT SOX9+ in CDK2 (Thr14 and Tyr15) in human Hct116 cell lines caused Sertoli cells (data not shown), suggesting that CDK2Y15S affects the premature S-phase progression, DNA-damage accumulation, and cell cycle of germ cells (Fig. 4) rather than Sertoli cells. Ultimately, genomic instability leading to S-phase arrest (Hughes et al., 2013). In these issues of potential niche defects could be addressed via germ these cells, degradation of cyclin E was found to be increased and this cell transplantation or cell type-specific ablation experiments. could be relieved by p21 depletion, suggesting the presence of Although there are that reduce the number of perinatal cellular feedback loops to attempt to reduce the levels of CDK2 gonocytes (typically stemming from PGC defects) (Hamer and de activity upon premature activation (Hughes et al., 2013). In Rooij, 2018), the Cdk2Y15S phenotype is unique in that the GST is consideration of the aforementioned data of others, and our data defective. Nevertheless, studies of other mutants give insight into showing elevated replicative activity and apoptosis of GFRA1+ cells how perturbations to cell cycle regulation impact gonocytes.

Fig. 8. Model for regulation of SSC differentiation by CDK2 phosphorylation. DEVELOPMENT

11 STEM CELLS AND REGENERATION Development (2019) 146, dev180273. doi:10.1242/dev.180273

Cdk2Y15S/Y15S germ cells fail to relocalize FOXO1 from the B6(Cg)-Tyrc-2J/J] using the reagent concentrations listed in Table S1. cytoplasm to the nucleus, a hallmark of the GST that normally Edited founders were identified either by subcloning followed by Sanger occurs between P3 and P21 (Goertz et al., 2011; Kang et al., 2016; sequencing using the primers and annealing temperatures listed in Table S1. For generating the Cdk2Y15S-T160A allele, Cdk2Y15S/Y15S females were used as Pui and Saga, 2017). This cytoplasmic localization occurs as a result Cdk2Y15S of CDK2-dependent phosphorylation (inactivation) of FOXO1 donors; thus, the alteration was not re-introduced by CRISPR. The Cdk2−/− allele contained a 2-nucleotide deletion in exon 1, (Huang et al., 2006), which is essential for SSC maintenance and leading to a premature termination codon and a predicted truncated CDK2 differentiation (Goertz et al., 2011). As FOXO1 nuclear localization of 31 amino acids. is disrupted in Cdk2Y15S mutants, we conclude that proper cell cycle regulation and/or CDK2 kinase activity is a pre-requisite for the Testes histology and immunohistochemistry GST, and thus lies upstream in the developmental evolution/ Glis3 For histological analyses, testes were fixed for 24 h at room temperature progression. Mice lacking the transcription factor partially (RT) in Bouin’s solution, paraffin-embedded, sectioned at 7 μm, and then Cdk2Y15S resemble mutants; they lack FOXO1 nuclear localization in stained with Hematoxylin & Eosin. For IHC, testes were fixed in 4% neonatal gonads and fail to undergo a normal GST, as indicated by paraformaldehyde for ∼24 h, paraffin-embedded, sectioned at 7 μm, and decreased expression of genes associated with the permanent pool of deparaffinized. Antigen retrieval for different antibodies was performed as undifferentiated spermatogonia (Kang et al., 2016). However, it is indicated in Table S4. Sections were blocked in PBS containing 5% goat unknown whether GLIS3 regulates these genes directly or indirectly, serum for 1 h at RT, followed by incubation with primary antibodies for 12 h so its relationship to CDK2 in the developmental hierarchy is unclear. at 4°C and detection by the secondary antibody (see Table S4 for antibody Interestingly, the first round of spermatogenesis in Cdk2Y15S/Y15S details). mice occurs normally, although meiosis is still interrupted, as in null mice. This first wave of spermatogenesis is claimed to arise directly Whole-mount seminiferous tubule staining from a subset of gonocytes that do not express neurogenin 3 Seminiferous tubule whole mounts were prepared as described previously (Yoshida et al., 2006). We can conclude that differentiation of this (Savitt et al., 2012), with minor modifications. Briefly, seminiferous tubules subtype of gonocytes neither requires CDK2 nor is affected by its were manually isolated in PBS and interstitial tissue was washed thoroughly abnormal activity in Cdk2Y15S mutants. This underscores the caution followed by fixation in 2% paraformaldehyde for 2 h. After extensive needed when extrapolating data from cultured cells or from somatic washing, tubules were processed for detection of progenitor spermatogonial markers and EdU labeling. To detect ZBTB16, FOXO1, cPARP1 and cells (such as spleen) to germ cells, and even from one germ cell LIN28, tubules were blocked in PBSS buffer (5% goat serum, 0.5% Triton type to another. Y15S X-100 in 1× PBS) for 2 h at RT following primary antibody incubation for These investigations into the Cdk2 mouse stemmed from a 12-18 h at 4°C in the same buffer. Tubules were washed at room temperature project to investigate the genetic basis of human infertility, and twice for 15 min, five times for 1 h in PBSS, and then incubated overnight at involves modeling human SNPs in mice to determine possible 4°C with secondary antibodies. To detect GFRA1, the blocking and all pathogenicity (Singh and Schimenti, 2015). A lesson learned from incubations and washing steps were carried out exclusively in PBS-AT (1% this and other alleles is that very few predicted