Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Development ; 151(12)2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38934417

ABSTRACT

Spermatogonial stem cells (SSCs) undergo self-renewal division to sustain spermatogenesis. Although it is possible to derive SSC cultures in most mouse strains, SSCs from a 129 background never proliferate under the same culture conditions, suggesting they have distinct self-renewal requirements. Here, we established long-term culture conditions for SSCs from mice of the 129 background (129 mice). An analysis of 129 testes showed significant reduction of GDNF and CXCL12, whereas FGF2, INHBA and INHBB were higher than in testes of C57BL/6 mice. An analysis of undifferentiated spermatogonia in 129 mice showed higher expression of Chrna4, which encodes an acetylcholine (Ach) receptor component. By supplementing medium with INHBA and Ach, SSC cultures were derived from 129 mice. Following lentivirus transduction for marking donor cells, transplanted cells re-initiated spermatogenesis in infertile mouse testes and produced transgenic offspring. These results suggest that the requirements of SSC self-renewal in mice are diverse, which has important implications for understanding self-renewal mechanisms in various animal species.


Subject(s)
Mice, Inbred C57BL , Spermatogenesis , Spermatogonia , Testis , Animals , Male , Mice , Spermatogonia/cytology , Spermatogonia/metabolism , Spermatogenesis/genetics , Spermatogenesis/physiology , Testis/metabolism , Testis/cytology , Cell Self Renewal , Adult Germline Stem Cells/metabolism , Adult Germline Stem Cells/cytology , Cells, Cultured , Receptors, Nicotinic/metabolism , Receptors, Nicotinic/genetics , Mice, Inbred Strains , Cell Differentiation , Cell Proliferation , Stem Cells/cytology , Stem Cells/metabolism , Mice, Transgenic
2.
Stem Cell Reports ; 19(4): 443-455, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38458191

ABSTRACT

Spermatogonial stem cell (SSC) transplantation is a valuable tool for studying stem cell-niche interaction. However, the conventional approach requires the removal of endogenous SSCs, causing damage to the niche. Here we introduce WIN18,446, an ALDH1A2 inhibitor, to enhance SSC colonization in nonablated recipients. Pre-transplantation treatment with WIN18,446 induced abnormal claudin protein expression, which comprises the blood-testis barrier and impedes SSC colonization. Consequently, WIN18,446 increased colonization efficiency by 4.6-fold compared with untreated host. WIN18,446-treated testes remained small despite the cessation of WIN18,446, suggesting its irreversible effect. Offspring were born by microinsemination using donor-derived sperm. While WIN18,446 was lethal to busulfan-treated mice, cyclophosphamide- or radiation-treated animals survived after WIN18,446 treatment. Although WIN18,446 is not applicable to humans due to toxicity, similar ALDH1A2 inhibitors may be useful for SSC transplantation into nonablated testes, shedding light on the role of retinoid metabolism on SSC-niche interactions and advancing SSC research in animal models and humans.


Subject(s)
Semen , Spermatogonia , Humans , Mice , Male , Animals , Spermatogonia/metabolism , Testis/metabolism , Fertility , Stem Cell Transplantation , Spermatogenesis
3.
J Reprod Dev ; 69(6): 347-355, 2023 Dec 08.
Article in English | MEDLINE | ID: mdl-37899250

