TISSUE-SPECIFIC STEM CELLS

Size: px
Start display at page:

Download "TISSUE-SPECIFIC STEM CELLS"

Transcription

1 TISSUE-SPECIFIC STEM CELLS NANOS2 Acts Downstream of Glial Cell Line-Derived Neurotrophic Factor Signaling to Suppress Differentiation of Spermatogonial Stem Cells AIKO SADA, a KAZUTERU HASEGAWA, b PUI HAN PIN, a YUMIKO SAGA a,b a Department of Genetics, SOKENDAI, Mishima, Japan; b Division of Mammalian Development, National Institute of Genetics, Mishima, Japan Key Words. Germline Differentiation Tissue-specific stem cells Targeted gene disruption Transgenic mouse Stem cell microenvironment interactions ABSTRACT Stem cells are maintained by both stem cell-extrinsic niche signals and stem cell-intrinsic factors. During murine spermatogenesis, glial cell line-derived neurotrophic factor (GDNF) signal emanated from Sertoli cells and germ cellintrinsic factor NANOS2 represent key regulators for the maintenance of spermatogonial stem cells. However, it remains unclear how these factors intersect in stem cells to control their cellular state. Here, we show that GDNF signaling is essential to maintain NANOS2 expression, and overexpression of Nanos2 can alleviate the stem cell loss phenotype caused by the depletion of Gfra1, a receptor for GDNF. By using an inducible Cre-loxP system, we show that NANOS2 Disclosure of potential conflicts of interest is found at the end of this article. expression is downregulated upon the conditional knockout (cko) of Gfra1, while ectopic expression of Nanos2 in GFRA1-negative spermatogonia does not induce de novo GFRA1 expression. Furthermore, overexpression of Nanos2 in the Gfra1-cKO testes prevents precocious differentiation of the Gfra1-knockout stem cells and partially rescues the stem cell loss phenotypes of Gfra1-deficient mice, indicating that the stem cell differentiation can be suppressed by NANOS2 even in the absence of GDNF signaling. Taken together, we suggest that NANOS2 acts downstream of GDNF signaling to maintain undifferentiated state of spermatogonial stem cells. STEM CELLS 2012;30: INTRODUCTION Spermatogonial stem cells represent a stem cell population in adult testes, and their proper biological activities ensure a continuous production of mature spermatozoa. The stem cell function resides in the undifferentiated spermatogonia, consisting of A single (A s ; isolated single cells), A paired (A pr ; interconnected two cells), and A aligned (A al ; interconnected 4, 8, 16, or 32 cells) [1 5]. The undifferentiated spermatogonia transform into A 1 differentiating spermatogonia, which subsequently undergo six mitotic and two meiotic divisions to form haploid spermatozoa [5]. In addition to the morphological classification, undifferentiated spermatogonia are also characterized by the expressions of various marker genes [6 10] (Supporting Information Fig. 1A): promyelocytic leukemia zinc finger (PLZF) is expressed in the all types of undifferentiated spermatogonia; NANOS2 and glial cell line-derived neurotrophic factor family receptor a 1 (GFRA1) are expressed in large subsets of A s and A pr ; NANOS3 and Neurogenin3 (Ngn3) are known as markers for A al spermatogonia. Although the entire undifferentiated spermatogonial pool might together constitute the stem cell population during homeostasis and regeneration [7, 11, 12], the heterogeneous gene expression within the population correlates strongly with the cell fates in the short term: Ngn3-expressing cells tend to differentiate, whereas NANOS2- or GFRA1-expressing cells are more likely to self-renew [7, 13]. Therefore, it would be valuable to ask how cellular states or fates of NANO- S2 þ GFRA1 þ and NANOS3 þ Ngn3 þ cells are regulated during spermatogenesis. In this article, we define hereafter NANO- S2 þ GFRA1 þ undifferentiated spermatogonia as spermatogonial stem cells and upregulation of NANOS3 and/or Ngn3 as spermatogonial stem cell differentiation. External niche stimuli and internal gene expression regulate self-renewal and differentiation of spermatogonial stem cells [13 22]. The growth factor glial cell line-derived neurotrophic factor (GDNF), a ligand for GFRA1, is secreted by somatic Sertoli cells and acts as one of the major niche signals for spermatogonial stem cells. GDNF signals via the GFRA1/ RET coreceptor through activation of Src family kinases, Ras and phosphoinositide 3-kinase-Akt pathway and subsequently induces several target genes in spermatogonial stem cells [23 31]. Previous study has shown that Gdnf haploinsufficiency led to a progressive germ cell loss phenotype, and that testicular overexpression of Gdnf resulted in an accumulation of Author contributions: A.S.: conception and design, data analysis and interpretation, collection and assembly of data, and manuscript writing; K.H.: collection and assembly of data, data analysis and interpretation, and manuscript writing; P.H.P.: collection and assembly of data and manuscript writing; Y.S.: conception and design, manuscript writing, and financial support. Correspondence: Yumiko Saga, Ph.D., Division of Mammalian Development, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka , Japan. Telephone: þ ; Fax: þ ; ysaga@lab.nig.ac.jp Received April 4, 2011; accepted for publication November 9, 2011; first published online in STEM CELLS EXPRESS November 18, VC AlphaMed Press /2012/$30.00/0 doi: /stem.790 STEM CELLS 2012;30:

2 Sada, Hasegawa, Pin et al. 281 undifferentiated spermatogonia [19]. Although mice lacking Gdnf or its receptors exhibit neonatal lethality, transplanted testes from each of Gdnf-, Gfra1-, and Ret-null newborn mice showed severe spermatogonial cell depletion, which might result from deficits in spermatogonial proliferation and/or in their maintenance of an undifferentiated state [20]. It is as yet unclear how GDNF signaling influences adult spermatogenesis, as germ cells are lost by postnatal day (P) 7 in the testes of null mutants of Gdnf or its receptors [20]. An RNA-binding protein NANOS2 is an intrinsic factor required for the maintenance of spermatogonial stem cells [13]. Previously, we reported that the removal of Nanos2 gene in adult testes resulted in the depletion of spermatogonial stem cells, which was evidenced by the absence of PLZF and GFRA1 expression [13]. In contrast, continuous Nanos2 expression in the germline caused an accumulation of spermatogonia, which were positive for PLZF and GFRA1 but exhibited no or lower levels of NANOS3 or Ngn3 expression [13]. Furthermore, the Nanos2-overexpressing cells showed slower proliferation and wild-type levels of apoptosis [13]. These results indicate that NANOS2 suppresses proliferation and differentiation of spermatogonial stem cells. As NANOS2 and GDNF (or its receptors) induced similar phenotypes upon loss of function and overexpression experiments with respect to spermatogonial stem cell maintenance, we examined the relationship between these two factors. We showed that GDNF signaling pathway induced/maintained NANOS2 expression but not vice versa. Furthermore, the stem cell loss phenotype of the Gfra1 conditional knockout (cko) mice was partially rescued by overexpressed Nanos2. To verify this phenotype, we examined how stem cell properties were affected by Gfra1 depletion and by Nanos2-overexpression. Our results indicated that NANOS2 was capable of suppressing differentiation of spermatogonial stem cells in the absence of GDNF signaling. MATERIALS AND METHODS Nanos2-Overexpression Induced by Ngn3-Cre To characterize recombined cells by Ngn3-Cre, we crossed Ngn3- Cre with Rosa yellow fluorescent protein (YFP) reporter (courtesy of Dr. R. Kopan) [32] (Supporting Information Fig. 1B) mice. We dissected Ngn3-Cre; Rosa-YFP double transgenic testes at P7 and 6 weeks (Supporting Information Fig. 1D). For conditional activation of Nanos2, we intercrossed the CAG-floxed CAT-3xFlag-Nanos2 (Tg-Nanos2) transgenic (Supporting Information Fig. 1C) female mice with Ngn3-Cre (courtesy of Dr. S. Yoshida) [33] male mice to obtain Tg-Nanos2; Ngn3-Cre double transgenic male pups. The littermate Tg-Nanos2 transgenic males (without Cre) were used as controls. We used 6-week-old mice for phenotypic analysis (Supporting Information Fig. 1D). Simultaneous Gfra1-cKO and Nanos2-Overexpression by a Tamoxifen-Inducible Cre To induce conditional inactivation of Gfra1, Gfra1 floxed mice (courtesy of Dr. H. Enomoto) [34] (Supporting Information Fig. 4A) were crossed with Cre-Ert2 line that ubiquitously expressed a tamoxifen (TM)-inducible Cre recombinase (Artemis Pharmaceuticals GmbH). For conditional activation of Nanos2, we subsequently introduced transgene, CAG-floxed CAT-3xFlag-Nanos2 (Tg-Nanos2) [35] (Supporting Information Fig. 4B) in the Gfra1-cKO background. We generated in a total four kinds of mice harboring the following alleles: (a) Gfra1 flox/þ ; Cre-Ert2, (b) Gfra1 flox/flox ; Cre- Ert2, (c) Gfra1 flox/þ ; Cre-Ert2; Tg-Nanos2, and (d) Gfra1 flox/flox ; Cre-Ert2; Tg-Nanos2. Correspondingly, activation of Cre recombinase produces (a) Gfra1 D/þ ; Cre-Ert2 (control), (b) Gfra1 D/D ; Cre-Ert2 (Gfra1-cKO), (c) Gfra1 D/þ ; Cre-Ert2; Tg-Nanos2 (Nanos2- overexpression), and (d) Gfra1 D/D ; Cre-Ert2; Tg-Nanos2 (Nanos2- overexpression in the Gfra1-cKO background). Cre activity was induced by intraperitoneal injection of TM (75 mg/kg body weight; Sigma, St. Louis, MO, dissolved in corn oil R) into 4-week-old mice for 5 consecutive days. The testes from each genotype of mice were dissected at indicated time points (Supporting Information Fig. 4C). Whole-Mount Immunohistochemistry After removing from the tunica albuginea, the seminiferous tubules were untangled physically. The tubules were fixed with 4% paraformaldehyde (PFA) for 2 hours at 4 C. After washing with PBS-T (0.3% TritonX-100/PBS), the specimens were incubated with 3% skim milk/pbs-t for 1 hour at room temperature (RT) and subsequently with primary antibodies at RT overnight. The following day, the tubules were washed with PBS-T and were incubated with secondary antibodies at 4 C overnight. After that, they were washed with PBS-T before mounting. The specimens were examined using a confocal laser scanning microscope (Zeiss, Germany, LSM510). Primary antibodies were used at the following dilutions: mouse anti-flag (1:5,000, Sigma), rabbit anti-plzf (1:1,000, Santa Cruz, Santa Cruz, CA, mouse anti-plzf (1:500, Calbiochem, Darmstadt, Germany, ence-research/calbiochem/c_m7eb.s1ogumaaaejt.h9.zf4), rabbit anti-nanos2 [36] (1:500), goat anti-gfra1 (1:200, R&D Systems, Minneapolis, MN, rat anti-green fluorescent protein (GFP; 1:500, Nacalai Tesque, Kyoto, Japan, co.jp/global/index.html), and rabbit anti-nanos3 [36] (1:500). All secondary antibodies were used at a 1:250 dilution: anti-rat, anti-rabbit, anti-goat, or anti-mouse IgG antibodies conjugated with either Alexa-488 or Alexa-594 (Molecular Probes, Eugene, OR, www. invitrogen.com/site/us/en/home/brands/molecular-probes.html). Histological Methods For immunofluorescence analysis, testes were fixed in 4% PFA at 4 C overnight before they were embedded in paraffin or OCT compound (Tissue Tek, Sakura, Tokyo, Japan, tek.com/top_e.html). Paraffin sections (6 lm) were autoclaved at 105 C for 15 minutes in antigen unmasking solution (Vector Laboratories, Burlingame, CA, after deparaffinization and hydration. Frozen sections (8 lm) were rinsed with PBS. After blocking with PBS containing 3% skim milk for 1 hour at RT, sections were rinsed and incubated overnight with primary antibodies at 4 C. The following day, the sections were washed in PBS and incubated with secondary antibodies for 1 hour at RT. After washing, these sections were counterstained with 4 0,6-diamidino-2-phenylindole (Sigma) and enclosed. Primary antibodies were used at the following dilutions: goat anti-gfra1 (1:200, Neuromics, Edina, MN, chick anti-gfp (1:500, Abcam, Cambridge, MA, chick anti-gfp (1:500, Aves Labs), rat TRA98 (1:5,000, courtesy of Dr. Y. Nishimune), rabbit anti-plzf (1:500, Santa Cruz), mouse anti- PLZF (1:100, Calbiochem), mouse anti-flag (1:5,000, Sigma), mouse anti-cleaved PARP (1:200, Cell Signaling, Beverly, MA, rabbit anti synaptonemal complex protein 3 (1:500, courtesy of Drs. S. Chuma and N. Nakatsuji), rabbit anti-phospho histone H3 (1:500, Millipore, Billerica, MA, com/index.do), and rabbit anti-nanos2 (1:100). All secondary antibodies were used at a 1:250 dilution: anti-goat, anti-chick, anti-rat, anti-rabbit, or anti-mouse IgG antibodies conjugated with either Alexa-488 or Alexa-594 (Molecular Probes). Statistics In each graph, the mean values were shown with the standard error. All statistical analyses in this study were performed using the Student s t test. Statistical significance was defined as p <.05.