deleterious variants bovine serum albumin, 0.1% Triton X-100 in 1× PBS) buffer. Briefly, are nulls, and that extensive phenotyping is required to understand isolated tubules were blocked for 2 h and incubated in goat anti-GFRA1 their impact. Generally speaking, unless this mutation is entirely antibody diluted in PBS-AT buffer. This was followed by five subsequent unusual in its effects, it raises the possibility that mice and people washes (30 min each) and detection by secondary antibodies. When performing co-immunolabeling of GFRA1 with other spermatogonial that present with non-obstructive azoospermia, and which have markers, ZBTB16, FOXO1, cPARP1 and LIN28 were always detected prior histopathology superficially resembling SCOS, may in fact have an to GFRA1. occult pool of SSCs that have lost the ability to differentiate, but For EdU incorporation, mice were injected intraperitoneally with 100 mg/ might be stimulated to do so after appropriate intervention. kg EdU (PY7562, Berry & Associates) before harvesting and processing Additionally, as an example of an autosomal semi-dominant male testes 4 h later for whole-mount staining, or fixed in 4% paraformaldehyde infertility allele, it emphasizes the importance of considering for IHC. Following antibody staining, freshly prepared Click reaction monoallelic alterations when attempting to identify genetic causes cocktail [10 mM (+)-sodium-L-ascorbate, 0.1 mM 6-caboxyfluorescein- of an individual patient’s infertility. TEG azide (FF 6110, Berry & Associates) or Alexa Fluor 594 Azide (A10270, Invitrogen) and 2 mM copper (II) sulfate in water] was added into whole-mount tubules or testes cross-sections and removed quickly after MATERIALS AND METHODS 45 s. Excess EdU was removed by extensive washing (2×1 h and 2×12 h at Mouse strains and breeding 4°C) using PBS-AT buffer. Following washing, tubules were mounted in Mice used were on a mixed genetic background [FVB/NJ and B6(Cg)- Vectashield (Vector Laboratories). Click reaction was always performed Tyrc-2J/J]. Experiments with the animals were performed under a protocol following antibody detection. In each animal, we counted all As,Apr and Aal (2004-0038) approved by Cornell’s Animal Care and Use Committee. For GFRA1+ cells positive or negative for EdU seminiferous tubules at least female fertility tests, 8- to 10-week-old WT or mutant homozygote females >8 mm in length. were mated with age-matched B6(Cg)-Tyrc-2J/J males. The Cdk2tm1Kald, Cdk2T160A Cdk2Y15S and alleles have been described previously (Berthet Imaging et al., 2003; Chauhan et al., 2016; Singh and Schimenti, 2015). Slide preparations were scanned, and tiled images of testes cross-sections or seminiferous tubule whole mounts were acquired using a laser scanning Production of CRISPR/Cas9-edited mice confocal microscope (u880, Carl Zeiss) using Plan Apo 40× water The Cdk2Y15S-T160A (formally Cdk2em2Jcs) and Cdk2−/− (formally immersion objective (1.1 NA) and Zen black software. Initial laser power Cdk2em3Jcs) alleles were generated using CRISPR/Cas9 editing, adjustment was performed to avoid saturation of signal: argon laser, 488 nm; essentially as described previously (Singh et al., 2015; Varshney et al., blue-diode, 405 nm; DPSS laser, 561 nm; HeNe, 633. Additional images of 2015). sgRNAs and ssODNs are listed in Table S1. Briefly, in vitro testes cross-sections were obtained through an Olympus BX51 microscope transcription of sgRNA was performed using a MEGAshortscript T7 with objectives 100×/1.35 NA infinity/0.17 or objective or 10×/0.3 NA, Transcription Kit (Ambion, AM1354), and ssODN were obtained from IDT. respectively, using Olympus Cell Sens software (Olympus). Following sgRNA, ssODN, Cas9 protein and mRNA (Addgene plasmid #44758) were identical background adjustments for all images, cropping, color and co-microinjected into zygotes [F1 hybrids between strains FVB/NJ and contrast adjustments were made with Adobe Photoshop CC 2017. DEVELOPMENT

12 STEM CELLS AND REGENERATION Development (2019) 146, dev180273. doi:10.1242/dev.180273

In vitro kinase assay using ‘FT mass analyzer’ in MS scan to select precursor ions followed by In vitro kinase assays were performed as described elsewhere (Lim et al., 3s ‘Top Speed’ data-dependent CID ion trap MS/MS scans at 1.6 m/z 2017). Briefly, 1 µg of anti CDK2 antibody was added to 0.5 µg of protein quadrupole isolation for precursor peptides with multiple charged ions lysate and rotated overnight (∼16 h) at 4°C. Twelve microliters of protein A above a threshold ion count of 10,000 and normalized collision energy of beads was added to each sample and left to mix for a further 2 h. Prior to 30%. MS survey scans at a resolving power of 120,000 (fwhm at m/z 200), their use, these beads were washed twice previously using 1 ml of EBN for the mass range of m/z 375-1575. Dynamic exclusion parameters were buffer (80 mM β-glycerophosphate, 15 mM MgCl2, 20 mM EGTA, set at 30 s of exclusion duration with ±10 ppm exclusion mass width. All adjusted to pH 7.3 with KOH; followed by adding 150 mM NaCl, 0.5% data were acquired using Xcalibur 3.0 operation software (Thermo Fisher NP40 and 1× protease inhibitor cocktail). After binding, the beads were Scientific). spun down and washed with EBN buffer three times. Pellets were re- suspended in 14 µl EBN buffer (containing protease inhibitors: cOmplete, Mass spectrometry data analysis Mini, 11836153001; Roche) followed by adding 6 µl of histone