ABSTRACT

Spermatogonial stem cells (SSCs) possess a unique ability to recolonize the seminiferous tubules. Upon microinjection into the adluminal compartment of the seminiferous tubules, SSCs transmigrate through the blood-testis barrier (BTB) to the basal compartment of the tubule and reinitiate spermatogenesis. It was recently discovered that inhibiting retinoic acid signaling with WIN18,446 enhances SSC colonization by transiently suppressing spermatogonia differentiation, thereby promoting fertility restoration. In this study, we report that WIN18,446 increases SSC colonization by disrupting the BTB. WIN18,446 altered the expression patterns of tight junction proteins (TJPs) and disrupted the BTB in busulfan-treated mice. WIN18,446 upregulated the expression of FGF2, one of the self-renewal factors for SSCs. While WIN18,446 enhanced SSC colonization in busulfan-treated wild-type mice, it did not increase colonization levels in busulfan-treated Cldn11-deficient mice, which lack the BTB, indicating that the enhancement of SSC colonization in wild-type testes depended on the loss of the BTB. Serial transplantation analysis revealed impaired self-renewal caused by WIN18,446, indicating that WIN18,446-mediated inhibition of retinoic acid signaling impaired SSC self-renewal. Strikingly, WIN18,446 administration resulted in the death of 45% of busulfan-treated recipient mice. These findings suggest that TJP modulation is the primary mechanism behind enhanced SSC homing by WIN18,446 and raise concerns regarding the use of WIN18,446 for human SSC transplantation.


Subject(s)
Blood-Testis Barrier , Busulfan , Male , Animals , Mice , Humans , Blood-Testis Barrier/metabolism , Busulfan/pharmacology , Busulfan/metabolism , Spermatogonia/metabolism , Testis , Spermatogenesis , Fertility , Cell Transplantation , Stem Cells , Tretinoin/pharmacology , Stem Cell Transplantation
4.
Stem Cell Reports ; 18(4): 985-998, 2023 04 11.
Article in English | MEDLINE | ID: mdl-36963391

ABSTRACT

The testis is an immune-privileged organ. It is considered that the testis somatic microenvironment is responsible for immune suppression. However, immunological properties of spermatogonial stem cells (SSCs) have remained unknown. Here, we report the birth of allogeneic offspring by enhanced expression of immunosuppressive PD-L1 in SSCs. In vitro supplementation of GDNF and FGF2 increased expression of PD-L1 in SSCs. Cultured SSCs maintained allogeneic spermatogenesis that persisted for >1 year. However, depletion or gene editing of Pd-l1 family genes in SSCs prevented allogeneic spermatogenesis, which suggested that germ cells are responsible for suppression of the allogeneic response. PD-L1 was induced by activation of the MAPK14-BCL6B pathway, which drives self-renewal by reactive oxygen species (ROS) generation. By contrast, reduced ROS or Mapk14 deficiency downregulated PD-L1. Allogeneic offspring were born after SSC transplantation into congenitally infertile and chemically castrated mice. Thus, SSCs have unique immunological properties, which make allogeneic recipients into "surrogate fathers."


Subject(s)
Hematopoietic Stem Cell Transplantation , Mitogen-Activated Protein Kinase 14 , Male , Mice , Animals , Spermatogonia , Reactive Oxygen Species/metabolism , Mitogen-Activated Protein Kinase 14/metabolism , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Cell Proliferation , Testis , Spermatogenesis/genetics
5.
J Reprod Dev ; 68(6): 369-376, 2022 Dec 19.
Article in English | MEDLINE | ID: mdl-36223953

ABSTRACT

Oogenesis depends on close interactions between oocytes and granulosa cells. Abnormal signaling between these cell types can result in infertility. However, attempts to manipulate oocyte-granulosa cell interactions have had limited success, likely due to the blood-follicle barrier (BFB), which prevents the penetration of exogenous materials into ovarian follicles. Here, we used adenoviruses (AVs) to manipulate the oocyte-granulosa cell interactions. AVs penetrated the BFB and transduced granulosa cells through ovarian microinjection. Although AVs caused transient inflammation, they did not impair fertility in wild-type mice. Introduction of Kitl-expressing AVs into congenitally infertile KitlSl-t/KitlSl-t mutant mouse ovaries, which contained only primordial follicles because of a lack of Kitl expression, restored fertility through natural mating. The offspring showed no evidence of AV integration and exhibited normal genomic imprinting patterns for imprinted genes. These results demonstrate the usefulness of AVs for manipulating oogenesis and suggest the possibility of gene therapies for human female infertility.