3 282 Relationship Between NANOS2 and GDNF Signaling RESULTS Ectopic Expression of Nanos2 in GFRA1-Negative Spermatogonia Did Not Induce De Novo GFRA1 Expression Our previous study showed that the majority of Nanos2-overexpressing cells expressed GFRA1 when we induced Nanos2 by Nanos3-Cre, in which Cre recombination occurs in the embryonic male gonocytes, precursors of spermatogonial stem cells [13]. The result raises two possibilities: (a) NANOS2 acts upstream of GDNF signaling and is directly involved in the induction/maintenance of GFRA1 expression, and (b) GFRA1 expression is maintained independently of NANOS2 where GFRA1 expression might be a coincidental but indirect consequence of the NANOS2-mediated differentiation block. To test whether NANOS2 directly affects GFRA1 expression, we investigated the effect of ectopic Nanos2-overexpression in GFRA1-negative spermatogonia. Given that Ngn3 expression commences around 7 days after birth [33, 37] (Supporting Information Fig. 1D) and the expression is restricted in a GFRA1-negative undifferentiated spermatogonia in adult [7, 8] (Supporting Information Fig. 1A), we expect that Nanos2 should be induced in GFRA1-negative population from early postnatal stage by Ngn3-Cre. To confirm this, we crossed Ngn3-Cre mice with Rosa-YFP reporter mice (Supporting Information Fig. 1B) that allowed us to visualize the cells that had experienced Ngn3 expression. At P7, we detected YFP-positive germ cells on the basement membrane of seminiferous tubules (Supporting Information Fig. 2A to 2F), reflecting Ngn3-positive germ cells and their descendants at this stage. These Ngn3-lineage cells did not overlap with GFRA1- positive spermatogonia, as evidenced that more than 90% of GFRA1-positive cells were YFP negative (Supporting Information Fig. 2G). In agreement with the high efficiency of this Cre line [33, 37], the 6-week-old double transgenic testes revealed that the majority of the spermatogenic germ cells including spermatogonia, spermatocytes, and spermatids were positive for YFP, as a result of the Ngn3-Cre-mediated recombination that occurred in spermatogonial population by this age (Fig. 1A, 1D). However, when we carefully observed YFP expression, most of GFRA1-positive undifferentiated spermatogonia remained as YFP negative (Fig. 1A 1F, Supporting Information Fig. 2G). Hence, Cre recombination was indeed introduced mostly in GFRA1-negative spermatogonia by using Ngn3-Cre. To address if NANOS2 could induce GFRA1 expression, we overexpressed Nanos2 in GFRA1-negative population by crossing Ngn3-Cre mice with CAG-floxed CAT-3xFlag-Nanos2 transgenic (hereafter referred to as Tg-Nanos2) mice (Supporting Information Fig. 1C). We examined marker expressions using whole-mount immunostaining in the control and Ngn3- Cre-driven Nanos2-overexpressing testes at age of 6 weeks. In controls, PLZF was expressed in undifferentiated spermatogonia including A s,a pr, and A al (Fig. 1G 1I). Whereas GFRA1 was expressed predominantly in A s and A pr spermatogonia, in which endogenous NANOS2 was coexpressed (Fig. 1M 1O). In the Nanos2-overexpressing testes, NANOS2-positive cells were positive for PLZF (Fig. 1J 1L), indicating their property of undifferentiated spermatogonia. Intriguingly, most of these Nanos2-overexpressing cells remained as GFRA1 negative (Fig. 1P 1R). This result indicates that Nanos2-overexpression by Ngn3-Cre did not induce de novo GFRA1 expression. We quantified the results by using testis cross-sections and found that 72.5% of FLAG-NANOS2-positive cells were GFRA1 negative (Supporting Information Fig. 3A 3C, 3J). Furthermore, the proportion of GFRA1-positive cells in PLZF-positive population was not changed in the condition of Nanos2-overexpression (Supporting Information Fig. 3D 3I, 3K). From these results, we ruled out the possibility that NANOS2 acts directly in the induction of GFRA1 expression. NANOS2 Suppresses Differentiation of GFRA1- Negative Undifferentiated Spermatogonia NANOS3 þ Ngn3 þ spermatogonia express neither NANOS2 nor GFRA1 (Supporting Information Fig. 1A) and might be in a more differentiated state compared with the NANO- S2 þ GFRA1 þ state. To test if NANOS2 could suppress spermatogonial differentiation and maintain stem cell-like state in the absence of GDNF signaling, we investigate the differentiation states of Nanos2-overexpressing spermatogonia driven by Ngn3-Cre, in which these Nanos2-overexpressing cells do not receive GDNF signals because of a lack of GFRA1-receptor expression (Fig. 1M 1R, Supporting Information Fig. 3). Similar to the case in which Nanos2 was induced via Nanos3-Cre [13], the Ngn3-Cre-driven Nanos2-overexpression resulted in the accumulation of PLZF-positive spermatogonia in the periphery of the tubules, while TRA98-positive germ cells in luminal side of the tubules were dramatically reduced (Fig. 2C, 2D). This result suggests that ectopic Nanos2 disturbs the proper production of differentiating germ cells. Furthermore, the PLZF-positive cells in the Nanos2-overexpressing mice showed lower proliferation rates (Fig. 2B) and similar levels of apoptosis (Fig. 2A) compared with control spermatogonia. Hence, Nanos2-overexpression decreases proliferation and differentiation but does not affect apoptosis even in the GFRA1-negative spermatogonia. To further ask the effects of ectopic Nanos2 on spermatogonial differentiation, we examined the expression of NANOS3, an earliest differentiation marker. We found that A al spermatogonia in control testes showed high levels of NANOS3 (Fig. 2E 2G); however, the majority of Nanos2- overexpressing cells were NANOS3-negative or -dim (Fig. 2H 2J). These results indicate that NANOS2 is able to suppress differentiation of spermatogonia without GDNF receptor expression. NANOS2 Expression Was Lost Upon the cko of Gfra1 We hypothesized that GDNF signaling pathway acts upstream of NANOS2 in the spermatogonial stem cells. To test this hypothesis, we examined NANOS2 expression in Gfra1-cKO mice. We crossed mice carrying a gene cassette composed of floxed Gfra1 cdna followed by EGFP cdna [34] (Supporting Information Fig. 4A) with Cre-ERT2 mice that ubiquitously express a TM-inducible Cre recombinase. We injected TM into 4-week-old of (a) Gfra1 flox/þ (heterozygous for the Gfra1 floxed allele); Cre-Ert2 or (b) Gfra1 flox/flox (homozygous for the Gfra1 floxed allele); Cre-Ert2 mice for 5 consecutive days, resulting in the generation of (a) Gfra1 D/þ (Gfra1 heterozygous used as control) or (b) Gfra1 D/D (Gfra1-cKO) mice, respectively (Supporting Information Fig. 4C). In the Gfra1 floxed mouse strain, the excision of the floxed Gfra1 gene results in the expression of a reporter EGFP gene, which is inserted into the Gfra1 locus (Supporting Information Fig. 4A). This allowed us to visualize Gfra1 enhancer/promoter activated cells that have undergone Cre recombination, through monitoring of GFP fluorescence. Therefore, we can compare Gfra1-positive, normal spermatogonial stem cells versus Gfra1-null spermatogonial stem cells just after the gene is deleted. At 5 weeks of age (1 week after the first TM injection), we detected GFRA1 protein expression in GFP (Dfloxed

4 Sada, Hasegawa, Pin et al. 283 Figure 1. Nanos2-overexpression in GFRA1-negative spermatogonia. (A F): Immunostaining of 6-week-old Rosa-YFP; Ngn3-Cre testes with the indicated markers. YFP expression is observed in Ngn3-Cre expressing cells and their offsprings. GFRA1- (C) or PLZF- (F) single positive cells were indicated by arrowheads. (G R): Whole-mount immunostaining of 6-week-old Tg-Nanos2; Ngn3-Cre testes with the indicated markers. The transgene-derived Nanos2 was detected by staining with either anti-flag or endogenous anti-nanos2 antibodies. The dotted lines show outlines of seminiferous tubules. Scale bars ¼ 100 lm. Abbreviations: Aal, Aaligned; Apr, Apaired; As, Asingle; DAPI, 40,6-diamidino-2-phenylindole; GFRA1, glial cell line-derived neurotrophic factor family receptor a 1; Ngn3, Neurogenin3; PLZF, promyelocytic leukemia zinc finger; Tg, transgene; YFP, yellow fluorescent protein.