H1 (260 ng/μl) The DDA raw files for CID MS/MS were subjected to database searches and 9 μl of kinase assay buffer (15 mM EGTA, 25 mM NaF, 250 mM sodium using Proteome Discoverer (PD) 2.2 software (Thermo Fisher Scientific) with the Sequest HT algorithm. All three raw MS files for three samples β-glycerophosphate, 5 mM DTT, 20 mM MgCl2,21µMATP).Following 20 min incubation at room temperature, 1 µl of ATP [γ-32P] containing a were used for the database search. The PD 2.2 processing workflow specific radioactivity of 5 µCi (Perkin Elmer, BLU502A) was added to containing an additional node of Minora Feature Detector for precursor ion- each sample and left for 30 min at 30°C. Then 6× SDS sample buffer [2 M based quantification was used for protein and post-translational β-mercaptoethanol, 0.375 M Tris (pH 6.8), 12% SDS, 60% glycerol, 0.6 M modifications identification. The database search was conducted against a ∼ DTT, 0.06% Bromophenol Blue] was added to a final concentration of 1× and mouse database containing 20,153 entries downloaded from NCBI each sample was boiled at 95°C for 5 min. Next, 12 µl of each sample were database on 12 January, 2018, plus some common contaminants (246 analyzed for Coomassie-stained histone H1 protein bands and phosphosignal entries). Two missed trypsin sites were allowed. The peptide on a phosphor screen cassette for 6-24 h. Phosphosignal was quantified using a precursor tolerance was set to 10 ppm and fragment ion tolerance was set to FLA7000 phosphorimager (Fujifilm). 0.6 Da. Variable modification of methionine oxidation, deamidation of asparagines/glutamine, phosphorylation of serine, threonine and tyrosine, Western blotting and fixed modification of cysteine carbamidomethylation, were set for the Protein was extracted using EBN buffer (see above) with protease inhibitors. database search. Only high-confidence peptides defined by Sequest HT with Samples were boiled at 95°C for 5 min and were separated by SDS/PAGE a 1% FDR by Percolator were considered for the peptide identification. The (12% acrylamide). Separated proteins were electrotransferred to 0.2 µM final protein IDs contained protein groups that were filtered with at least two nitrocellulose (Bio-Rad, 1620112) or PVDF membrane (Immobilon-P peptides per protein. membrane, IPVH00010, EMD Millipore), and then blocked in 5% nonfat milk for 45 min at RT. Membrane was washed 3×10 min in TBST (0.14 M Sperm counting NaCl, 15 mM KCl, 25 mM Tris Base, 1% Tween20) at room temperature Sperm counting was performed as described previously (Singh and followed by ∼16 h incubation with primary antibody, washing, and ∼1h Schimenti, 2015). incubation with HRP-conjugated secondary antibody (as stated in Table S4) for 45 min at room temperature. Signal was detected using Luminata scRNA seq sample preparation Classico Western HRP substrate (WBLUC0100; EMD Millipore). At P3.5 testes were decapsulated in HBSS (Mediatech) and digested with Densiometric analysis of western blot bands was performed using 0.642 ml of Trypsin/EDTA (0.25%, Invitrogen) and 0.071 ml DNase I Fujifilm Multi Gauge software Ver. 3.1. (1 mg/ml in HBSS, Sigma-Aldrich) at 37°C for 3-5 min. Digestion was stopped by adding 0.5 ml of media (HBSS+10% FBS), the cells were Site-directed mutagenesis filtered through a 70-µm cell strainer and re-suspended in HBSS with 0.04% ′ Site-directed mutagenesis for obtaining CDK2-Y15F and CDK2-Y15S FBS. Single cell 3 RNA-seq sequencing libraries for Illumina were cDNA was accomplished using the QuikChange Lightning Site-Directed constructed using a 10x Genomics Chromium instrument, using the ′ ’ Mutagenesis Kit (Agilent, 210518) using the primer sets listed in Table S3. Chromium Single Cell 3 Reagent kits (v2) following the manufacturer s All mutations were confirmed by Sanger sequencing. protocols. The final libraries were quantified by digital PCR and sequenced on Illumina sequencers, using an Illumina NextSeq500/550 75 bp CDK2 overexpression and immunoprecipitation (26 bp+8 bp index read+58 bp). The target number of cells captured from The (Myc-DDK-tagged)-CDK2 cDNA was obtained from Origene the input, single cell suspension was 8700. The data were demultiplexed and (RC200494). Y15F and Y15F mutant cDNA were generated as described aligned to the reference genome using the 10x Genomics Cellranger above and transfected into HEK293T cells (ATCC; CRL-3216) using software (v2.2) and visualized using the 10x Genomics Loupe Cell Browser TransIT-LT1 Transfection Reagent (Mirus, MIR2305) following the software (v2.0) packages. manufacturer’s instructions. Lysates were prepared from harvested cells in lysis buffer (50 mM Tris pH7.5, 150 mM NaCl, 0.5% Triton X-100, 5 mM scRNA-seq data analysis EDTA) containing phosphatase (Thermo Fisher Scientific, 78428) and The raw scRNA-seq data were analyzed using cell ranger from the 10x protease inhibitors (Roche, 04693159001). 