Subject(s)
Infertility, Female , Mice , Female , Animals , Humans , Infertility, Female/genetics , Infertility, Female/therapy , Infertility, Female/metabolism , Adenoviridae/genetics , Ovarian Follicle/metabolism , Granulosa Cells/metabolism , Oocytes/metabolism , Fertility/genetics
6.
Cell Rep Med ; 3(5): 100606, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35584625

ABSTRACT

Oocytes and granulosa cells closely interact with each other during follicular development, and a lack of appropriate signaling between them results in infertility. Attempts to manipulate oocyte microenvironment have been impeded by the impermeability of the blood-follicle barrier (BFB). To establish a strategy for manipulating oogenesis, we use adeno-associated viruses (AAVs), which have a unique ability of transcytosis. Microinjecting of AAVs into the ovarian stroma penetrates the BFB and achieves long-term gene expression. Introduction of an AAV carrying the mouse Kitl gene restores oogenesis in congenitally infertile KitlSl-t/KitlSl-t mutant mouse ovaries, which lack Kitl expression but contain only primordial follicles. Healthy offspring without AAV integration are born by natural mating. Therefore, AAV-mediated gene delivery not only provides a means for studying oocyte-granulosa interactions through the manipulation of the oocyte microenvironment but could also be a powerful method to treat female infertility resulting from somatic cell defects.


Subject(s)
Infertility, Female , Ovary , Animals , Dependovirus/genetics , Female , Fertility/genetics , Humans , Infertility, Female/genetics , Mice , Ovarian Follicle
7.
Stem Cell Reports ; 17(4): 924-935, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35334214

ABSTRACT

Gametogenesis requires close interactions between germ cells and somatic cells. Derivation of sperm from spermatogonial stem cells (SSCs) is hampered by the inefficiency of spermatogonial transplantation technique in many animal species because it requires a large number of SSCs and depletion of endogenous spermatogenesis. Here we used mouse testis primordia and organoids to induce spermatogenesis from SSCs. We microinjected mouse SSCs into embryonic gonads or reaggregated neonatal testis organoids, which were transplanted under the tunica albuginea of mature testes. As few as 1 × 104 donor cells colonized both types of transplants and produced sperm. Moreover, rat embryonic gonads supported xenogeneic spermatogenesis from mouse SSCs when transplanted in testes of immunodeficient mice. Offspring with normal genomic imprinting patterns were born after microinsemination. These results demonstrate remarkable flexibility of the germ cell-somatic cell interaction and raise new strategies of SSC manipulation for animal transgenesis and analysis of male infertility.


Subject(s)
Hematopoietic Stem Cell Transplantation , Testis , Animals , Male , Mice , Organoids , Rats , Spermatogenesis/genetics , Spermatogonia/transplantation , Stem Cell Transplantation
8.
Stem Cell Reports ; 16(7): 1832-1844, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34143973

ABSTRACT

Spermatogonial transplantation has been used as a standard assay for spermatogonial stem cells (SSCs). After transplantation into the seminiferous tubules, SSCs transmigrate through the blood-testis barrier (BTB) between Sertoli cells and settle in a niche. Unlike in the repair of other self-renewing systems, SSC transplantation is generally performed after complete destruction of endogenous spermatogenesis. Here, we examined the impacts of recipient conditioning on SSC homing. Germ cell ablation downregulated the expression of glial cell line-derived neurotrophic factor, which has been shown to attract SSCs to niches, implying that nonablated niches would attract SSCs more efficiently. As expected, SSCs colonized nonablated testes when transplanted into recipients with the same genetic background. Moreover, although spermatogenesis was arrested at the spermatocyte stage in Cldn11-deficient mice without a BTB, transplantation not only enhanced donor colonization but also restored normal spermatogenesis. The results show promise for the development of a new transplantation strategy to overcome male infertility.