5 284 Relationship Between NANOS2 and GDNF Signaling Figure 2. Characterization of Nanos2-overexpressing spermatogonia induced by Ngn3-Cre. (A, B): Quantification of apoptotic (A) and proliferative (B) spermatogonia in 6-week-old mice (N=3). PARP is one of the main targets of CASPASE-3, therefore the PARP cleavage serves as a marker of cells undergoing apoptosis. **, p <.01. (C, D): Six-week-old testes were stained with the indicated markers. TRA98 is a germ cell marker that is expressed in spermatogonia, spermatocytes, and round spermatids. (E J): Whole-mount immunostaining of 6-week-old testes with the indicated markers. Scale bars ¼ 100 lm. Abbreviations: A al,a aligned ; DAPI, 4 0,6-diamidino-2-phenylindole; Ngn3, Neurogenin3; PARP, poly (ADP-ribose) polymerase; PH3, phospho histone H3; PLZF, promyelocytic leukemia zinc finger; Tg, transgene. Gfra1)-positive cells in control testes but not in Gfra1-cKO testes (Fig. 3A, 3B, 3I), indicating a successful deletion of Gfra1 upon TM injections. Then, we examined NANOS2 expression in the GFP-positive cells. Consistent with coexpressions of GFRA1 and NANOS2, more than 95% of GFPpositive cells were NANOS2-positive in the Gfra1 D/þ mice (Fig. 3E, 3J). Upon the knockout of Gfra1, NANOS2 expression was dramatically decreased in these cells although they still showed undifferentiated cell morphology such as A s or A pr (Fig. 3F, 3J). This result suggests that GFRA1-mediated GDNF signaling might induce/maintain NANOS2 expression. Induction of Nanos2-Overexpression in the Gfra1-cKO Background The essential role of GDNF signaling in the spermatogonial stem cell maintenance is shown by in vivo and in vitro studies [19, 20, 38, 39]. If NANOS2 were an essential downstream factor to achieve this function, we speculate that continuous expression of NANOS2 could negate the stem cell loss phenotype caused by the loss of GDNF signaling. To test this hypothesis, we overexpressed Nanos2 simultaneously with the cko of Gfra1. We introduced CAG-floxed CAT-3xFlag- Nanos2 transgene (Tg-Nanos2) (Supporting Information Fig. 4B)

6 Sada, Hasegawa, Pin et al. 285 Figure 3. GFRA1 and NANOS2 expression in Gfra1-positive spermatogonia. (A H): Whole-mount immunostaining of 5-week-old testes with the indicated markers. Scale bar ¼ 100 lm. (I, J): The ratio of either GFRA1-positive (I) or NANOS2-positive (J) clusters (2 n cells: 1, 2, 4, 8, 16) was counted in GFP-positive spermatogonial polulation (n ¼ 3). **, p <.01. Abbreviations: A s,a single ;A pr,a paired ;A al,a aligned ; GFP, green fluorescent protein; GFRA1, glial cell line-derived neurotrophic factor family receptor a 1; Tg, transgene. in the Gfra1 flox/þ ; Cre-Ert2 or Gfra1 flox/flox ; Cre-Ert2 mouse backgrounds. Injections of TM to (a) Gfra1 flox/þ ; Cre-Ert2; Tg- Nanos2 or (b) Gfra1 flox/flox ; Cre-Ert2; Tg-Nanos2 mice resulted in the generation of (a) Gfra1 D/þ ; Tg-Nanos2 (Nanos2-overexpression in control background) and (b) Gfra1 D/D ; Tg-Nanos2 (Nanos2-overexpression in Gfra1-cKO background), respectively. As Cre-Ert2 is ubiquitously expressed from Rosa26 locus, we expected that Nanos2 expression would be induced in both somatic cells and all stages of germ cells. However, after TM injections, NANOS2 expression was predominantly observed in spermatogonia and a part of spermatocyte, while later stages of germ cells and somatic cells were negative for NANOS2 (Supporting Information Fig. 5). This might be due to a higher efficiency of Cre recombination in mitotic/meiotic cells or a post-transcriptional repression of NANOS2 in later stage germ cells and somatic cells. In spite of the differential efficiency of NANOS2 expression among cell types, Cre-Ert2 successfully induced Nanos2-overexpression in GFP-positive spermatogonia in both Gfra1 D/þ and Gfra1 D/D backgrounds (Fig. 3C, 3D, 3G 3J). Partial Rescue of Gfra1-Knockout Phenotype by Overexpressed Nanos2 To determine the speed in which spermatogonial stem cells are lost upon Gfra1 deletion and the extent to which overexpressed Nanos2 could alleviate this phenotype, we examined the testes histology (Fig. 4) and changes in the numbers of PLZF (undifferentiated spermatogonia)- and GFP (Dfloxed Gfra1)-positive cells in each genotype at 5, 6, 8 and 12 weeks of age (Fig. 5). At 5 weeks, seminiferous tubules of control and all three mutant mice seemed similar, with multiple layers of germ cells (Fig. 4A, 4D, 4G, 4J). However, in the Gfra1 D/D mice, in addition to the overall decrease of PLZF-positive spermatogonia by 8 weeks (Fig. 5A, blue bar, Supporting Information Fig. 6C), there was a severe depletion of GFP-positive spermatogonia (Fig. 5B, blue bar, Supporting Information Fig. 6D) compared with controls (Fig. 5A, 5B, white bars, Supporting Information Fig. 6A, 6B). We could detect GFPpositive cells in the Gfra1-cKO testes until 5 weeks of age but never observed thereafter (Fig. 5B, blue bar). By 12 weeks, all types of germ cells had disappeared from the Gfra1 D/D testes, revealing Sertoli cell-only phenotypes (Fig. 4F). Hence, we confirmed that Gfra1 deficiency in adult testes resulted in the stem cell loss phenotype. On the contrary, if we compared the number of PLZFpositive spermatogonia in Gfra1-cKO testes with (Fig. 5A, green bar) or without (Fig. 5, blue bar) Tg-Nanos2, these cells were maintained for a prolonged period in the presence of Tg-Nanos2. At 8 weeks, we could observe PLZF-positive spermatogonia in Gfra1 D/D ; Tg-Nanos2 testes (Fig. 5A, green bar, Supporting Information Fig. 6G) but not in Gfra1 D/D

7 286 Relationship Between NANOS2 and GDNF Signaling Figure 4. Testicular histology of Gfra1-conditional knockout mice with or without Nanos2-overexpression. (A L): Testes from each mutant were immunostained with the indicated markers. Scale bar ¼ 100 lm. Abbreviations: DAPI, 40,6-diamidino-2-phenylindole; GFRA1, glial cell line-derived neurotrophic factor family receptor a 1; PLZF, promyelocytic leukemia zinc finger; Tg, transgene. testes (Fig. 5A, blue bar, Supporting Information Fig. 6C). Consistently, GFP-positive cells were maintained in the 6-week- or 8-week-old Gfra1-cKO testes only when the Tg-Nanos2 was induced (Fig. 5B, green and blue bars, Supporting Information Fig. 6D, 6H). Eventually, Gfra1D/D; TgNanos2 mice lost their germ cells including PLZF-positive spermatogonia by 12 weeks (Fig. 4L). From these results, we conclude that a partial or a short-term rescue of the Gfra1cKO phenotype can be achieved by Nanos2-overexpression. Apoptotic Cell Death Is Not Responsible for the Loss of Spermatogonial Stem Cells in the Gfra1-Knockout Testes The stem cell state or stemness is regulated via the following cellular processes: (a) cell proliferation to expand the stem cell population; (b) suppression of differentiation to maintain stem cells in undifferentiated state; and (c) cell death/survival to control their quality and quantity. GDNF signaling might control these cellular events in a NANOS2-dependent or -independent manner. To address which cellular event(s) are affected by the knockout of Gfra1 and rescued by Nanos2-overexpression, we compared four different genotypes shown above. First, we quantified the apoptosis of PLZF- (Supporting Information Fig. 9A) and GFP (Dfloxed Gfra1)- (Fig. 6A) positive cells immediately after the loss of Gfra1. At 5 weeks of age, apoptotic cells were detected at a very low frequency in the control testes and their numbers were not altered by Gfra1 deletion (Fig. 6A, Supporting Information Fig. 7A, 7B). In the Nanos2-overexpressing testes, regardless of the presence or absence of Gfra1, we observed an increased apoptosis in spermatocytes and a part of spermatogonia (Supporting Information Fig. 7C 7G), indicating that Nanos2-overexpression by the ubiquitous Cre-Ert2 may cause inappropriate effects on spermatogenic germ cells. Nevertheless, there were no significant differences in the proportion of cleaved PARP-positive cells within PLZF- (Supporting Information Fig. 9A) or GFP- (Fig. 6A) positive undifferentiated spermatogonia in any comparison we examined. We obtained similar results

8 Sada, Hasegawa, Pin et al. 287 Figure 5. Temporal changes in the number of spermatogonia in each mutant. The numbers of PLZF-positive (A) or GFP-positive (B) cells per seminiferous tubule were counted at the indicated time points (N=3). The average numbers were normalized using the control of each stage (white bars). **, p <.01; *, p <.05. Abbreviations: GFP, green fluorescent protein; GFRA1, glial cell line-derived neurotrophic factor family receptor alpha 1; PLZF, promyelocytic leukemia zinc finger; Tg, transgene. by using testes from 8-week-old mice (Supporting Information Fig. 10A). From these results, we ruled out the possibility that Nanos2-overexpression rescued the stem cell loss phenotype of Gfra1-cKO testes by interfering with the apoptotic pathway. Overexpressed Nanos2 Cannot Rescue the Reduced Proliferation of Gfra1-Knockout Spermatogonia Given that GDNF signaling is implicated in the proliferation of spermatogonial stem cells [20, 23 25, 28, 38], we next addressed whether Nanos2-overexpression could rescue the proliferation defects of Gfra1-cKO mice. We quantified the proliferation of spermatogonial stem cells using phosphohistone H3 (PH3) as a marker of cell proliferation (Fig. 6B, Supporting Information Fig. 9B). Normally (i.e., in Gfra1 D/þ testes), 30.9% of GFP-positive spermatogonia were positive for PH3 (Fig. 6B, Supporting Information Fig. 8A, 8B), whereas only 8.7% of Gfra1-cKO cells showed the signal (Fig. 6B, Supporting Information Fig. 8C, 8D). Hence, we confirmed that the proliferation of spermatogonial stem cells was highly dependent on GDNF-stimulated pathway. Upon the overexpression of Nanos2, we found a significant reduction of proliferation in PLZF- (Supporting Information Fig. 9B) or GFP-positive cells (Fig. 6B) regardless of the presence or absence of a functional Gfra1 allele. Significantly, in the Gfra1 D/D ; Tg-Nanos2 mice, the proliferation of GFPpositive cells constituted only 1.5% of the total GFP-positive cells (Fig. 6B). Similar results were obtained by using testes from 8-week-old mice (Supporting Information Fig. 10B). Hence, spermatogonial proliferation might not be the reason why Nanos2-overexpression could rescue the stem cell loss phenotype of Gfra1-cKO mice. Rather, NANOS2 and GDNF signaling might regulate the proliferation of spermatogonial stem cells in different pathways: GDNF signaling stimulates the proliferation via known [23, 24, 27, 28, 30] or unknown pathways, while NANOS2 might delay cell cycle progression of the stem cells independent of GDNF signaling. Overexpression of Nanos2 Blocks Stem Cell Differentiation Caused by cko of Gfra1 The stem cell loss is also caused by the inability of stem cells to maintain an undifferentiated state, which may result in the depletion of the stem cell population through differentiation. Indeed, Gdnf-overexpression suppresses differentiation of spermatogonia, and the deletion of Gdnf or its receptors promotes differentiation of spermatogonia and causes the stem cell loss phenotype in vivo and in vitro [19, 20, 38]. To test if Nanos2-overexpression could suppress differentiation of Gfra1-deficient stem cells and contribute to the stem cell maintenance, we evaluated the differentiation state of Gfra1- deficient stem cells with or without overexpressed Nanos2. We performed whole-mount immunostaining for GFP (Dfloxed Gfra1) and NANOS3 at 5 weeks (Fig. 7). In control low or testes, NANOS3 expression was much higher in GFP spermatogonia (morphologically defined as A al ) than in GFP þ spermatogonia (morphologically defined as A s or A pr ) (Fig. 7A). In the Gfra1-deficient testes, however, most GFPpositive spermatogonia, including A s or A pr expressed NANOS3 strongly (Fig. 7B). Quantitative analyses revealed that more than 80% of GFP-positive spermatogonial cluster in the Gfra1-cKO testes exhibited a strong NANOS3 signal, while