400 µg of each lysate was platform to generate a matrix of raw read counts, which was further analyzed immunoprecipitated using the c-Myc-Tag IP/Co-IP Kit (Pierce, 23620). in R using Seurat (Butler et al., 2018; Satija et al., 2015) and Monocle (Qiu Y15S/Y15S Proteins were separated on 12% polyacrylamide gel by SDS-PAGE and et al., 2017). A cluster of germ cells from WT and Cdk2 were CDK2 protein was visualized by western blot analysis. separated from the rest of the testis cells based on Ddx4 gene expression. We then isolated cell barcodes that had more than 10,000 UMIs per cell (see Protein identification by nanoLC-ESI-MS/MS analysis Results). Raw read counts for these selected germ cells were further filtered In-gel trypsin digestion of immunoprecipitated CDK2 protein was to keep only those genes that were detected in at least 10% of the cells. This performed as described previously (Yang et al., 2007). The tryptic resulted in an expression matrix of 10,451 genes across 141 cells (WT=69; digests were subjected to nanoLC-ESI-MS/MS analysis on an Orbitrap Cdk2Y15S/Y15S=72). Because of the high rate of drop-outs in mRNA-capture Fusion Tribrid (Thermo Fisher Scientific) mass spectrometer equipped in scRNA-seq approaches, the expression values for mid- and low- with a nanospray Flex Ion Source, and a Dionex UltiMate3000RSLCnano expressed genes are often unreliable due to missing information. Hence, we system (Thermo Fisher Scientific) following a protocol described recovered expression for each gene using SAVER (Huang et al., 2018), an previously (Thomas et al., 2017; Yang et al., 2018). The column was approach that estimates gene expression from UMI-based scRNAseq data re-equilibrated with 0.1% formic acid for 23 min prior to the next run. For using information across all genes and cells. This estimated gene expression data-dependent acquisition (DDA) analysis, the instrument was operated was used for further analysis. The most variable genes across all cells were DEVELOPMENT

13 STEM CELLS AND REGENERATION Development (2019) 146, dev180273. doi:10.1242/dev.180273 identified using principal component analysis (PCA). Specifically, PCA was Chakraborty, P., Buaas, F. W., Sharma, M., Snyder, E., de Rooij, D. G. and performed using log-transformed counts-per-million (CPM) expression Braun, R. E. (2014a). LIN28A marks the spermatogonial progenitor population values. The top 300 genes with highest absolute loading for PC1, top 300 and regulates its cyclic expansion. Stem Cells 32, 860-873. doi:10.1002/stem.1584 Chakraborty, P., William Buaas, F., Sharma, M., Smith, B. E., Greenlee, A. R., genes for PC2 and top 300 genes for PC3 were chosen, which resulted in 744 Eacker, S. M. and Braun, R. E. (2014b). Androgen-dependent sertoli cell tight unique genes. Furthermore, this list of variable genes was filtered to remove junction remodeling is mediated by multiple tight junction components. Mol. mitochondrial genes (n=10). The 141 germ cells were clustered into five Endocrinol. 28, 1055-1072. doi:10.1210/me.2013-1134 groups using the ‘Ward.D2’ method based on expression estimates for the 734 Chauhan, S., Diril, M. K., Lee, J. H. S., Bisteau, X., Manoharan, V., Adhikari, D., highly variable genes. The pseudotime and trajectory analyses were performed Ratnacaram, C. K., Janela, B., Noffke, J., Ginhoux, F. et al. (2016). Cdk2 based on the expression of the 734 most variable genes using Monocle R code. catalytic activity is essential for meiotic cell division in vivo. Biochem. J. 473, RNAvelocity analysis (La Manno et al., 2018) was performed using Velocyto, 2783-2798. doi:10.1042/BCJ20160607 Clement, T. M., Inselman, A. L., Goulding, E. H., Willis, W. D. and Eddy, E. M. available at velocyto.org . GSEA (Subramanian et al., 2005) was performed on (2015). Disrupting cyclin dependent kinase 1 in spermatocytes causes late the web server software.broadinstitute.org/gsea/index.jsp. meiotic arrest and infertility in mice. Biol. Reprod. 93, 137. doi:10.1095/biolreprod. 115.134940 Statistics Costoya, J. A., Hobbs, R. M., Barna, M., Cattoretti, G., Manova, K., Sukhwani, P-values were calculated from unpaired Student’s t-tests for all IHC and M., Orwig, K. E., Wolgemuth, D. J. and Pandolfi, P. P. (2004). Essential role of Plzf in maintenance of spermatogonial stem cells. Nat. Genet. 36, 653-659. whole-mount experiments. doi:10.1038/ng1367 Cuijpers, S. A. G. and Vertegaal, A. C. O. (2018). Guiding mitotic progression by Acknowledgements crosstalk between post-translational modifications. Trends Biochem. Sci. 43, ’ The authors would like to thank R. Munroe and C. Abratte of Cornell s transgenic 251-268. doi:10.1016/j.tibs.2018.02.004 T160A Y15S facility for generating the Cdk2 allele alone and in cis to the Cdk2 change. Culty, M. (2013). Gonocytes, from the fifties to the present: is there a reason to Authors also thank the Proteomics Facility of Cornell University for providing the change the name? Biol. Reprod. 89, 46. doi:10.1095/biolreprod.113.110544 mass spectrometry data and NIH SIG 1S10 OD017992-01 grant support for the Dalton, S. (2015). Linking the cell cycle to cell fate decisions. Trends Cell Biol. 25, Orbitrap Fusion mass spectrometer. Confocal imaging data was acquired in the 592-600. doi:10.1016/j.tcb.2015.07.007 Cornell BRC-Imaging Facility using the NIH-funded (S10OD018516) Zeiss LSM880 de Rooij, D. G. (2017). The nature and dynamics of spermatogonial stem cells. confocal/multiphoton microscope (u880). Development 144, 3022-3030. doi:10.1242/dev.146571 Endo, T., Romer, K. A., Anderson, E. L., Baltus, A. E., de Rooij, D. G. and Page, Competing interests D. C. (2015). Periodic retinoic acid-STRA8 signaling intersects with periodic germ- The authors declare no competing or financial interests. cell competencies to regulate spermatogenesis. Proc. Natl. Acad. Sci. USA 112, E2347-E2356. doi:10.1073/pnas.1505683112 Author contributions Gaytan, F., Sangiao-Alvarellos, S., Manfredi-Lozano, M., Garcıa-Galiano,́ D., Conceptualization: P.S., D.P., J.C.S.; Methodology: N.P., P.K., A.G.; Formal analysis: Ruiz-Pino, F., Romero-Ruiz, A., León, S., Morales, C., Cordido, F., Pinilla, L. P.S., R.K.P., N.P., J.K.G.; Investigation: P.S., N.P., D.P.; Writing - original draft: P.S., et al. (2013). Distinct expression patterns predict differential roles of the miRNA- J.K.G., J.C.S.; Writing - review & editing: R.K.P., J.C.S., P.K., A.G.; Supervision: P.K., binding proteins, Lin28 and Lin28b, in the mouse testis: studies during postnatal A.G., J.C.S.; Project administration: J.C.S.; Funding acquisition: J.C.S. development and in a model of hypogonadotropic hypogonadism. Endocrinology 154, 1321-1336. doi:10.1210/en.2012-1745 Ginsburg, M., Snow, M. H. and McLaren, A. (1990). Primordial germ cells in the Funding mouse embryo during gastrulation. Development 110, 521-528. doi:10.1016/ This research was funded by National Institutes of Health grants (R01 HD082568 to 0168-9525(91)90008-e J.C.S. and P50 HD076210 to A.G. and D.P.), the Biomedical Research Council, the Goertz, M. J., Wu, Z., Gallardo, T. D., Hamra, F. K. and Castrillon, D. H. (2011). Agency for Science, Technology and Research (A*STAR; to P.K.), a Singapore Foxo1 is required in mouse spermatogonial stem cells for their maintenance and International Graduate Award (SINGA; to N.P.), a Biomedical Research Council – the initiation of spermatogenesis. J. Clin. Invest. 121, 3456-3466. doi:10.1172/ Joint Council Office grant (1231AFG031 to P.K.), the National Medical Research JCI57984 Council Singapore, NMRC (NMRC/CBRG/0091/2015 to P.K.), a National Research Gu, Y., Rosenblatt, J. and Morgan, D. O. (1992). Cell cycle regulation of CDK2 Foundation Singapore grant (NRF2016-CRP001-103 to P.K.) and contract activity by phosphorylation of Thr160 and Tyr15. EMBO J. 11, 3995-4005. doi:10. CO29155 from the New York State Stem Cell Science Program (NYSTEM). 1002/j.1460-2075.1992.tb05493.x Deposited in PMC for release after 12 months. Hamer, G. and de Rooij, D. G. (2018). Mutations causing specific arrests in the development of mouse primordial germ cells and gonocytes. Biol. Reprod. 99, Data availability 75-86. doi:10.1093/biolre/ioy075 The 10x single cell RNAseq data have been deposited in Gene Expression Omnibus Hara, K., Nakagawa, T., Enomoto, H., Suzuki, M., Yamamoto, M., Simons, B. D. with accession GSE130554. and Yoshida, S. (2014). Mouse spermatogenic stem cells continually interconvert between equipotent singly isolated and syncytial states. Cell Stem Cell 14, 658-672. doi:10.1016/j.stem.2014.01.019 Supplementary information Hobbs, R. M., Seandel, M., Falciatori, I., Rafii, S. and Pandolfi, P. P. (2010). Plzf Supplementary information available online at regulates germline progenitor self-renewal by opposing mTORC1. Cell 142, http://dev.biologists.org/lookup/doi/10.1242/dev.180273.supplemental 468-479. doi:10.1016/j.cell.2010.06.041 Hofmann, M.-C. (2008). Gdnf signaling pathways within the mammalian References spermatogonial stem cell niche. Mol. Cell. Endocrinol. 288, 95-103. doi:10. Artegiani, B., Lindemann, D. and Calegari, F. (2011). Overexpression of cdk4 and 1016/j.mce.2008.04.012 cyclinD1 triggers greater expansion of neural stem cells in the adult mouse brain. Hu, Y.-C., de Rooij, D. G. and Page, D. C. (2013). Tumor suppressor gene Rb is J. Exp. Med. 208, 937-948. doi:10.1084/jem.20102167 required for self-renewal of spermatogonial stem cells in mice. Proc. Natl. Acad. Bellve, A., Cavicchia, J., Millette, C., O’Brien, D., Bhatnagar, Y. and Dym, M. Sci. USA 110, 12685-12690. doi:10.1073/pnas.1311548110 (1977). Spermatogenic cells of the prepuberal mouse: isolation and Huang, H., Regan, K. M., Lou, Z., Chen, J. and Tindall, D. J. (2006). CDK2- morphological characterization. J. Cell. Biol. 74, 68-85. doi:10.1083/jcb.74.1.68 dependent phosphorylation of FOXO1 as an apoptotic response to DNA damage. Berthet, C., Aleem, E., Coppola, V., Tessarollo, L. and Kaldis, P. (2003). Cdk2 Science 314, 294-297. doi:10.1126/science.1130512 knockout mice are viable. Curr. Biol. 13, 1775-1785. doi:10.1016/j.cub.2003.09.024 Huang, M., Wang, J., Torre, E., Dueck, H., Shaffer, S., Bonasio, R., Murray, J. I., Bhang, D. H., Kim, B.-J., Kim, B. G., Schadler, K., Baek, K.-H., Kim, Y. H., Hsiao, Raj, A., Li, M. and Zhang, N. R. (2018). SAVER: gene expression recovery for single-cell RNA sequencing. Nat. Methods 15, 539-542. doi:10.1038/s41592- W., Ding, B.-S., Rafii, S., Weiss, M. J. et al. (2018). Testicular endothelial cells 018-0033-z are a critical population in the germline stem cell niche. Nat. Commun. 9, 4379. Huckins, C. (1971a). The spermatogonial stem cell population in adult rats. I. Their doi:10.1038/s41467-018-06881-z morphology, proliferation and maturation. Anat. Rec. 169, 533-557. doi:10.1002/ Buaas, F. W., Kirsh, A. L., Sharma, M., McLean, D. J., Morris, J. L., Griswold, ar.1091690306 M. D., de Rooij, D. G. and Braun, R. E. (2004). Plzf is required in adult male germ Huckins, C. (1971b). The spermatogonial stem cell population in adult rats. III. cells for stem cell self-renewal. Nat. Genet. 36, 647-652. doi:10.1038/ng1366 Evidence for a long-cycling population. Cell Tissue Kinet. 4, 335-349. doi:10. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. and Satija, R. (2018). Integrating 1111/j.1365-2184.1971.tb01544.x single-cell transcriptomic data across different conditions, technologies, and Hughes, B. T., Sidorova, J., Swanger, J., Monnat, R. J. and Clurman, B. E.