Subject(s)
Spermatogonia/cytology , Spermatogonia/transplantation , Stem Cell Transplantation , Testis/cytology , Animals , Apoptosis , Biomarkers/metabolism , Busulfan/pharmacology , Claudins/metabolism , Cytokines/metabolism , Germ Cells/drug effects , Germ Cells/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Male , Mice, Knockout , Regeneration/drug effects , Spermatogenesis
9.
Genes Dev ; 35(3-4): 250-260, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33446567

ABSTRACT

Reactive oxygen species (ROS) produced by NADPH1 oxidase 1 (NOX1) are thought to drive spermatogonial stem cell (SSC) self-renewal through feed-forward production of ROS by the ROS-BCL6B-NOX1 pathway. Here we report the critical role of oxygen on ROS-induced self-renewal. Cultured SSCs proliferated poorly and lacked BCL6B expression under hypoxia despite increase in mitochondria-derived ROS. Due to lack of ROS amplification under hypoxia, NOX1-derived ROS were significantly reduced, and Nox1-deficient SSCs proliferated poorly under hypoxia but normally under normoxia. NOX1-derived ROS also influenced hypoxic response in vivo because Nox1-deficient undifferentiated spermatogonia showed significantly reduced expression of HIF1A, a master transcription factor for hypoxic response. Hypoxia-induced poor proliferation occurred despite activation of MYC and suppression of CDKN1A by HIF1A, whose deficiency exacerbated self-renewal efficiency. Impaired proliferation of Nox1- or Hif1a-deficient SSCs under hypoxia was rescued by Cdkn1a depletion. Consistent with these observations, Cdkn1a-deficient SSCs proliferated actively only under hypoxia but not under normoxia. On the other hand, chemical suppression of mitochondria-derived ROS or Top1mt mitochondria-specific topoisomerase deficiency did not influence SSC fate, suggesting that NOX1-derived ROS play a more important role in SSCs than mitochondria-derived ROS. These results underscore the importance of ROS origin and oxygen tension on SSC self-renewal.


Subject(s)
Adult Germline Stem Cells/cytology , Cell Hypoxia/physiology , Oxygen/metabolism , Reactive Oxygen Species/metabolism , Animals , Cell Division/genetics , Cell Proliferation/genetics , Cells, Cultured , DNA Topoisomerases, Type I/genetics , Gene Expression Regulation, Developmental , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Mice , Mice, Knockout , Mitochondria/physiology , NADPH Oxidase 1/metabolism
10.
J Reprod Dev ; 66(4): 341-349, 2020 Aug 20.
Article in English | MEDLINE | ID: mdl-32213736

ABSTRACT

The spermatogonial stem cell (SSC) population in testis is small, and the lack of SSC markers has severely handicapped research on these cells. During our attempt to identify genes involved in SSC aging, we found that CD2 is expressed in cultured SSCs. Flow cytometric analysis and spermatogonial transplantation experiments showed that CD2 is expressed in SSCs from mature adult mouse testes. Cultured SSCs transfected with short hairpin RNAs (shRNAs) against CD2 proliferated poorly and showed an increased frequency of apoptosis. Moreover, functional analysis of transfected cells revealed impairment of SSC activity. Fluorescence activated cell sorting and spermatogonial transplantation experiments showed that CD2 is expressed not only in mouse but also in rat SSCs. The results indicate that CD2 is a novel SSC surface marker conserved between mouse and rat SSCs.


Subject(s)
Adult Germline Stem Cells/metabolism , CD2 Antigens/metabolism , Spermatogenesis/physiology , Spermatogonia/metabolism , Animals , Flow Cytometry , Male , Mice , Rats
11.
Biol Reprod ; 102(1): 220-232, 2020 02 12.
Article in English | MEDLINE | ID: mdl-31403678

ABSTRACT

Spermatogonial stem cells (SSCs) undergo continuous self-renewal division in response to self-renewal factors. The present study identified ephrin type-A receptor 2 (EPHA2) on mouse SSCs and showed that supplementation of glial cell-derived neurotrophic factor (GDNF) and fibroblast growth factor 2 (FGF2), which are both SSC self-renewal factors, induced EPHA2 expression in cultured SSCs. Spermatogonial transplantation combined with magnetic-activated cell sorting or fluorescence-activated cell sorting also revealed that EPHA2 was expressed in SSCs. Additionally, ret proto-oncogene (RET) phosphorylation levels decreased following the knockdown (KD) of Epha2 expression via short hairpin ribonucleic acid (RNA). Although the present immunoprecipitation experiments did not reveal an association between RET with EPHA2, RET interacted with FGFR2. The Epha2 KD decreased the proliferation of cultured SSCs and inhibited the binding of cultured SSCs to laminin-coated plates. The Epha2 KD also significantly reduced the colonization of testis cells by spermatogonial transplantation. EPHA2 was also expressed in human GDNF family receptor alpha 1-positive spermatogonia. The present results indicate that SSCs express EPHA2 and suggest that it is a critical modifier of self-renewal signals in SSCs.


