Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
1.
Genes Dev ; 35(3-4): 250-260, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33446567

RESUMO

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.


Assuntos
Células-Tronco Germinativas Adultas/citologia , Hipóxia Celular/fisiologia , Oxigênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Divisão Celular/genética , Proliferação de Células/genética , Células Cultivadas , DNA Topoisomerases Tipo I/genética , Regulação da Expressão Gênica no Desenvolvimento , Subunidade alfa do Fator 1 Induzível por Hipóxia/deficiência , Camundongos , Camundongos Knockout , Mitocôndrias/fisiologia , NADPH Oxidase 1/metabolismo
2.
Development ; 151(12)2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38934417

RESUMO

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.


Assuntos
Camundongos Endogâmicos C57BL , Espermatogênese , Espermatogônias , Testículo , Animais , Masculino , Camundongos , Espermatogônias/citologia , Espermatogônias/metabolismo , Espermatogênese/genética , Espermatogênese/fisiologia , Testículo/metabolismo , Testículo/citologia , Autorrenovação Celular , Células-Tronco Germinativas Adultas/metabolismo , Células-Tronco Germinativas Adultas/citologia , Células Cultivadas , Receptores Nicotínicos/metabolismo , Receptores Nicotínicos/genética , Camundongos Endogâmicos , Diferenciação Celular , Proliferação de Células , Células-Tronco/citologia , Células-Tronco/metabolismo , Camundongos Transgênicos
3.
Annu Rev Cell Dev Biol ; 29: 163-87, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24099084

RESUMO

Spermatogenesis originates from spermatogonial stem cells (SSCs). Development of the spermatogonial transplantation technique in 1994 provided the first functional assay to characterize SSCs. In 2000, glial cell line-derived neurotrophic factor was identified as a SSC self-renewal factor. This discovery not only provided a clue to understand SSC self-renewing mechanisms but also made it possible to derive germline stem (GS) cell cultures in 2003. In vitro culture of GS cells demonstrated their potential pluripotency and their utility in germline modification. However, in vivo SSC analyses have challenged the traditional concept of SSC self-renewal and have revealed their relationship with the microenvironment. An improved understanding of SSC self-renewal through functional assays promises to uncover fundamental principles of stem cell biology and will enable us to use these cells for applications in animal transgenesis and medicine.


Assuntos
Células-Tronco Adultas/citologia , Células-Tronco Adultas/fisiologia , Espermatogênese , Animais , Diferenciação Celular , Masculino , Nicho de Células-Tronco , Transplante de Células-Tronco , Fatores de Transcrição/fisiologia
4.
Development ; 150(20)2023 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-36897562

RESUMO

Reactive oxygen species (ROS) are generated from NADPH oxidases and mitochondria; they are generally harmful for stem cells. Spermatogonial stem cells (SSCs) are unique among tissue-stem cells because they undergo ROS-dependent self-renewal via NOX1 activation. However, the mechanism by which SSCs are protected from ROS remains unknown. Here, we demonstrate a crucial role for Gln in ROS protection using cultured SSCs derived from immature testes. Measurements of amino acids required for SSC cultures revealed the indispensable role of Gln in SSC survival. Gln induced Myc expression to drive SSC self-renewal in vitro, whereas Gln deprivation triggered Trp53-dependent apoptosis and impaired SSC activity. However, apoptosis was attenuated in cultured SSCs that lacked NOX1. In contrast, cultured SSCs lacking Top1mt mitochondria-specific topoisomerase exhibited poor mitochondrial ROS production and underwent apoptosis. Gln deprivation reduced glutathione production; supra-molar Asn supplementation allowed offspring production from SSCs cultured without Gln. Therefore, Gln ensures ROS-dependent SSC-self-renewal by providing protection against NOX1 and inducing Myc.


Assuntos
Glutamina , Espermatogônias , Masculino , Camundongos , Animais , Espermatogônias/metabolismo , Glutamina/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proliferação de Células , Células-Tronco , Células Cultivadas
5.
J Reprod Dev ; 2024 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-38735740

RESUMO

Intracytoplasmic sperm injection (ICSI) is clinically used to treat obstructive/nonobstructive azoospermia. This study compared the efficacy of ICSI with cauda epididymal and testicular sperm in Wistar (WI) and Brown-Norway (BN) rats. The transfer of ICSI oocytes with cryopreserved epididymal and testicular WI sperm resulted in offspring production of 26.2% and 3.7%-4.7%, respectively (P < 0.05). Treatments for artificial oocyte activation (AOA) and acrosome removal improved pronuclear formation in BN-ICSI oocytes; however, only AOA treatment was effective in producing offspring (3.7%-6.5%). In the case of ICSI with testicular sperm (TESE-ICSI), one offspring (0.6%) was derived from the BN-TESE-ICSI oocytes. The application of AOA or a hypo-osmotic sperm suspension did not improve the production of TESE-ICSI offspring. Thus, outbred WI rat offspring can be produced by using ICSI and less efficiently by using TESE-ICSI. Challenges in producing offspring by using ICSI/TESE-ICSI in inbred BN strains require further investigation.