species. Nat. Biotechnol. 36, 411-420. doi:10.1038/nbt.4096 (2013). Essential role for Cdk2 inhibitory phosphorylation during replication stress DEVELOPMENT

14 STEM CELLS AND REGENERATION Development (2019) 146, dev180273. doi:10.1242/dev.180273

revealed by a human Cdk2 knockin mutation. Proc. Natl. Acad. Sci. USA 110, Oatley, J. M. and Brinster, R. L. (2012). The germline stem cell niche unit in 8954-8959. doi:10.1073/pnas.1302927110 mammalian testes. Physiol. Rev. 92, 577-595. doi:10.1152/physrev.00025.2011 Johnston, D. S., Wright, W. W., Dicandeloro, P., Wilson, E., Kopf, G. S. and Ohbo, K., Yoshida, S., Ohmura, M., Ohneda, O., Ogawa, T., Tsuchiya, H., Jelinsky, S. A. (2008). Stage-specific gene expression is a fundamental Kuwana, T., Kehler, J., Abe, K., Schöler, H. R. et al. (2003). Identification and characteristic of rat spermatogenic cells and Sertoli cells. Proc. Natl. Acad. Sci. characterization of stem cells in prepubertal spermatogenesis in mice. Dev. Biol. USA 105, 8315-8320. doi:10.1073/pnas.0709854105 258, 209-225. doi:10.1016/S0012-1606(03)00111-8 Kaldis, P. (1999). The cdk-activating kinase (CAK): from yeast to mammals. Cell Ohmura, M., Yoshida, S., Ide, Y., Nagamatsu, G., Suda, T. and Ohbo, K. (2004). Mol. Life Sci. 55, 284-296. doi:10.1007/s000180050290 Spatial analysis of germ stem cell development in Oct-4/EGFP transgenic mice. Kang, H. S., Chen, L.-Y., Lichti-Kaiser, K., Liao, G., Gerrish, K., Bortner, C. D., Arch. Histol. Cytol. 67, 285-296. doi:10.1679/aohc.67.285 Yao, H. H.-C., Eddy, E. M. and Jetten, A. M. (2016). Transcription factor GLIS3: A Ortega, S., Prieto, I., Odajima, J., Martın,́ A., Dubus, P., Sotillo, R., Barbero, J. L., new and critical regulator of postnatal stages of mouse spermatogenesis. Stem Malumbres, M. and Barbacid, M. (2003). Cyclin-dependent kinase 2 is essential Cells 34, 2772-2783. doi:10.1002/stem.2449 for meiosis but not for mitotic cell division in mice. Nat. Genet. 35, 25-31. doi:10. Klein, A. M., Nakagawa, T., Ichikawa, R., Yoshida, S. and Simons, B. D. (2010). 1038/ng1232 Mouse germ line stem cells undergo rapid and stochastic turnover. Cell Stem Cell Percharde, M., Wong, P. and Ramalho-Santos, M. (2017). Global 7, 214-224. doi:10.1016/j.stem.2010.05.017 hypertranscription in the mouse embryonic germline. Cell Rep. 19, 1987-1996. Krasinska, L., Cot, E. and Fisher, D. (2008). Selective chemical inhibition as a tool doi:10.1016/j.celrep.2017.05.036 to study Cdk1 and Cdk2 functions in the cell cycle. Cell Cycle 7, 1702-1708. Pietras, E. M., Warr, M. R. and Passegué,E.(2011). Cell cycle regulation in doi:10.4161/cc.7.12.6101 hematopoietic stem cells. J. Cell Biol. 195, 709-720. doi:10.1083/jcb.201102131 La Manno, G., Soldatov, R., Zeisel, A., Braun, E., Hochgerner, H., Petukhov, V., Pui, H. P. and Saga, Y. (2017). Gonocytes-to-spermatogonia transition initiates prior Lidschreiber, K., Kastriti, M. E., Lönnerberg, P., Furlan, A. et al. (2018). RNA to birth in murine testes and it requires FGF signaling. Mech. Dev. 144, 125-139. velocity of single cells. Nature 560, 494-498. doi:10.1038/s41586-018-0414-6 doi:10.1016/j.mod.2017.03.002 Lange, C., Huttner, W. B. and Calegari, F. (2009). Cdk4/cyclinD1 overexpression Qiu, X., Hill, A., Packer, J., Lin, D., Ma, Y.-A. and Trapnell, C. (2017). Single-cell in neural stem cells shortens G1, delays neurogenesis, and promotes the mRNA quantification and differential analysis with Census. Nat. Methods 14, generation and expansion of basal progenitors. Cell Stem Cell 5, 320-331. doi:10. 309-315. doi:10.1038/nmeth.4150 1016/j.stem.2009.05.026 Ravnik, S. E. and Wolgemuth, D. J. (1999). Regulation of meiosis during Law, N. C., Oatley, M. J. and Oatley, J. M. (2019). Developmental kinetics and mammalian spermatogenesis: the A-type cyclins and their associated cyclin- transcriptome dynamics of stem cell specification in the spermatogenic lineage. dependent kinases are differentially expressed in the germ-cell lineage. Dev. Biol. Nat. Commun. 10, 2787. doi:10.1038/s41467-019-10596-0 207, 408-418. doi:10.1006/dbio.1998.9156 Liao, J., Ng, S. H., Tu, J., Luk, A. C. S., Qian, Y., Tang, N. L. S., Feng, B., Chan, Rubin, S. M. (2013). Deciphering the retinoblastoma protein phosphorylation code. W.-Y., Fouchet, P. and Lee, T.-L. (2017). Single-cell RNA-seq resolves cellular Trends Biochem. Sci. 38, 12-19. doi:10.1016/j.tibs.2012.10.007 heterogeneity and transcriptional dynamics during spermatogonia stem cells Sasaki, H. and Matsui, Y. (2008). Epigenetic events in mammalian germ-cell establishment and differentiation. bioRxiv. doi:10.1101/194696 development: and beyond. Nat. Rev. Genet. 9, 129-140. doi:10. Lim, S. and Kaldis, P. (2012). Loss of Cdk2 and Cdk4 induces a switch from 1038/nrg2295 proliferation to differentiation in neural stem cells. Stem Cells 30, 1509-1520. Satija, R., Farrell, J. A., Gennert, D., Schier, A. F. and Regev, A. (2015). Spatial doi:10.1002/stem.1114 reconstruction of single-cell gene expression data. Nat. Biotechnol. 33, 495-502. Lim, S. and Kaldis, P. (2013). Cdks, cyclins and CKIs: roles beyond cell cycle doi:10.1038/nbt.3192 regulation. Development 140, 3079-3093. doi:10.1242/dev.091744 Satyanarayana, A. and Kaldis, P. (2009). Mammalian cell-cycle regulation: several Lim, S., Bhinge, A., Bragado Alonso, S., Aksoy, I., Aprea, J., Cheok, C. F., Cdks, numerous cyclins and diverse compensatory mechanisms. Oncogene 28, Calegari, F., Stanton, L. W. and Kaldis, P. (2017). Cyclin-dependent kinase- 2925-2939. doi:10.1038/onc.2009.170 dependent phosphorylation of Sox2 at Serine 39 regulates neurogenesis. Mol. Savitt, J., Singh, D., Zhang, C., Chen, L.