Subject(s)
Adult Germline Stem Cells/metabolism , Receptors, Eph Family/metabolism , Spermatogonia/metabolism , Testis/metabolism , Adult Germline Stem Cells/cytology , Animals , Cell Proliferation/physiology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Male , Mice , Phosphorylation , Proto-Oncogene Mas , RNA, Small Interfering , Receptors, Eph Family/genetics , Spermatogonia/cytology
12.
Life Sci Alliance ; 2(2)2019 04.
Article in English | MEDLINE | ID: mdl-30940732

ABSTRACT

Reactive oxygen species (ROS) play critical roles in self-renewal division for various stem cell types. However, it remains unclear how ROS signals are integrated with self-renewal machinery. Here, we report that the MAPK14/MAPK7/BCL6B pathway creates a positive feedback loop to drive spermatogonial stem cell (SSC) self-renewal via ROS amplification. The activation of MAPK14 induced MAPK7 phosphorylation in cultured SSCs, and targeted deletion of Mapk14 or Mapk7 resulted in significant SSC deficiency after spermatogonial transplantation. The activation of this signaling pathway not only induced Nox1 but also increased ROS levels. Chemical screening of MAPK7 targets revealed many ROS-dependent spermatogonial transcription factors, of which BCL6B was found to initiate ROS production by increasing Nox1 expression via ETV5-induced nuclear translocation. Because hydrogen peroxide or Nox1 transfection also induced BCL6B nuclear translocation, our results suggest that BCL6B initiates and amplifies ROS signals to activate ROS-dependent spermatogonial transcription factors by forming a positive feedback loop.


Subject(s)
Adult Germline Stem Cells/physiology , Cell Self Renewal/physiology , Reactive Oxygen Species/metabolism , Animals , Benzodiazepinones/pharmacology , Cell Proliferation/drug effects , DNA-Binding Proteins/metabolism , Feedback, Physiological/physiology , Gene Knockout Techniques , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Mitogen-Activated Protein Kinase 14/genetics , Mitogen-Activated Protein Kinase 14/metabolism , Mitogen-Activated Protein Kinase 7/genetics , Mitogen-Activated Protein Kinase 7/metabolism , NADPH Oxidase 1/genetics , NADPH Oxidase 1/metabolism , Real-Time Polymerase Chain Reaction , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection
13.
J Reprod Dev ; 64(6): 511-522, 2018 Dec 14.
Article in English | MEDLINE | ID: mdl-30175719

ABSTRACT

Stem cell homing is a complex phenomenon that involves multiple steps; thus far, attempts to increase homing efficiency have met with limited success. Spermatogonial stem cells (SSCs) migrate to the niche after microinjection into seminiferous tubules, but the homing efficiency is very low. Here we report that reversible disruption of the blood-testis barrier (BTB) between Sertoli cells enhances the homing efficiency of SSCs. We found that SSCs on a C57BL/6 background are triggered to proliferate in vitro when MHY1485, which stimulates MTORC, were added to culture medium. However, the cultured cells did not produce offspring by direct injection into the seminiferous tubules. When acyline, a gonadotropin-releasing hormone (GnRH) analogue, was administered into infertile recipients, SSC colonization increased by ~5-fold and the recipients sired offspring. In contrast, both untreated individuals and recipients that received leuprolide, another GnRH analogue, remained infertile. Acyline not only decreased CLDN5 expression but also impaired the BTB, suggesting that increased colonization was caused by efficient SSC migration through the BTB. Enhancement of stem cell homing by tight junction protein manipulation constitutes a new approach to improve homing efficiency, and similar strategy may be applicable to other self-renewing tissues.