9 288 Relationship Between NANOS2 and GDNF Signaling Figure 6. Quantification of apoptosis and proliferation. The rates of apoptotic (A) and proliferative (B) spermatogonia were quantified within GFP-positive populations in 5-week-old mice (N=3). **, p <.01 and *, p <.05. Abbreviations: GFP, green fluorescent protein; GFRA1, glial cell line-derived neurotrophic factor family receptor a 1; PARP, poly (ADP-ribose) polymerase; PH3, phospho histone H3; Tg, transgene. 37.0% of GFP-positive cells in the control testes showed the signal (Fig. 7E). Consistent with this result, the proportion of NANOS3-positive cells was increased in the entire PLZF-positive spermatogonia upon the cko of Gfra1 (Supporting Information Fig. 11A, 11B, 11E). These results suggest that GDNF signaling acts as a suppressive factor on differentiation of spermatogonial stem cells. On the contrary, Nanos2-overexpression decreased the level of NANOS3 expression in spermatogonia both in PLZF- (Supporting Information Fig. 11C, 11E) and GFP- (Fig. 7C, 7E) positive population. Notably, the elevated NANOS3 expression caused by Gfra1 deletion was suppressed in the cells overexpressing Nanos2 (Fig. 7D, 7E, Supporting Information Fig. 11D, 11E). In the Gfra1 D/D ; Tg-Nanos2 mice, only 9.8% of GFP-positive cells showed the NANOS3 signals (Fig. 7E). Hence, overexpressed Nanos2 can delay the differentiation progression of spermatogonial stem cells caused by Gfra1 deficiency. Taken together, we conclude that NANOS2 acts downstream of GDNF signaling to suppress the differentiation of spermatogonial stem cells. DISCUSSION NANOS2 Is a Possible Target of GDNF Signaling and Suppresses Differentiation of Spermatogonial Stem Cells In vivo studies showed that deletion of GDNF signaling components [20] (Figs. 5, 7) or Nanos2 [13] caused a precocious differentiation of spermatogonia and subsequent loss of stem Figure 7. Forced expression of Nanos2 suppresses differentiation of Gfra1-knockout spermatogonia. (A D): Whole-mount immunostaining of 5-week-old testes with the indicated markers. Scale bar ¼ 100 lm. (E): Frequency of NANOS3-positive spermatogonial clusters (2 n cells: 1, 2, 4, 8, 16) in GFP-positive clusters was counted by whole-mount immunostainig of tubules (n ¼ 3). **, p <.01. Abbreviations: A s,a single ;A pr,a paired ;A al,a aligned ; GFP, green fluorescent protein; GFRA1, glial cell line-derived neurotrophic factor family receptor a 1; Tg, transgene. cell population. In cultured spermatogonia, several genes related to differentiation, for example, Ngn3 or Kit, were upregulated by GDNF/GFRA1 removal or Gfra1-knockdown experiments [29, 38]. In contrast, overexpression of Gdnf resulted in the accumulation of undifferentiated spermatogonia due to a blocked differentiation [19]. Similarly, Nanos2-overexpression using Nanos3- Cre line showed that NANOS2 cell autonomously maintained spermatogonia as NANOS3 - Ngn3 undifferentiated state in the presence of GFRA1 [13]. Therefore, GDNF signaling and NANOS2 might have suppressive roles for initial/early step of spermatogonial differentiation, probably at the transition point from NANOS2 þ GFRA1 þ to NANOS3 þ Ngn3 þ.

10 Sada, Hasegawa, Pin et al. 289 In this study, we used several combinations of mutant mice and examined the interplay between NANOS2 and GDNF signaling (summarized in Supporting Information Fig. 12). We found that NANOS2 expression was dependent on GDNF signaling pathway, as cko of Gfra1 resulted in an immediate loss of NANOS2 expression (Fig. 3E, 3F, 3J). When we manipulated NANOS2 to be continuously expressed after the knockout of Gfra1, NANOS2 suppressed differentiation of Gfra1-null spermatogonia (Fig. 7) and maintained the stem cell population for a longer period when compared with simple Gfra1-null spermatogonia (Fig. 5). NANOS2 also has an ability to repress the differentiation of GFRA1-negative, Ngn3-positive population (Fig. 2E 2J). From these results, we propose that NANOS2 is one of the strong candidates of downstream factor of GDNF signaling with respect to the suppression of spermatogonial stem cell differentiation. Although the relationship between GDNF signaling and NANOS2 begins to be understood, we need further studies to answer the following questions. (a) Although NANOS3 expression was increased in Gfra1-cKO cells, it is still unclear whether these cells retain the ability to enter normal differentiation pathway just like endogenous NANOS3 þ Ngn3 þ population. The long-term lineage tracing experiments using ubiquitous lineage tracing marker will be needed to tell the fate of the mutant cells. (b) We still do not know whether GDNF signaling directly regulates NANOS2 transcription/translation or indirectly affects NANOS2 expression as a result of maintaining proper cellular state of spermatogonial stem cells. We need to test whether NANOS2 expression is upregulated in the Gdnfoverexpressing mice; if so, the deletion of Nanos2 cause spermatogonial cell loss, due to de-repression of differentiation even in the presence of overexpressed Gdnf. The answers to these questions will surely lead to better understanding of the molecular mechanisms in spermatogonial stem cell regulation. Opposite Roles of NANOS2 and GDNF Signaling in the Proliferation of Spermatogonial Stem Cells Given that Nanos2-overexpression could not rescue the phenotype of Gfra1-cKO mice completely (Figs. 4, 5), GDNF signaling might also have a NANOS2-independent pathway to maintain spermatogonial stem cells. A possible explanation is that the proliferation of spermatogonial stem cells is highly dependent on the GDNF-stimulated pathway. It is supported by the analyses in our Gfra1-cKO mice (Fig. 6B) or Gdnf-, Gfra1- and Ret-null mice [20], in which the spermatogonial proliferation is significantly reduced. It is also shown that, the GDNF and its downstream factors are essential for the growth of spermatogonial stem cells in culture [23 25, 28, 38 40]. On the other hand, our previous [13] and current studies (Figs. 2B, 6B, Supporting Information Fig. 9B) revealed that Nanos2-overexpressing cells showed a lower proliferation rate than that of control spermatogonia. As Gfra1 is required for the promotion of proliferation of spermatogonial stem cells and the proliferation is further decreased by the presence of overexpressed Nanos2, the number of GFP (Dfloxed Gfra1)- positive cells in the Gfra1 D/D ; Tg-Nanos2 mice gradually reduced (Fig. 5B) due to the significantly lower growth rate (Fig. 6B) and normal level of apoptosis (Fig. 6A). The suppressive role of NANOS2 on spermatogonial proliferation is an interesting feature of the stem cell regulation. Because, it has long been proposed that stem cells divide infrequently and are predominantly quiescent in several other tissues [41, 42]. However, it remains unknown (a) differences between NANOS2 þ GFRA1 þ versus NANOS3 þ Ngn3 þ population in terms of cell division frequency; (b) a link between slow-cycling nature and stem cell activity/potential in spermatogonial stem cells; and (c) crosstalk between pathways for regulating proliferation and differentiation. We will address these issues as future works, especially by focusing on the identification of NANOS2 targets, which are specifically involved in the regulation of either differentiation or cell cycle/division. Nanos2-Overexpression Phenotypes by Using Different Cre Lines In our previous study, we used Nanos3-Cre to induce Nanos2-overexpression, in which Cre expression commences in all embryonic male gonocytes [13]. After birth, the male gonocytes resume proliferation and migrate from their original central position toward the periphery in the seminiferous tubules and transform into spermatogonia by P7 [1, 3, 43]. We think that Nanos3-Cre-driven Nanos2-overexpression blocks differentiation of spermatogonia, and the Nanos2- overexpressing spermatogonia never proceed from NANO- S2 þ GFRA1 þ into NANOS3 þ Ngn3 þ state (Supporting Information Fig. 12B). Consistent with this idea, the majority of Nanos3-driven Nanos2-overexpressing cells were identified as GFRA1-positive, A s or A pr cells in the adult stage [13]. As shown in the results of lineage tracing experiment of Ngn3-Cre, most of GFRA1-positive cells are not marked by Ngn3-Cre in either P7 (Supporting Information Fig. 2) or adult (Fig. 1A 1F) stage, indicating that Cre recombination occurred in the cells that have already exited from NANO- S2 þ GFRA1 þ state and have become NANOS3 þ Ngn3 þ. Consistently, the Ngn3-driven Nanos2-overexpressing cells in the adult testes exhibited A al -like properties reflecting the characteristic of Ngn3-expressing cells (Fig. 1G 1R). However, the effects of Nanos2-overexpression by Ngn3-Cre were similar to that observed in the Tg-Nanos2/Nanos3-Cre mice: NANOS2 slows down proliferation and suppresses cell differentiation (Fig. 2, Supporting Information Fig. 12C). Nanos2-overexpression induced via Cre-Ert2 occurred in the entire undifferentiated spermatogonia (and much broader cells) (Supporting Information Figs. 5, 12E, 12F). In both Gfra1 D/þ ; Tg-Nanos2 and Gfra1 D/D ; Tg-Nanos2 mice, GFP (Dfloxed Gfra1)-positive cells were A s or A pr and showed no NANOS3 expression (Fig. 7) similar to the case of Tg- Nanos2/Nanos3-Cre [13]. Furthermore, similar to the case of Ngn3-Cre, Cre-Ert2 driven Nanos2-overexpression might suppress differentiation of GFRA1-negative (NANOS3 þ Ngn3 þ ) population, as shown by NANOS3 downregulation in the entire PLZF-positive population (Supporting Information Fig. 11). The proliferation rate was also decreased in both GFP- and PLZF-positive cells by Cre-Ert2-driven Nanos2-overexpression irrespective of the presence or absence of Gfra1 (Fig. 6B, Supporting Information Fig. 9B). Together, these results strongly support our idea that NANOS2 can repress differentiation and proliferation of both GFRA1-positive cells and the cells that have lost GFRA1 expression, either by Gfra1-knockout or in a process of normal differentiation into NANOS3 þ Ngn3 þ state. Stem Cells in Murine Spermatogenesis Although studies using functional transplantation assays along with lineage tracing experiments have demonstrated that the stem cell activity/potential is highly enriched in the undifferentiated spermatogonial population, an unequivocal identification of the stem cells within the subpopulation of undifferentiated spermatogonia is still under argument [6, 7, 9, 12, 13, 44]. In particular, several studies indicate that GFRA1 selection did not result in the enrichment of transplantable stem cells in adult testes [6, 12]. However, we denoted GFRA1 þ NANOS2 þ spermatogonia as stem cells in this study, as our previous long-term lineage tracing analysis indicated that NANOS2-expressing spermatogonia (frequently coexpress