-C., Folmer, J., Shokat, K. M. and Cell. Biol. 37, e00201-17. doi:10.1128/MCB.00201-17 Wright, W. W. (2012). The in vivo response of stem and other undifferentiated Liu, D., Matzuk, M. M., Sung, W. K., Guo, Q., Wang, P. and Wolgemuth, D. J. spermatogonia to the reversible inhibition of glial cell line-derived neurotrophic (1998). Cyclin A1 is required for meiosis in the male mouse. Nat. Genet. 20, factor signaling in the adult. Stem Cells 30, 732-740. doi:10.1002/stem.1028 377-380. doi:10.1038/3855 Sharma, M., Srivastava, A., Fairfield, H. E., Bergstrom, D., Flynn, W. F. and Macosko, E. Z., Basu, A., Satija, R., Nemesh, J., Shekhar, K., Goldman, M., Braun, R. E. (2019). Identification of EOMES-expressing spermatogonial stem Tirosh, I., Bialas, A. R., Kamitaki, N., Martersteck, E. M. et al. (2015). Highly cells and their regulation by PLZF. eLife 8, e43352. doi:10.7554/eLife.43352 parallel genome-wide expression profiling of individual cells using nanoliter Singh, P. and Schimenti, J. C. (2015). The genetics of human infertility by droplets. Cell 161, 1202-1214. doi:10.1016/j.cell.2015.05.002 Martinerie, L., Manterola, M., Chung, S. S. W., Panigrahi, S. K., Weisbach, M., functional interrogation of SNPs in mice. Proc. Natl. Acad. Sci. USA 112, Vasileva, A., Geng, Y., Sicinski, P. and Wolgemuth, D. J. (2014). Mammalian 10431-10436. doi:10.1073/pnas.1506974112 ’ E-type cyclins control chromosome pairing, telomere stability and CDK2 Singh, P., Schimenti, J. C. and Bolcun-Filas, E. (2015). A mouse geneticist s localization in male meiosis. PLoS Genet. 10, e1004165. doi:10.1371/journal. practical guide to CRISPR applications. Genetics 199, 1-15. doi:10.1534/ pgen.1004165 genetics.114.169771 McConnell, M. J., Chevallier, N., Berkofsky-Fessler, W., Giltnane, J. M., Malani, Song, H.-W., Bettegowda, A., Lake, B. B., Zhao, A. H., Skarbrevik, D., Babajanian, R. B., Staudt, L. M. and Licht, J. D. (2003). Growth suppression by acute E., Sukhwani, M., Shum, E. Y., Phan, M. H., Plank, T.-D. M. et al. (2016). The promyelocytic leukemia-associated protein PLZF is mediated by repression of homeobox transcription factor RHOX10 drives mouse spermatogonial stem cell c-myc expression. Mol. Cell. Biol. 23, 9375-9388. doi:10.1128/MCB.23.24.9375- establishment. Cell Rep. 17, 149-164. doi:10.1016/j.celrep.2016.08.090 9388.2003 Subramanian, A., Tamayo, P., Mootha, V. K., Mukherjee, S., Ebert, B. L., Gillette, McCurdy, S. R., Pacal, M., Ahmad, M. and Bremner, R. (2017). A CDK2 activity M. A., Paulovich, A., Pomeroy, S. L., Golub, T. R., Lander, E. S. et al. (2005). signature predicts outcome in CDK2-low . Oncogene 36, 2491-2502. Gene set enrichment analysis: a knowledge-based approach for interpreting doi:10.1038/onc.2016.409 genome-wide expression profiles. Proc. Natl. Acad. Sci. USA 102, 15545-15550. McGinnis, C. S., Murrow, L. M. and Gartner, Z. J. (2019). DoubletFinder: doublet doi:10.1073/pnas.0506580102 detection in single-cell RNA sequencing data using artificial nearest neighbors. Szabó, P. E., Hübner, K., Schöler, H. and Mann, J. R. (2002). Allele-specific Cell Syst. 8, 329-337.e4. doi:10.1016/j.cels.2019.03.003 expression of imprinted genes in mouse migratory primordial germ cells. Mech. Meng, X., Lindahl, M., Hyvönen, M. E., Parvinen, M., de Rooij, D. G., Hess, M. W., Dev. 115, 157-160. doi:10.1016/S0925-4773(02)00087-4 Raatikainen-Ahokas, A., Sainio, K., Rauvala, H., Lakso, M. et al. (2000). Szmyd, R., Niska-Blakie, J., Diril, M. K., Renck Nunes, P., Tzelepis, K., Lacroix, Regulation of cell fate decision of undifferentiated spermatogonia by GDNF. A., van Hul, N., Deng, L.-W., Matos, J., Dreesen, O. et al. (2019). Premature Science 287, 1489-1493. doi:10.1126/science.287.5457.1489 activation of Cdk1 leads to mitotic events in S phase and embryonic lethality. Morgan, D. O. (1995). Principles of CDK regulation. Nature 374, 131-134. doi:10. Oncogene 38, 998-1018. doi:10.1038/s41388-018-0464-0 1038/374131a0 Tam, P. P. and Snow, M. H. (1981). Proliferation and migration of primordial germ Morris, L., Allen, K. E. and La Thangue, N. B. (2000). Regulation of E2F cells during compensatory growth in mouse embryos. J. Embryol. Exp. Morphol. transcription by cyclin E-Cdk2 kinase mediated through p300/CBP co-activators. 64, 133-147. Nat. Cell Biol. 2, 232-239. doi:10.1038/35008660 Tang, J.-X., Li, J., Cheng, J.-M., Hu, B., Sun, T.-C., Li, X.-Y., Batool, A., Wang, Z.- Nagano, R., Tabata, S., Nakanishi, Y., Ohsako, S., Kurohmaru, M. and Hayashi, P., Wang, X.-X., Deng, S.-L. et al. (2017). Requirement for CCNB1 in mouse Y. (2000). Reproliferation and relocation of mouse male germ cells (gonocytes) spermatogenesis. Cell Death Dis. 8, e3142. doi:10.1038/cddis.2017.555 during prespermatogenesis. Anat. Rec. 258, 210-220. doi:10.1002/(SICI)1097- Thomas, C. J., Cleland, T. P., Zhang, S., Gundberg, C. M. and Vashishth, D. 0185(20000201)258:2<210::AID-AR10>3.0.CO;2-X (2017). Identification and characterization of glycation adducts on osteocalcin. Nakagawa, T., Sharma, M., Nabeshima, Y., Braun, R. E. and Yoshida, S. (2010). Anal. Biochem. 525, 46-53. doi:10.1016/j.ab.2017.02.011 Functional hierarchy and reversibility within the murine spermatogenic stem cell Toyooka, Y., Tsunekawa, N., Takahashi, Y., Matsui, Y., Satoh, M. and Noce, T.