Subject(s)
Blood-Testis Barrier/metabolism , Sertoli Cells/metabolism , Spermatogonia/metabolism , Testis/metabolism , Animals , Blood-Testis Barrier/drug effects , Cell Proliferation/drug effects , Claudin-5/metabolism , Male , Mice , Morpholines/pharmacology , Oligopeptides/pharmacology , Sertoli Cells/cytology , Sertoli Cells/drug effects , Spermatogenesis/drug effects , Spermatogenesis/physiology , Spermatogonia/cytology , Spermatogonia/drug effects , Stem Cell Niche/drug effects , Testis/drug effects , Triazines/pharmacology
14.
Stem Cell Reports ; 9(4): 1180-1191, 2017 10 10.
Article in English | MEDLINE | ID: mdl-28943251

ABSTRACT

The introduction of megabase-sized large DNA fragments into the germline has been a difficult task. Although microcell-mediated chromosome transfer into mouse embryonic stem cells (ESCs) allows the production of transchromosomic mice, ESCs have unstable karyotypes and germline transmission is unreliable by chimera formation. As spermatogonial stem cells (SSCs) are the only stem cells in the germline, they represent an attractive target for germline modification. Here, we report successful transfer of a mouse artificial chromosome (MAC) into mouse germline stem cells (GSCs), cultured spermatogonia enriched for SSCs. MAC-transferred GSCs maintained the host karyotype and MAC more stably than ESCs, which have significant variation in chromosome number. Moreover, MAC-transferred GSCs produced transchromosomic mice following microinjection into the seminiferous tubules of infertile recipients. Successful transfer of MACs to GSCs overcomes the problems associated with ESC-mediated germline transmission and provides new possibilities in germline modification.


Subject(s)
Chromosomes, Artificial , Gene Transfer Techniques , Spermatogonia/cytology , Spermatogonia/metabolism , Animals , Biomarkers , Cell Tracking , Gene Expression , Genes, Reporter , Genomic Instability , Immunophenotyping , Karyotype , Male , Mice , Mice, Transgenic , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Phenotype , Spermatogenesis
15.
Genes Dev ; 30(23): 2637-2648, 2016 12 01.
Article in English | MEDLINE | ID: mdl-28007786

ABSTRACT

Myc plays critical roles in the self-renewal division of various stem cell types. In spermatogonial stem cells (SSCs), Myc controls SSC fate decisions because Myc overexpression induces enhanced self-renewal division, while depletion of Max, a Myc-binding partner, leads to meiotic induction. However, the mechanism by which Myc acts on SSC fate is unclear. Here we demonstrate a critical link between Myc/Mycn gene activity and glycolysis in SSC self-renewal. In SSCs, Myc/Mycn are regulated by Foxo1, whose deficiency impairs SSC self-renewal. Myc/Mycn-deficient SSCs not only undergo limited self-renewal division but also display diminished glycolytic activity. While inhibition of glycolysis decreased SSC activity, chemical stimulation of glycolysis or transfection of active Akt1 or Pdpk1 (phosphoinositide-dependent protein kinase 1 ) augmented self-renewal division, and long-term SSC cultures were derived from a nonpermissive strain that showed limited self-renewal division. These results suggested that Myc-mediated glycolysis is an important factor that increases the frequency of SSC self-renewal division.