11 290 Relationship Between NANOS2 and GDNF Signaling GFRA1) acted as spermatogonial stem cells in vivo and the lack of Nanos2 led to a stem cell loss phenotype [13]. In addition, we showed for the first time that cko of Gfra1 gene in the adult stage caused a depletion of spermatogonia, followed by a complete loss of spermatogenic germ cells (Figs. 4, 5), strongly indicating that GFRA1-positive spermatogonia constitute the stem cell population. As indicated by the recent study [44], one cannot exclude the possibility that ex vivo manipulation of germ cells and pretreatment of recipients involved in functional transplantation assays may have altered the isolated donor cells or the recipient tissues, which might affect cell behavior. Future experiments are required to determine the fates of GFRA1-positive cells in intact testes by using an inducible Cre-loxP system. CONCLUSIONS This study shows a genetic interaction of NANOS2 and GDNF signaling for the maintenance of stem cells during murine spermatogenesis. We suggest that NANOS2 maintains the undifferentiated state of spermatogonial stem cells downstream of GDNF signaling. Our results offer a novel insight into the understanding of how spermatogonial stem cells are regulated by stem cell-intrinsic and -extrinsic mechanisms. ACKNOWLEDGMENTS We thank Dr. H. Enomoto for the Gfra1 floxed mice; Dr. S. Yoshida for the Ngn3-Cre transgenic mice; R. Kopan for the Rosa-YFP mice; Dr. Y. Nishimune for the TRA98 antibody; and Drs. S. Chuma and N. Nakatsuji for the SCP3 antibody. This work was supported in part by Grants-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology (Japan) to Y.S. and a Research Fellowship for Young Scientists from the Japan Society for the Promotion of Science to A.S. DISCLOSURE OF POTENTIAL CONFLICT OF INTEREST The authors indicate no potential conflicts of interests. REFERENCES 1 de Rooij DG. Stem cells in the testis. Int J Exp Pathol 1998;79: de Rooij DG, Grootegoed JA. Spermatogonial stem cells. Curr Opin Cell Biol 1998;10: de Rooij DG, Russell LD. All you wanted to know about spermatogonia but were afraid to ask. J Androl 2000;21: de Rooij DG. Proliferation and differentiation of spermatogonial stem cells. Reproduction 2001;121: Russell L, Ettlin R, Sinha Hikim A et al. Histological and Histopathological Evaluation of the Testis. Clearwater, FL: Cache River Press, Grisanti L, Falciatori I, Grasso M et al. Identification of spermatogonial stem cell subsets by morphological analysis and prospective isolation. Stem Cells 2009;27: Nakagawa T, Sharma M, Nabeshima Y et al. Functional hierarchy and reversibility within the murine spermatogenic stem cell compartment. Science 2010;328: Suzuki H, Sada A, Yoshida S et al. The heterogeneity of spermatogonia is revealed by their topology and expression of marker proteins including the germ cell-specific proteins Nanos2 and Nanos3. Dev Biol 2009;336: Tokuda M, Kadokawa Y, Kurahashi H et al. CDH1 is a specific marker for undifferentiated spermatogonia in mouse testes. Biol Reprod 2007;76: Zheng K, Wu X, Kaestner KH et al. The pluripotency factor LIN28 marks undifferentiated spermatogonia in mouse. BMC Dev Biol 2009; 9: Klein AM, Nakagawa T, Ichikawa R et al. Mouse germ line stem cells undergo rapid and stochastic turnover. Cell Stem Cell 2010;7: Oatley JM, Brinster RL. Regulation of spermatogonial stem cell selfrenewal in mammals. Annu Rev Cell Dev Biol 2008;24: Sada A, Suzuki A, Suzuki H et al. The RNA-binding protein NANOS2 is required to maintain murine spermatogonial stem cells. Science 2009;325: Atchison FW, Means AR. Spermatogonial depletion in adult Pin1- deficient mice. Biol Reprod 2003;69: Buaas FW, Kirsh AL, Sharma M et al. Plzf is required in adult male germ cells for stem cell self-renewal. Nat Genet 2004;36: Chen C, Ouyang W, Grigura V et al. ERM is required for transcriptional control of the spermatogonial stem cell niche. Nature 2005;436: Costoya JA, Hobbs RM, Barna M et al. Essential role of Plzf in maintenance of spermatogonial stem cells. Nat Genet 2004;36: Falender AE, Freiman RN, Geles KG et al. Maintenance of spermatogenesis requires TAF4b, a gonad-specific subunit of TFIID. Genes Dev 2005;19: Meng X, Lindahl M, Hyvonen ME et al. Regulation of cell fate decision of undifferentiated spermatogonia by GDNF. Science 2000;287: Naughton CK, Jain S, Strickland AM et al. Glial cell-line derived neurotrophic factor-mediated RET signaling regulates spermatogonial stem cell fate. Biol Reprod 2006;74: Schlesser HN, Simon L, Hofmann MC et al. Effects of ETV5 (ets variant gene 5) on testis and body growth, time course of spermatogonial stem cell loss, and fertility in mice. Biol Reprod 2008;78: Takubo K, Ohmura M, Azuma M et al. Stem cell defects in ATMdeficient undifferentiated spermatogonia through DNA damageinduced cell-cycle arrest. Cell Stem Cell 2008;2: Braydich-Stolle L, Kostereva N, Dym M et al. Role of Src family kinases and N-Myc in spermatogonial stem cell proliferation. Dev Biol 2007;304: He Z, Jiang J, Kokkinaki M et al. Gdnf upregulates c-fos transcription via the Ras/Erk1/2 pathway to promote mouse spermatogonial stem cell proliferation. Stem Cells 2008;26: Hofmann MC, Braydich-Stolle L, Dym M. Isolation of male germline stem cells; influence of GDNF. Dev Biol 2005;279: Hofmann MC. Gdnf signaling pathways within the mammalian spermatogonial stem cell niche. Mol Cell Endocrinol 2008;288: Lee J, Kanatsu-Shinohara M, Inoue K et al. Akt mediates self-renewal division of mouse spermatogonial stem cells. Development 2007;134: Lee J, Kanatsu-Shinohara M, Morimoto H et al. Genetic reconstruction of mouse spermatogonial stem cell self-renewal in vitro by Rascyclin D2 activation. Cell Stem Cell 2009;5: Oatley JM, Avarbock MR, Telaranta AI et al. Identifying genes important for spermatogonial stem cell self-renewal and survival. Proc Natl Acad Sci USA 2006;103: Oatley JM, Avarbock MR, Brinster RL. Glial cell line-derived neurotrophic factor regulation of genes essential for self-renewal of mouse spermatogonial stem cells is dependent on Src family kinase signaling. J Biol Chem 2007;282: Wu X, Oatley JM, Oatley MJ et al. The POU domain transcription factor POU3F1 is an important intrinsic regulator of GDNF-induced survival and self-renewal of mouse spermatogonial stem cells. Biol Reprod 2010;82: Liu Z, Turkoz A, Jackson EN et al. Notch1 loss of heterozygosity causes vascular tumors and lethal hemorrhage in mice. J Clin Invest 2011;121: Yoshida S, Takakura A, Ohbo K et al. Neurogenin3 delineates the earliest stages of spermatogenesis in the mouse testis. Dev Biol 2004;269: Uesaka T, Jain S, Yonemura S et al. Conditional ablation of GFRalpha1 in postmigratory enteric neurons triggers unconventional

Testicular stem cells

Testicular stem cells Testicular stem cells Dirk G. de Rooij Department of Endocrinology Faculty of Biology, Utrecht University 1. Knowledge on the development of the spermatogenic stem cell lineage 2. Principals of the nature

More information

SUPPLEMENTAL INFORMATION FOR. PAX7 expression defines germline stem cells in the adult testis

SUPPLEMENTAL INFORMATION FOR. PAX7 expression defines germline stem cells in the adult testis SUPPLEMENTAL INFORMATION FOR PAX7 expression defines germline stem cells in the adult testis Gina M. Aloisio, Yuji Nakada, Hatice D. Saatcioglu, Christopher G. Peña, Michael D. Baker, Edward D. Tarnawa,

More information

Maintaining the male germline: regulation of spermatogonial stem cells

Maintaining the male germline: regulation of spermatogonial stem cells 133 REVIEW Maintaining the male germline: regulation of spermatogonial stem cells Kyle Caires, Johnathan Broady and Derek McLean Department of Animal Sciences, Center for Reproductive Biology, Washington

More information

Spermatogenic Stem Cell System in the Mouse Testis

Spermatogenic Stem Cell System in the Mouse Testis Spermatogenic Stem Cell System in the Mouse Testis S. YOSHIDA Division of Germ Cell Biology, National Institute for Basic Biology, Higashiyama, Myodaiji, Okazaki 444-8787, Okazaki, Japan Mouse spermatogenesis

More information

TISSUE-SPECIFIC STEM CELLS

TISSUE-SPECIFIC STEM CELLS TISSUE-SPECIFIC STEM CELLS LIN28A Marks the Spermatogonial Progenitor Population and Regulates Its Cyclic Expansion PAPIA CHAKRABORTY, a F. WILLIAM BUAAS, a MANJU SHARMA, a ELIZABETH SNYDER, a DIRK G.

More information

TO BE OR NOT TO BE: THE MOLECULAR MECHANISMS THAT REGULATE SPERMATOGONIAL STEM CELL FATE

TO BE OR NOT TO BE: THE MOLECULAR MECHANISMS THAT REGULATE SPERMATOGONIAL STEM CELL FATE TO BE OR NOT TO BE: THE MOLECULAR MECHANISMS THAT REGULATE SPERMATOGONIAL STEM CELL FATE By FRIEDA K. CHAN A dissertation submitted in partial fulfillment of the requirements for the degree of DOCTOR OF

More information

TISSUE-SPECIFIC STEM CELLS

TISSUE-SPECIFIC STEM CELLS TISSUE-SPECIFIC STEM CELLS TAF4b is Required for Mouse Spermatogonial Stem Cell Development LINDSAY A. LOVASCO, a ERIC A. GUSTAFSON, a KIMBERLY A. SEYMOUR, a DIRK G. DE ROOIJ, b RICHARD N. FREIMAN a Key

More information

Impact of Sox9 Dosage and Hes1-mediated Notch Signaling in Controlling the Plasticity of Adult Pancreatic Duct Cells in Mice

Impact of Sox9 Dosage and Hes1-mediated Notch Signaling in Controlling the Plasticity of Adult Pancreatic Duct Cells in Mice Impact of Sox9 Dosage and Hes1-mediated Notch Signaling in Controlling the Plasticity of Adult Pancreatic Duct Cells in Mice Shinichi Hosokawa 1,3,Kenichiro Furuyama 1,3, Masashi Horiguchi 1,3,Yoshiki

More information

Development Supplementary information

Development Supplementary information Supplemental Materials and Methods Mosaic clonal analysis GSC and SP clones were induced with the FLP/FRT-mediated mitotic recombination technique (Xu and Rubin, 1993) in files with following genotypes:

More information

The first round of mouse spermatogenesis is a distinctive program that lacks the self-renewing spermatogonia stage

The first round of mouse spermatogenesis is a distinctive program that lacks the self-renewing spermatogonia stage RESEARCH ARTICLE 1495 Development 133, 1495-1505 (2006) doi:10.1242/dev.02316 The first round of mouse spermatogenesis is a distinctive program that lacks the self-renewing spermatogonia stage Shosei Yoshida

More information

Supplemental Information. Otic Mesenchyme Cells Regulate. Spiral Ganglion Axon Fasciculation. through a Pou3f4/EphA4 Signaling Pathway

Supplemental Information. Otic Mesenchyme Cells Regulate. Spiral Ganglion Axon Fasciculation. through a Pou3f4/EphA4 Signaling Pathway Neuron, Volume 73 Supplemental Information Otic Mesenchyme Cells Regulate Spiral Ganglion Axon Fasciculation through a Pou3f4/EphA4 Signaling Pathway Thomas M. Coate, Steven Raft, Xiumei Zhao, Aimee K.

More information

TISSUE-SPECIFIC STEM CELLS

TISSUE-SPECIFIC STEM CELLS TISSUE-SPECIFIC STEM CELLS The Transition from Stem Cell to Progenitor Spermatogonia and Male Fertility Requires the SHP2 Protein Tyrosine Phosphatase PAWAN PURI, a BART T. PHILLIPS, a HITOMI SUZUKI, a

More information

Supplemental Figure 1. (A) The localization of Cre DNA recombinase in the testis of Cyp19a1-Cre mice was detected by immunohistchemical analyses

Supplemental Figure 1. (A) The localization of Cre DNA recombinase in the testis of Cyp19a1-Cre mice was detected by immunohistchemical analyses Supplemental Figure 1. (A) The localization of Cre DNA recombinase in the testis of Cyp19a1-Cre mice was detected by immunohistchemical analyses using an anti-cre antibody; testes at 1 week (left panel),

More information

Functional and molecular features of the Id4 + germline stem cell population in mouse testes

Functional and molecular features of the Id4 + germline stem cell population in mouse testes Functional and molecular features of the Id4 + germline stem cell population in mouse testes Frieda Chan, 1 Melissa J. Oatley, 1 Amy V. Kaucher, 1 Qi-En Yang, 1 Charles J. Bieberich, 2 Cooduvalli S. Shashikant,

More information

THE EFFECT OF RESTORING GDNF SIGNALING ON SPERMATOGONIAL STEM CELL DIFFERENTIATION

THE EFFECT OF RESTORING GDNF SIGNALING ON SPERMATOGONIAL STEM CELL DIFFERENTIATION THE EFFECT OF RESTORING GDNF SIGNALING ON SPERMATOGONIAL STEM CELL DIFFERENTIATION by Andrew J. Laychur A thesis submitted to the Johns Hopkins University in conformity with the requirements for the degree

More information

Quantification of early stage lesions for loss of p53 should be shown in the main figures.