compartment. Science 328, 62-67. doi:10.1126/science.1182868 (2000). Expression and intracellular localization of mouse Vasa-homologue DEVELOPMENT

15 STEM CELLS AND REGENERATION Development (2019) 146, dev180273. doi:10.1242/dev.180273

protein during germ cell development. Mech. Dev. 93, 139-149. doi:10.1016/ proteins in single spots on comparative proteomics in conventional 2-D gel/MALDI S0925-4773(00)00283-5 workflow. Electrophoresis 28, 2080-2094. doi:10.1002/elps.200600524 Trapnell, C., Cacchiarelli, D., Grimsby, J., Pokharel, P., Li, S., Morse, M., Yang, Y., Anderson, E. and Zhang, S. (2018). Evaluation of six sample Lennon, N. J., Livak, K. J., Mikkelsen, T. S. and Rinn, J. L. (2014). The preparation procedures for qualitative and quantitative proteomics analysis of dynamics and regulators of cell fate decisions are revealed by pseudotemporal milk fat globule membrane. Electrophoresis 39, 2332-2339. doi:10.1002/elps. ordering of single cells. Nat. Biotechnol. 32, 381-386. doi:10.1038/nbt.2859 201800042 Varshney, G. K., Pei, W., LaFave, M. C., Idol, J., Xu, L., Gallardo, V., Carrington, Yeyati, P. L., Shaknovich, R., Boterashvili, S., Li, J., Ball, H. J., Waxman, S., B., Bishop, K., Jones, M. P., Li, M. et al. (2015). High-throughput gene targeting Nason-Burchenal, K., Dmitrovsky, E., Zelent, A. and Licht, J. D. (1999). and phenotyping in zebrafish using CRISPR/Cas9. Genome Res.. 25, 1030-1042. Leukemia translocation protein PLZF inhibits cell growth and expression of cyclin doi:10.1101/gr.186379.114 A. Oncogene 18, 925-934. doi:10.1038/sj.onc.1202375 Viera, A., Rufas, J. S., Martınez,I.,Barbero,J.L.,Ortega,S.andSuja,J.A.́ (2009). Yoshida, S., Takakura, A., Ohbo, K., Abe, K., Wakabayashi, J., Yamamoto, M., CDK2 is required for proper homologous pairing, recombination and sex-body formation Suda, T. and Nabeshima, Y.-I. (2004). Neurogenin3 delineates the earliest during male mouse meiosis. J. Cell Sci. 122, 2149-2159. doi:10.1242/jcs.046706 stages of spermatogenesis in the mouse testis. Dev. Biol. 269, 447-458. doi:10. Viera, A., Alsheimer, M., Gómez, R., Berenguer, I., Ortega, S., Symonds, C. E., 1016/j.ydbio.2004.01.036 Santamarıa,́ D., Benavente, R. and Suja, J. A. (2015). CDK2 regulates nuclear Yoshida, S., Sukeno, M., Nakagawa, T., Ohbo, K., Nagamatsu, G., Suda, T. and envelope protein dynamics and telomere attachment in mouse meiotic prophase. Nabeshima, Y.-I. (2006). The first round of mouse spermatogenesis is a J. Cell Sci. 128, 88-99. doi:10.1242/jcs.154922 distinctive program that lacks the self-renewing spermatogonia stage. Welburn, J. P. I., Tucker, J. A., Johnson, T., Lindert, L., Morgan, M., Willis, A., Development 133, 1495-1505. doi:10.1242/dev.02316 Noble, M. E. M. and Endicott, J. A. (2007). How tyrosine 15 phosphorylation Zhao, H., Chen, X., Gurian-West, M. and Roberts, J. M. (2012). Loss of cyclin- inhibits the activity of cyclin-dependent kinase 2-cyclin A. J. Biol. Chem. 282, dependent kinase 2 (CDK2) inhibitory phosphorylation in a CDK2AF knock-in 3173-3181. doi:10.1074/jbc.M609151200 mouse causes misregulation of DNA replication and centrosome duplication. Mol. Wolgemuth, D. J. and Roberts, S. S. (2010). Regulating and meiosis in the Cell. Biol. 32, 1421-1432. doi:10.1128/MCB.06721-11 male germ line: critical functions for cyclins. Philos. Trans. R. Soc. Lond. B Biol. Zheng, K., Wu, X., Kaestner, K. H. and Wang, P. J. (2009). The pluripotency factor Sci. 365, 1653-1662. doi:10.1098/rstb.2009.0254 LIN28 marks undifferentiated spermatogonia in mouse. BMC Dev. Biol. 9, 38. Yang, Q.-E. and Oatley, J. M. (2014). Spermatogonial stem cell functions in doi:10.1186/1471-213X-9-38 physiological and pathological conditions. Curr. Top. Dev. Biol. 107, 235-267. Ziegenhain, C., Vieth, B., Parekh, S., Reinius, B., Guillaumet-Adkins, A., Smets, doi:10.1016/B978-0-12-416022-4.00009-3 M., Leonhardt, H., Heyn, H., Hellmann, I. and Enard, W. (2017). Comparative Yang, Y., Thannhauser, T. W., Li, L. and Zhang, S. (2007). Development of an analysis of single-cell RNA sequencing methods. Mol. Cell 65, 631-643.e4. integrated approach for evaluation of 2-D gel image analysis: impact of multiple doi:10.1016/j.molcel.2017.01.023 DEVELOPMENT

16