Subject(s)
Cell Self Renewal/genetics , Gene Expression Regulation, Developmental/genetics , Glycolysis/genetics , N-Myc Proto-Oncogene Protein/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Spermatogonia/cytology , Stem Cells/metabolism , 3-Phosphoinositide-Dependent Protein Kinases/metabolism , Animals , Cell Division/genetics , Cell Proliferation/genetics , Gene Knockout Techniques , Male , Mice , Mice, Inbred C57BL , N-Myc Proto-Oncogene Protein/genetics , Proto-Oncogene Proteins c-myc/genetics , RNA Splicing Factors/metabolism , Stem Cells/enzymology
16.
Biol Reprod ; 94(5): 112, 2016 05.
Article in English | MEDLINE | ID: mdl-27053363

ABSTRACT

Spermatogonial stem cells (SSCs) provide the foundation for spermatogenesis. Earlier studies have shown that the transplantation of SSCs restores fertility to infertile recipients. However, most of the previously described experiments have depended on transplantation using sexually immature animals, and the effectiveness of spermatogonial transplantation in mature animals has not been examined in detail. In this study, we evaluated the efficiency of offspring production by adult recipients of spermatogonial transplantation using germline stem (GS) cells, cultured spermatogonia with enriched SSC activity. GS cells were transplanted into mature WBB6F1-W/W(v) (W) or busulfan-treated mice, which were then mated with female mice to obtain offspring from donor cells. We found that GS cells produced offspring most efficiently by transplantation into busulfan (44 mg/kg)-treated mice and all recipients produced progeny within 4 mo (76-111 days) after transplantation. When the dose dependence of offspring production was examined in W mice, approximately 40-80 SSCs were estimated to be required for fertility restoration. Efficient offspring production using GS cells and spermatogonial transplantation will be useful for analyzing factors involved in male fertility.


Subject(s)
Adult Germline Stem Cells/transplantation , Fertility , Infertility, Male/therapy , Stem Cell Transplantation/methods , Animals , Female , Infertility, Male/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Pregnancy , Spermatogenesis/physiology , Spermatogonia/physiology , Testis/pathology
17.
Biol Reprod ; 94(1): 13, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26607720

ABSTRACT

Spermatogonial stem cells (SSCs) comprise a small population of germ cells with self-renewal potential. Previous studies have shown that SSCs share several common features with stem cells in other self-renewing tissues, including surface markers and proliferative machinery. However, studies of SSCs are severely handicapped by the small number of SSCs and the lack of SSC-specific markers. In the present study, we examined the utility of CDy1 and Rh123, both of which are used for the collection of stem cells in several self-renewing tissues. CDy1 stained germline stem (GS) cells, cultured spermatogonia enriched for SSC activity, after in vitro incubation without exerting toxic effects. Unlike previously reported stem cell-specific dyes, CDy1 was also useful for enrichment of SSCs in both GS cell culture and mature adult testes. Spermatogonial transplantation showed that ∼1 in 66.7 cells exhibited SSC activity after CDH1-based magnetic cell selection and CDy1 staining. In contrast, although Rh123 was previously used successfully to collect SSCs from cryptorchid testes, it was not possible to recover SSCs from both GS cell cultures and wild-type testes. Thus, CDy1 staining will provide a useful strategy for the enrichment of SSCs and may be used in conjunction with other reagents for the enrichment of SSCs.


Subject(s)
Anthracenes/chemistry , Fluorescent Dyes/chemistry , Morpholines/chemistry , Spermatogonia/ultrastructure , Stem Cells/ultrastructure , Animals , Cdh1 Proteins/metabolism , Cells, Cultured , Cryptorchidism/pathology , Germ Cells/ultrastructure , Magnetics , Male , Mice , Mice, Transgenic , Rhodamine 123/chemistry , Spermatogonia/transplantation , Testis/cytology , Tetraspanin 29/metabolism
18.
Biol Reprod ; 92(6): 147, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25947060

ABSTRACT

Spermatogonial stem cells (SSCs) represent a unique population of germ cells with self-renewal potential. Although reactive oxygen species (ROS) are considered toxic to germ cells, we recently showed that moderate levels of ROS are required for SSC self-renewal and that Nox1 is involved in ROS generation. In this study, we showed that self-renewal factor treatment induces Nox3 to trigger SSC self-renewal. Nox3 was transiently expressed in cultured spermatogonia by FGF2 and GDNF stimulation, whereas Nox1 was expressed predominantly during the stable phase of proliferation. Nox3 inhibition by short hairpin RNA reduced cytokine-induced ROS generation and limited the proliferation of cultured spermatogonia. Although Nox3 overexpression revealed no apparent effect, depletion of Nox3 decreased the number of SSCs in both cultured spermatogonia and freshly isolated testis cells. Our results suggest that self-renewal of SSCs is regulated by sequential activation of different Nox genes, and underscore the complexity of ROS regulation in the self-renewal division of SSCs.