Quantification of early stage lesions for loss of p53 should be shown in the main figures. Reviewer #1 (Remarks to the Author): Expert in prostate cancer The manuscript "Clonal dynamics following p53 loss of heterozygosity in Kras-driven cancers" uses a number of novel genetically engineered

More information

Recent developments in the spermatogonial stem cell field

Recent developments in the spermatogonial stem cell field DOI: 10.21451/1984-3143-AR890 Anim. Reprod., v.14, n.1, p.82-88, Jan./Mar. 2017 Recent developments in the spermatogonial stem cell field Dirk G. de Rooij 1 Reproductive Biology Group, Division of Developmental

More information

Supplementary Figure 1 Madm is not required in GSCs and hub cells. (a,b) Act-Gal4-UAS-GFP (a), Act-Gal4-UAS- GFP.nls (b,c) is ubiquitously expressed

Supplementary Figure 1 Madm is not required in GSCs and hub cells. (a,b) Act-Gal4-UAS-GFP (a), Act-Gal4-UAS- GFP.nls (b,c) is ubiquitously expressed Supplementary Figure 1 Madm is not required in GSCs and hub cells. (a,b) Act-Gal4-UAS-GFP (a), Act-Gal4-UAS- GFP.nls (b,c) is ubiquitously expressed in the testes. The testes were immunostained with GFP

More information

Problem Set 8 Key 1 of 8

Problem Set 8 Key 1 of 8 7.06 2003 Problem Set 8 Key 1 of 8 7.06 2003 Problem Set 8 Key 1. As a bright MD/PhD, you are interested in questions about the control of cell number in the body. Recently, you've seen three patients

More information

Supplementary Figure 1: Hsp60 / IEC mice are embryonically lethal (A) Light microscopic pictures show mouse embryos at developmental stage E12.

Supplementary Figure 1: Hsp60 / IEC mice are embryonically lethal (A) Light microscopic pictures show mouse embryos at developmental stage E12. Supplementary Figure 1: Hsp60 / IEC mice are embryonically lethal (A) Light microscopic pictures show mouse embryos at developmental stage E12.5 and E13.5 prepared from uteri of dams and subsequently genotyped.

More information

SUPPLEMENTARY FIGURES

SUPPLEMENTARY FIGURES SUPPLEMENTARY FIGURES 1 Supplementary Figure 1, Adult hippocampal QNPs and TAPs uniformly express REST a-b) Confocal images of adult hippocampal mouse sections showing GFAP (green), Sox2 (red), and REST

More information

Hematopoiesis. - Process of generation of mature blood cells. - Daily turnover of blood cells (70 kg human)

Hematopoiesis. - Process of generation of mature blood cells. - Daily turnover of blood cells (70 kg human) Hematopoiesis - Process of generation of mature blood cells - Daily turnover of blood cells (70 kg human) 1,000,000,000,000 total cells 200,000,000,000 red blood cells 70,000,000,000 neutrophils Hematopoiesis

More information

Spermatogenesis. What is it and what does it look like? How do hormones regulate spermatogenesis?

Spermatogenesis. What is it and what does it look like? How do hormones regulate spermatogenesis? Spermatogenesis What is it and what does it look like? How do hormones regulate spermatogenesis? FSH, androgens, growth factors Animal Physiology (Hill, Wise, Anderson): Ch. 15 435-438 1 Spermatogenesis:

More information

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION DOI: 10.1038/ncb2697 Figure S1 Cytokeratin 5 is a specific marker for basal and intermediate cells in all mouse prostate lobes. (a) Immunofluorescence staining showing co-localization of YFP with p63 in

More information

Figure S1. (A) Schematic diagram of dnrar transgene allele. (B) X-Gal staining of testis from

Figure S1. (A) Schematic diagram of dnrar transgene allele. (B) X-Gal staining of testis from Figure S1. (A) Schematic diagram of dnrar transgene allele. (B) X-Gal staining of testis from germ cell mutants (dnrar flox/flox, Stra8-Cre +, RARElacZ) (A ), controls (dnrar flox/flox, RARElacZ) (B ),

More information

Sami Ventelä, 1,2 Hiroshi Ohta, 3 Martti Parvinen, 2 and Yoshitake Nishimune 3

Sami Ventelä, 1,2 Hiroshi Ohta, 3 Martti Parvinen, 2 and Yoshitake Nishimune 3 BIOLOGY OF REPRODUCTION 66, 1422 1429 (2002) Development of the Stages of the Cycle in Mouse Seminiferous Epithelium after Transplantation of Green Fluorescent Protein-Labeled Spermatogonial Stem Cells

More information

PAX7 expression defines germline stem cells in the adult testis

PAX7 expression defines germline stem cells in the adult testis The Journal of Clinical Investigation PAX7 expression defines germline stem cells in the adult testis Gina M. Aloisio, 1 Yuji Nakada, 1 Hatice D. Saatcioglu, 1 Christopher G. Peña, 1 Michael D. Baker,

More information

SUPPLEMENTARY FIG. S2. Representative counting fields used in quantification of the in vitro neural differentiation of pattern of dnscs.

SUPPLEMENTARY FIG. S2. Representative counting fields used in quantification of the in vitro neural differentiation of pattern of dnscs. Supplementary Data SUPPLEMENTARY FIG. S1. Representative counting fields used in quantification of the in vitro neural differentiation of pattern of anpcs. A panel of lineage-specific markers were used

More information

Germ Cell Transplantation in Fish

Germ Cell Transplantation in Fish Larvi 2009 Germ Cell Transplantation in Fish Goro Yoshizaki (Tokyo University of Marine Science and Technology, SORST/JST) Tuna Mackerel Body weight; 300 kg 300 g Body length; 3 m 30 cm Scombridae family

More information

SUPPLEMENTARY DATA. Supplementary Table 2. Antibodies used for Immunofluoresence. Supplementary Table 3. Real-time PCR primer sequences.

SUPPLEMENTARY DATA. Supplementary Table 2. Antibodies used for Immunofluoresence. Supplementary Table 3. Real-time PCR primer sequences. Supplementary Table 2. Antibodies used for Immunofluoresence. Antibody Dilution Source Goat anti-pdx1 1:100 R&D Systems Rabbit anti-hnf6 1:100 Santa Cruz Biotechnology Mouse anti-nkx6.1 1:200 Developmental

More information

Introduction. K. Zohni 1, X. Zhang 1, S.L. Tan 1, P. Chan 2, and M.C. Nagano 1, * ORIGINAL ARTICLE Andrology

Introduction. K. Zohni 1, X. Zhang 1, S.L. Tan 1, P. Chan 2, and M.C. Nagano 1, * ORIGINAL ARTICLE Andrology Human Reproduction, Vol.27, No.1 pp. 44 53, 2012 Advanced Access publication on November 14, 2011 doi:10.1093/humrep/der357 ORIGINAL ARTICLE Andrology The efficiency of male fertility restoration is dependent

More information

Supporting Information

Supporting Information Supporting Information Franco et al. 10.1073/pnas.1015557108 SI Materials and Methods Drug Administration. PD352901 was dissolved in 0.5% (wt/vol) hydroxyl-propyl-methylcellulose, 0.2% (vol/vol) Tween

More information

Supplemental Figure 1. Western blot analysis indicated that MIF was detected in the fractions of

Supplemental Figure 1. Western blot analysis indicated that MIF was detected in the fractions of Supplemental Figure Legends Supplemental Figure 1. Western blot analysis indicated that was detected in the fractions of plasma membrane and cytosol but not in nuclear fraction isolated from Pkd1 null

More information

Supplemental Data. Wnt/β-Catenin Signaling in Mesenchymal Progenitors. Controls Osteoblast and Chondrocyte

Supplemental Data. Wnt/β-Catenin Signaling in Mesenchymal Progenitors. Controls Osteoblast and Chondrocyte Supplemental Data Wnt/β-Catenin Signaling in Mesenchymal Progenitors Controls Osteoblast and Chondrocyte Differentiation during Vertebrate Skeletogenesis Timothy F. Day, Xizhi Guo, Lisa Garrett-Beal, and

More information

Supplementary Figure 1: Expression of Gli1-lacZ in E17.5 ovary and mesonephros. a,

Supplementary Figure 1: Expression of Gli1-lacZ in E17.5 ovary and mesonephros. a, Supplementary Figure 1: Expression of Gli1-lacZ in E17.5 ovary and mesonephros. a, Transverse sections of E17.5 ovary and mesonephros from Gli1-LacZ reporter embryos (n=3) after LacZ staining (blue). The

More information

a Control IgG Intestine c Testis b Thymus 1 3 2 S S 2 1 3 4 4 Figure S1 The wild-type mouse (C57BL/6J) organs (intestine, thymus and testis) were frozen in liquid nitrogen and sectioned at 5 µm on a cryostat.

More information

Journal Club. 03/04/2012 Lama Nazzal

Journal Club. 03/04/2012 Lama Nazzal Journal Club 03/04/2012 Lama Nazzal NOTCH and the kidneys Is an evolutionarily conserved cell cell communication mechanism. Is a regulator of cell specification, differentiation, and tissue patterning.

More information

Supplementary Materials and Methods

Supplementary Materials and Methods Supplementary Materials and Methods Whole Mount X-Gal Staining Whole tissues were collected, rinsed with PBS and fixed with 4% PFA. Tissues were then rinsed in rinse buffer (100 mm Sodium Phosphate ph

More information

Oct4 Lineage Tracing of Spermatogonial Stem Cells in the Adult Mouse Testes. A Thesis SUBMITTED TO THE FACULTY OF UNIVERSITY OF MINNESOTA BY

Oct4 Lineage Tracing of Spermatogonial Stem Cells in the Adult Mouse Testes. A Thesis SUBMITTED TO THE FACULTY OF UNIVERSITY OF MINNESOTA BY Oct4 Lineage Tracing of Spermatogonial Stem Cells in the Adult Mouse Testes A Thesis SUBMITTED TO THE FACULTY OF UNIVERSITY OF MINNESOTA BY Kerri Tana Haider IN PARTIAL FULFILLMENT OF THE REQUIREMENTS

More information

ICSI with sperm derived from cultured testes (Nature 2011) Establishment of rabbit ips cells (J Biol Chem 2010)

ICSI with sperm derived from cultured testes (Nature 2011) Establishment of rabbit ips cells (J Biol Chem 2010) Efficient production of offspring in wild-derived strains of mice (Biol Reprod in press) (Presented by Keiji Mochida) Birth of offspring from ectopically transplanted PGCs (Biol Reprod 211) ICSI with sperm

More information

(Stratagene, La Jolla, CA) (Supplemental Fig. 1A). A 5.4-kb EcoRI fragment

(Stratagene, La Jolla, CA) (Supplemental Fig. 1A). A 5.4-kb EcoRI fragment SUPPLEMENTAL INFORMATION Supplemental Methods Generation of RyR2-S2808D Mice Murine genomic RyR2 clones were isolated from a 129/SvEvTacfBR λ-phage library (Stratagene, La Jolla, CA) (Supplemental Fig.