Subject(s)
NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Spermatogenesis/physiology , Spermatogonia/metabolism , Stem Cells/metabolism , Animals , Cell Proliferation , Cells, Cultured , Male , Mice , NADH, NADPH Oxidoreductases/genetics , NADH, NADPH Oxidoreductases/metabolism , NADPH Oxidase 1 , NADPH Oxidases/genetics , Spermatogonia/cytology , Stem Cells/cytology
19.
Stem Cell Reports ; 4(3): 489-502, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25684228

ABSTRACT

Spermatogonial stem cells (SSCs) are required for spermatogenesis. Earlier studies showed that glial cell line-derived neurotrophic factor (GDNF) was indispensable for SSC self-renewal by binding to the GFRA1/RET receptor. Mice with mutations in these molecules showed impaired spermatogenesis, which was attributed to SSC depletion. Here we show that SSCs undergo GDNF-independent self-renewal. A small number of spermatogonia formed colonies when testis fragments from a Ret mutant mouse strain were transplanted into heterologous recipients. Moreover, fibroblast growth factor 2 (FGF2) supplementation enabled in vitro SSC expansion without GDNF. Although GDNF-mediated self-renewal signaling required both AKT and MAP2K1/2, the latter was dispensable in FGF2-mediated self-renewal. FGF2-depleted testes exhibited increased levels of GDNF and were enriched for SSCs, suggesting that the balance between FGF2 and GDNF levels influences SSC self-renewal in vivo. Our results show that SSCs exhibit at least two modes of self-renewal and suggest complexity of SSC regulation in vivo.


Subject(s)
Cell Self Renewal , Fibroblast Growth Factor 2/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Spermatogonia/cytology , Spermatogonia/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Proliferation , Fibroblast Growth Factor 2/pharmacology , Gene Expression , Germ Cells/cytology , Germ Cells/metabolism , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Male , Mice , Mice, Transgenic , Mutation , Proto-Oncogene Proteins c-ret/genetics , Seminiferous Tubules/metabolism , Spermatogonia/drug effects , Stem Cell Transplantation , Stem Cells/drug effects , Testis/metabolism
20.
Stem Cell Reports ; 3(4): 676-89, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25358794

ABSTRACT

Germ cells are thought to exhibit a unique DNA damage response that differs from that of somatic stem cells, and previous studies suggested that Trp53 is not involved in the survival of spermatogonial stem cells (SSCs) after irradiation. Here, we report a critical role for the Trp53-Trp53inp1-Tnfrsf10b pathway during radiation-induced SSC apoptosis. Spermatogonial transplantation revealed that Trp53 deficiency increased the survival of SSCs after irradiation. Although Bbc3, a member of the intrinsic apoptotic pathway, was implicated in apoptosis of germ and somatic stem cells, Bbc3 depletion inhibited apoptosis in committed spermatogonia, but not in SSCs. In contrast, inhibition of Tnfrsf10b, an extrinsic apoptosis regulator, rescued SSCs. Tnfrsf10b, whose deficiency protected SSCs, was upregulated by Trp53inp1 upon irradiation. These results suggest that the Trp53-Trp53inp1-Tnfrsf10b pathway responds to genotoxic damage in SSCs and that stem and progenitor cells exhibit distinct DNA damage responses in self-renewing tissue.


Subject(s)
Adult Stem Cells/metabolism , Carrier Proteins/metabolism , Heat-Shock Proteins/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Spermatogonia/metabolism , Tumor Suppressor Protein p53/metabolism , Adult Stem Cells/radiation effects , Animals , Apoptosis , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Carrier Proteins/genetics , Heat-Shock Proteins/genetics , Male , Mice , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Spermatogonia/radiation effects , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...