More information

EMBO REPORT SUPPLEMENTARY SECTION. Quantitation of mitotic cells after perturbation of Notch signalling.

EMBO REPORT SUPPLEMENTARY SECTION. Quantitation of mitotic cells after perturbation of Notch signalling. EMBO REPORT SUPPLEMENTARY SECTION Quantitation of mitotic cells after perturbation of Notch signalling. Notch activation suppresses the cell cycle indistinguishably both within and outside the neural plate

More information

Supplemental Figure 1. Intracranial transduction of a modified ptomo lentiviral vector in the mouse

Supplemental Figure 1. Intracranial transduction of a modified ptomo lentiviral vector in the mouse Supplemental figure legends Supplemental Figure 1. Intracranial transduction of a modified ptomo lentiviral vector in the mouse hippocampus targets GFAP-positive but not NeuN-positive cells. (A) Stereotaxic

More information

Supporting Online Material for

Supporting Online Material for www.sciencemag.org/cgi/content/full/1171320/dc1 Supporting Online Material for A Frazzled/DCC-Dependent Transcriptional Switch Regulates Midline Axon Guidance Long Yang, David S. Garbe, Greg J. Bashaw*

More information

Induction of spermatogenic synchrony by retinoic acid in neonatal mice

Induction of spermatogenic synchrony by retinoic acid in neonatal mice EDITOR'S Letter to CORNER the Editor Spermatogenesis 3:1, e23180; January/February/March 2013 2013; 2013 Landes Bioscience EDITOR'S CORNER Induction of spermatogenic synchrony by retinoic acid in neonatal

More information

Spermatogonial stem cells: What does the future hold?

Spermatogonial stem cells: What does the future hold? F, V & V IN OBGYN, 2011, 3 (1): 36-40 Viewpoint Spermatogonial stem cells: What does the future hold? H. TOURNAYE, E. GOOSSENS Research unit Biology of the Testis; Department of Embryology and Genetics;

More information

Emx2 patterns the neocortex by regulating FGF positional signaling

Emx2 patterns the neocortex by regulating FGF positional signaling Emx2 patterns the neocortex by regulating FGF positional signaling Tomomi Fukuchi-Shimogori and Elizabeth A Grove Presented by Sally Kwok Background Cerebral cortex has anatomically and functionally distinct

More information

Early cell death (FGF) B No RunX transcription factor produced Yes No differentiation

Early cell death (FGF) B No RunX transcription factor produced Yes No differentiation Solution Key - Practice Questions Question 1 a) A recent publication has shown that the fat stem cells (FSC) can act as bone stem cells to repair cavities in the skull, when transplanted into immuno-compromised

More information

Effective activity of cytokine-induced killer cells against autologous metastatic melanoma including cells with stemness features

Effective activity of cytokine-induced killer cells against autologous metastatic melanoma including cells with stemness features Effective activity of cytokine-induced killer cells against autologous metastatic melanoma including cells with stemness features Loretta Gammaitoni, Lidia Giraudo, Valeria Leuci, et al. Clin Cancer Res

More information

SPOC1 (PHF13) is required for spermatogonial stem cell differentiation and sustained spermatogenesis

SPOC1 (PHF13) is required for spermatogonial stem cell differentiation and sustained spermatogenesis JCS epress online publication date 18 August 2011 Research Article 1 SPOC1 (PHF13) is required for spermatogonial stem cell differentiation and sustained spermatogenesis Annegret Bördlein 1, *, Harry Scherthan

More information

Meeting Report. From December 8 to 11, 2012 at Atlanta, GA, U.S.A

Meeting Report. From December 8 to 11, 2012 at Atlanta, GA, U.S.A Meeting Report Affiliation Department of Transfusion Medicine and Cell Therapy Name Hisayuki Yao Name of the meeting Period and venue Type of your presentation Title of your presentation The 54 th Annual

More information

follicles and spermatogonia

follicles and spermatogonia 5 th World Congress of the International Society for Fertility Preservation Vienna, Austria. November 16-18; 2017 Session 2: Stem cells and in vitro growth of gametes Development, sex differentiation and

More information

Knockout TM SR : ; ; ; : R ; R : A : X(2013) , ,, B. , (Knockout TM

Knockout TM SR : ; ; ; : R ; R : A : X(2013) , ,, B. , (Knockout TM 33 1 Vol.33 No.1 013 1 Dec. 013 Reproduction & Contraception doi: 10.7669/j.issn.03-37X.013.1.0804 E-mail: randc_journal@163.com Knockout TM SR ; ; ; 400014 : FBS Knockout TM SRKSR : FBS KSR HE TUNEL RT-PCR

More information

Studying The Role Of DNA Mismatch Repair In Brain Cancer Malignancy

Studying The Role Of DNA Mismatch Repair In Brain Cancer Malignancy Kavya Puchhalapalli CALS Honors Project Report Spring 2017 Studying The Role Of DNA Mismatch Repair In Brain Cancer Malignancy Abstract Malignant brain tumors including medulloblastomas and primitive neuroectodermal

More information

AP VP DLP H&E. p-akt DLP

AP VP DLP H&E. p-akt DLP A B AP VP DLP H&E AP AP VP DLP p-akt wild-type prostate PTEN-null prostate Supplementary Fig. 1. Targeted deletion of PTEN in prostate epithelium resulted in HG-PIN in all three lobes. (A) The anatomy

More information

Stem Cell Reports Ar ticle

Stem Cell Reports Ar ticle Please cite this article in press as: Takashima et al., Functional Differences between GDNF-Dependent and FGF-Dependent Mouse Spermatogonial Stem Cell Self-Renewal, Stem Cell Reports (5), http://dx.doi.org/.6/j.stemcr.5..

More information

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION SUPPLEMENTARY INFORMATION doi:10.1038/nature11306 Supplementary Figures Supplementary Figure 1. Basic characterization of GFP+ RGLs in the dentate gyrus of adult nestin-gfp mice. a, Sample confocal images

More information

Biology 4361 Developmental Biology. October 11, Multiple choice (one point each)

Biology 4361 Developmental Biology. October 11, Multiple choice (one point each) Biology 4361 Developmental Biology Exam 1 October 11, 2005 Name: ID#: Multiple choice (one point each) 1. Sertoli cells a. surround spermatocytes b. are the structural components of the seminiferous tubules

More information

Supplemental Information. Tissue Myeloid Progenitors Differentiate. into Pericytes through TGF-b Signaling. in Developing Skin Vasculature

Supplemental Information. Tissue Myeloid Progenitors Differentiate. into Pericytes through TGF-b Signaling. in Developing Skin Vasculature Cell Reports, Volume 18 Supplemental Information Tissue Myeloid Progenitors Differentiate into Pericytes through TGF-b Signaling in Developing Skin Vasculature Tomoko Yamazaki, Ani Nalbandian, Yutaka Uchida,

More information

Tissue-dependent consequences of Apc inactivation on proliferation and differentiation of ciliated cell progenitors via Wnt and Notch signaling

Tissue-dependent consequences of Apc inactivation on proliferation and differentiation of ciliated cell progenitors via Wnt and Notch signaling Washington University School of Medicine Digital Commons@Becker Open Access Publications 1-1-2013 Tissue-dependent consequences of Apc inactivation on proliferation and differentiation of ciliated cell

More information

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION b 350 300 250 200 150 100 50 0 E0 E10 E50 E0 E10 E50 E0 E10 E50 E0 E10 E50 Number of organoids per well 350 300 250 200 150 100 50 0 R0 R50 R100 R500 1st 2nd 3rd Noggin 100 ng/ml Noggin 10 ng/ml Noggin

More information

Pattern and Kinetics of Mouse Donor Spermatogonial Stem Cell Colonization in Recipient Testes 1

Pattern and Kinetics of Mouse Donor Spermatogonial Stem Cell Colonization in Recipient Testes 1 BIOLOGY OF REPRODUCTION 60, 1429 1436 (1999) Pattern and Kinetics of Mouse Donor Spermatogonial Stem Cell Colonization in Recipient Testes 1 Makoto Nagano, Mary R. Avarbock, and Ralph L. Brinster 2 School

More information

spermatogonial transplantation (testis/tem cels/spermaoneis/transec mice/fertty)

spermatogonial transplantation (testis/tem cels/spermaoneis/transec mice/fertty) Proc. Natl. Acad. Sci. USA Vol. 91, pp. 11303-11307, November 1994 Developmental Biology Germline transmission of donor haplotype following spermatogonial transplantation (testis/tem cels/spermaoneis/transec

More information

Supplementary Materials for

Supplementary Materials for www.sciencetranslationalmedicine.org/cgi/content/full/4/117/117ra8/dc1 Supplementary Materials for Notch4 Normalization Reduces Blood Vessel Size in Arteriovenous Malformations Patrick A. Murphy, Tyson

More information

Supplementary Figure 1: si-craf but not si-braf sensitizes tumor cells to radiation.

Supplementary Figure 1: si-craf but not si-braf sensitizes tumor cells to radiation. Supplementary Figure 1: si-craf but not si-braf sensitizes tumor cells to radiation. (a) Embryonic fibroblasts isolated from wildtype (WT), BRAF -/-, or CRAF -/- mice were irradiated (6 Gy) and DNA damage

More information

Development, sex differentiation and clonal expansion of PGCs to create primordial follicles and spermatogonia. Scenarios for in vitro gametogenesis

Development, sex differentiation and clonal expansion of PGCs to create primordial follicles and spermatogonia. Scenarios for in vitro gametogenesis 5 th World Congress of the International Society for Fertility Preservation Vienna, Austria. November 16-18; 2017 Session 2: Stem cells and in vitro growth of gametes Development, sex differentiation and

More information

Chapter 7 Conclusions

Chapter 7 Conclusions VII-1 Chapter 7 Conclusions VII-2 The development of cell-based therapies ranging from well-established practices such as bone marrow transplant to next-generation strategies such as adoptive T-cell therapy

More information

TISSUE-SPECIFIC STEM CELLS

TISSUE-SPECIFIC STEM CELLS TISSUE-SPECIFIC STEM CELLS Gdnf Upregulates c-fos Transcription via the Ras/Erk1/2 Pathway to Promote Mouse Spermatogonial Stem Cell Proliferation ZUPING HE, a JIJI JIANG, a MARIA KOKKINAKI, a NADY GOLESTANEH,

More information

Reproductive Toxicology

Reproductive Toxicology Reproductive Toxicology 32 (2011) 395 406 Contents lists available at SciVerse ScienceDirect Reproductive Toxicology jo u r n al hom epa ge: ww w.elsevier.com/locate/reprotox Effects of multiple doses

More information

Zhu et al, page 1. Supplementary Figures

Zhu et al, page 1. Supplementary Figures Zhu et al, page 1 Supplementary Figures Supplementary Figure 1: Visual behavior and avoidance behavioral response in EPM trials. (a) Measures of visual behavior that performed the light avoidance behavior

More information

Neural stem cells and the neurobiology of ageing. Chen Siyun 1, Dawe G.S. 2

Neural stem cells and the neurobiology of ageing. Chen Siyun 1, Dawe G.S. 2 ABSTRACT Neural stem cells and the neurobiology of ageing Chen Siyun 1, Dawe G.S. 2 Department of Physics, Faculty of Science, National University of Singapore 10 Kent Ridge Road, Singapore 117546 The

More information

Supplementary Figure 1. EC-specific Deletion of Snail1 Does Not Affect EC Apoptosis. (a,b) Cryo-sections of WT (a) and Snail1 LOF (b) embryos at

Supplementary Figure 1. EC-specific Deletion of Snail1 Does Not Affect EC Apoptosis. (a,b) Cryo-sections of WT (a) and Snail1 LOF (b) embryos at Supplementary Figure 1. EC-specific Deletion of Snail1 Does Not Affect EC Apoptosis. (a,b) Cryo-sections of WT (a) and Snail1 LOF (b) embryos at E10.5 were double-stained for TUNEL (red) and PECAM-1 (green).

More information

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION Supplementary Figure 1. Ras V12 expression in the entire eye-antennal disc does not cause invasive tumours. a, Eye-antennal discs expressing Ras V12 in all cells (marked with GFP, green) overgrow moderately

More information

Sestrin2 and BNIP3 (Bcl-2/adenovirus E1B 19kDa-interacting. protein3) regulate autophagy and mitophagy in renal tubular cells in. acute kidney injury

Sestrin2 and BNIP3 (Bcl-2/adenovirus E1B 19kDa-interacting. protein3) regulate autophagy and mitophagy in renal tubular cells in. acute kidney injury Sestrin2 and BNIP3 (Bcl-2/adenovirus E1B 19kDa-interacting protein3) regulate autophagy and mitophagy in renal tubular cells in acute kidney injury by Masayuki Ishihara 1, Madoka Urushido 2, Kazu Hamada

More information

Computer Simulation of the Rodent Spermatogonial Stem Cell Niche 1

Computer Simulation of the Rodent Spermatogonial Stem Cell Niche 1 BIOLOGY OF REPRODUCTION (2013) 88(5):131, 1 11 Published online before print 27 March 2013. DOI 10.1095/biolreprod.113.108639 Computer Simulation of the Rodent Spermatogonial Stem Cell Niche 1 Dirk G.

More information

(a) Significant biological processes (upper panel) and disease biomarkers (lower panel)

(a) Significant biological processes (upper panel) and disease biomarkers (lower panel) Supplementary Figure 1. Functional enrichment analyses of secretomic proteins. (a) Significant biological processes (upper panel) and disease biomarkers (lower panel) 2 involved by hrab37-mediated secretory

More information

marker. DAPI labels nuclei. Flies were 20 days old. Scale bar is 5 µm. Ctrl is

marker. DAPI labels nuclei. Flies were 20 days old. Scale bar is 5 µm. Ctrl is Supplementary Figure 1. (a) Nos is detected in glial cells in both control and GFAP R79H transgenic flies (arrows), but not in deletion mutant Nos Δ15 animals. Repo is a glial cell marker. DAPI labels

More information

Supplemental Information. Emi2 Is Essential for Mouse Spermatogenesis

Supplemental Information. Emi2 Is Essential for Mouse Spermatogenesis Cell Reports, Volume 20 Supplemental Information Emi2 Is Essential for Mouse Spermatogenesis Lakshmi Gopinathan, Radoslaw Szmyd, Diana Low, M. Kasim Diril, Heng-Yu Chang, Vincenzo Coppola, Kui Liu, Lino

More information

Developmental Biology

Developmental Biology Developmental Biology 371 (212) 35 46 Contents lists available at SciVerse ScienceDirect Developmental Biology journal homepage: www.elsevier.com/locate/developmentalbiology Geminin is required for mitotic

More information

Supplemental Information. Induction of Expansion and Folding. in Human Cerebral Organoids

Supplemental Information. Induction of Expansion and Folding. in Human Cerebral Organoids Cell Stem Cell, Volume 20 Supplemental Information Induction of Expansion and Folding in Human Cerebral Organoids Yun Li, Julien Muffat, Attya Omer, Irene Bosch, Madeline A. Lancaster, Mriganka Sur, Lee

More information

Supplementary Table; Supplementary Figures and legends S1-S21; Supplementary Materials and Methods

Supplementary Table; Supplementary Figures and legends S1-S21; Supplementary Materials and Methods Silva et al. PTEN posttranslational inactivation and hyperactivation of the PI3K/Akt pathway sustain primary T cell leukemia viability Supplementary Table; Supplementary Figures and legends S1-S21; Supplementary

More information

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION doi:10.1038/nature10495 WWW.NATURE.COM/NATURE 1 2 WWW.NATURE.COM/NATURE WWW.NATURE.COM/NATURE 3 4 WWW.NATURE.COM/NATURE WWW.NATURE.COM/NATURE 5 6 WWW.NATURE.COM/NATURE WWW.NATURE.COM/NATURE 7 8 WWW.NATURE.COM/NATURE

More information

Nature Genetics: doi: /ng Supplementary Figure 1. Assessment of sample purity and quality.

Nature Genetics: doi: /ng Supplementary Figure 1. Assessment of sample purity and quality. Supplementary Figure 1 Assessment of sample purity and quality. (a) Hematoxylin and eosin staining of formaldehyde-fixed, paraffin-embedded sections from a human testis biopsy collected concurrently with

More information

Nature Immunology: doi: /ni Supplementary Figure 1. Huwe1 has high expression in HSCs and is necessary for quiescence.

Nature Immunology: doi: /ni Supplementary Figure 1. Huwe1 has high expression in HSCs and is necessary for quiescence. Supplementary Figure 1 Huwe1 has high expression in HSCs and is necessary for quiescence. (a) Heat map visualizing expression of genes with a known function in ubiquitin-mediated proteolysis (KEGG: Ubiquitin

More information

Testis( Dissertation_ 全文 )

Testis( Dissertation_ 全文 ) Glycosylation Properties Associated TitleDifferentiation of Spermatogonial S Testis( Dissertation_ 全文 ) Author(s) Kim, Sung Min Citation Kyoto University ( 京都大学 ) Issue Date 2013-05-23 URL https://doi.org/10.14989/doctor.k17

More information

Supplementary Figure 1. A. Bar graph representing the expression levels of the 19 indicated genes in the microarrays analyses comparing human lung

Supplementary Figure 1. A. Bar graph representing the expression levels of the 19 indicated genes in the microarrays analyses comparing human lung Supplementary Figure 1. A. Bar graph representing the expression levels of the 19 indicated genes in the microarrays analyses comparing human lung immortalized broncho-epithelial cells (AALE cells) expressing

More information

a) Primary cultures derived from the pancreas of an 11-week-old Pdx1-Cre; K-MADM-p53

a) Primary cultures derived from the pancreas of an 11-week-old Pdx1-Cre; K-MADM-p53 1 2 3 4 5 6 7 8 9 10 Supplementary Figure 1. Induction of p53 LOH by MADM. a) Primary cultures derived from the pancreas of an 11-week-old Pdx1-Cre; K-MADM-p53 mouse revealed increased p53 KO/KO (green,

More information

Regulation of spermatogonia

Regulation of spermatogonia Qing Zhou and Michael D Griswold, School of Molecular Biosciences, Washington State University, Pullman, WA 99164 4660, USA Table of Contents 1. Introduction.........2 2. Spermatogenesis: in general......

More information

The Biology and Genetics of Cells and Organisms The Biology of Cancer

The Biology and Genetics of Cells and Organisms The Biology of Cancer The Biology and Genetics of Cells and Organisms The Biology of Cancer Mendel and Genetics How many distinct genes are present in the genomes of mammals? - 21,000 for human. - Genetic information is carried

More information

NOTCH1 Gain of Function in Germ Cells Causes Failure of Spermatogenesis in Male Mice

NOTCH1 Gain of Function in Germ Cells Causes Failure of Spermatogenesis in Male Mice Florida International University FIU Digital Commons HWCOM Faculty Publications Herbert Wertheim College of Medicine 7-30-2013 NOTCH1 Gain of Function in Germ Cells Causes Failure of Spermatogenesis in

More information

SUPPLEMENTAL INFORMATIONS

SUPPLEMENTAL INFORMATIONS 1 SUPPLEMENTAL INFORMATIONS Figure S1 Cumulative ZIKV production by testis explants over a 9 day-culture period. Viral titer values presented in Figure 1B (viral release over a 3 day-culture period measured

More information

Supplementary Figure 1. Expression of phospho-sik3 in normal and osteoarthritic articular cartilage in the knee. (a) Semiserial histological sections

Supplementary Figure 1. Expression of phospho-sik3 in normal and osteoarthritic articular cartilage in the knee. (a) Semiserial histological sections Supplementary Figure 1. Expression of phospho-sik3 in normal and osteoarthritic articular cartilage in the knee. (a) Semiserial histological sections of normal cartilage were stained with safranin O-fast

More information

Significance. 1 T.E., K.A.R, and E.L.A. contributed equally to this work.

Significance. 1 T.E., K.A.R, and E.L.A. contributed equally to this work. eriodic retinoic acid STRA8 signaling intersects with periodic germ-cell competencies to regu spermatogenesis Tsutomu Endo a,1,2, Katherine A. Romer a,b,1, Ericka L. Anderson a,c,1, Andrew E. altus a,c,

More information

Tumor suppressor Spred2 interaction with LC3 promotes autophagosome maturation and induces autophagy-dependent cell death

Tumor suppressor Spred2 interaction with LC3 promotes autophagosome maturation and induces autophagy-dependent cell death www.impactjournals.com/oncotarget/ Oncotarget, Supplementary Materials 2016 Tumor suppressor Spred2 interaction with LC3 promotes autophagosome maturation and induces autophagy-dependent cell death Supplementary

More information

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION doi:10.1038/nature12215 Supplementary Figure 1. The effects of full and dissociated GR agonists in supporting BFU-E self-renewal divisions. BFU-Es were cultured in self-renewal medium with indicated GR

More information

Supplementary Information POLO-LIKE KINASE 1 FACILITATES LOSS OF PTEN-INDUCED PROSTATE CANCER FORMATION

Supplementary Information POLO-LIKE KINASE 1 FACILITATES LOSS OF PTEN-INDUCED PROSTATE CANCER FORMATION Supplementary Information POLO-LIKE KINASE 1 FACILITATES LOSS OF PTEN-INDUCED PROSTATE CANCER FORMATION X. Shawn Liu 1, 3, Bing Song 2, 3, Bennett D. Elzey 3, 4, Timothy L. Ratliff 3, 4, Stephen F. Konieczny

More information

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION doi:10.1038/nature12652 Supplementary Figure 1. PRDM16 interacts with endogenous EHMT1 in brown adipocytes. Immunoprecipitation of PRDM16 complex by flag antibody (M2) followed by Western blot analysis

More information

Epithelial cell death is an important contributor to oxidant-mediated acute lung injury SUPPORTING INFORMATION 60611, USA

Epithelial cell death is an important contributor to oxidant-mediated acute lung injury SUPPORTING INFORMATION 60611, USA Epithelial cell death is an important contributor to oxidant-mediated acute lung injury SUPPORTING INFORMATION G.R. Scott Budinger 1,2 *, Gökhan M. Mutlu 1 *, Daniela Urich 2, Saul Soberanes 1, Leonard

More information

Programmed Cell Death (apoptosis)

Programmed Cell Death (apoptosis) Programmed Cell Death (apoptosis) Stereotypic death process includes: membrane blebbing nuclear fragmentation chromatin condensation and DNA framentation loss of mitochondrial integrity and release of

More information

Supplementary Figure 1.TRIM33 binds β-catenin in the nucleus. a & b, Co-IP of endogenous TRIM33 with β-catenin in HT-29 cells (a) and HEK 293T cells

Supplementary Figure 1.TRIM33 binds β-catenin in the nucleus. a & b, Co-IP of endogenous TRIM33 with β-catenin in HT-29 cells (a) and HEK 293T cells Supplementary Figure 1.TRIM33 binds β-catenin in the nucleus. a & b, Co-IP of endogenous TRIM33 with β-catenin in HT-29 cells (a) and HEK 293T cells (b). TRIM33 was immunoprecipitated, and the amount of

More information