6.
Genes Dev ; 30(23): 2637-2648, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-28007786

RESUMO

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.


Assuntos
Autorrenovação Celular/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Glicólise/genética , Proteína Proto-Oncogênica N-Myc/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Espermatogônias/citologia , Células-Tronco/metabolismo , Proteínas Quinases Dependentes de 3-Fosfoinositídeo/metabolismo , Animais , Divisão Celular/genética , Proliferação de Células/genética , Técnicas de Inativação de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Proto-Oncogênica N-Myc/genética , Proteínas Proto-Oncogênicas c-myc/genética , Fatores de Processamento de RNA/metabolismo , Células-Tronco/enzimologia
7.
Biol Reprod ; 108(4): 682-693, 2023 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-36648447

RESUMO

Characterization of spermatogonial stem cells (SSCs) has been hampered by their low frequency and lack of features that distinguish them from committed spermatogonia. Few conserved SSC markers have been discovered. To identify a new SSC marker, we evaluated SIRPA expression in mouse and rat SSCs. SIRPA was expressed in a small population of undifferentiated spermatogonia. SIRPA, and its ligand CD47 were expressed in cultured SSCs. Expression of both SIRPA and CD47 was upregulated by supplementation of GDNF and FGF2, which promoted SSC self-renewal. Sirpa depletion by short hairpin RNA impaired the proliferation of cultured SSCs, and these cells showed decreased MAP2K1 activation and PTPN11 phosphorylation. Immunoprecipitation experiments showed that SIRPA associates with PTPN11. Ptpn11 depletion impaired SSC activity in a manner similar to Sirpa depletion. SIRPA was expressed in undifferentiated spermatogonia in rat and monkey testes. Xenogenic transplantation experiments demonstrated that SIRPA is expressed in rat SSCs. These results suggest that SIRPA is a conserved SSC marker that promotes SSC self-renewal division by activating the MAP2K1 pathway via PTPN11.


Assuntos
Antígeno CD47 , Células-Tronco , Masculino , Camundongos , Ratos , Animais , Antígeno CD47/metabolismo , Células-Tronco/metabolismo , Proliferação de Células , Espermatogônias/metabolismo , Testículo/metabolismo , Células Cultivadas
8.
J Reprod Dev ; 69(6): 347-355, 2023 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-37899250

RESUMO

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.


Assuntos
Barreira Hematotesticular , Bussulfano , Masculino , Animais , Camundongos , Humanos , Barreira Hematotesticular/metabolismo , Bussulfano/farmacologia , Bussulfano/metabolismo , Espermatogônias/metabolismo , Testículo , Espermatogênese , Fertilidade , Transplante de Células , Células-Tronco , Tretinoína/farmacologia , Transplante de Células-Tronco
9.
Proc Natl Acad Sci U S A ; 117(14): 7837-7844, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32229564

RESUMO

The blood-testis barrier (BTB) is thought to be indispensable for spermatogenesis because it creates a special environment for meiosis and protects haploid cells from the immune system. The BTB divides the seminiferous tubules into the adluminal and basal compartments. Spermatogonial stem cells (SSCs) have a unique ability to transmigrate from the adluminal compartment to the basal compartment through the BTB upon transplantation into the seminiferous tubule. Here, we analyzed the role of Cldn11, a major component of the BTB, in spermatogenesis using spermatogonial transplantation. Cldn11-deficient mice are infertile due to the cessation of spermatogenesis at the spermatocyte stage. Cldn11-deficient SSCs failed to colonize wild-type testes efficiently, and Cldn11-deficient SSCs that underwent double depletion of Cldn3 and Cldn5 showed minimal colonization, suggesting that claudins on SSCs are necessary for transmigration. However, Cldn11-deficient Sertoli cells increased SSC homing efficiency by >3-fold, suggesting that CLDN11 in Sertoli cells inhibits transmigration of SSCs through the BTB. In contrast to endogenous SSCs in intact Cldn11-deficient testes, those from WT or Cldn11-deficient testes regenerated sperm in Cldn11-deficient testes. The success of this autologous transplantation appears to depend on removal of endogenous germ cells for recipient preparation, which reprogrammed claudin expression patterns in Sertoli cells. Consistent with this idea, in vivo depletion of Cldn3/5 regenerated endogenous spermatogenesis in Cldn11-deficient mice. Thus, coordinated claudin expression in both SSCs and Sertoli cells expression is necessary for SSC homing and regeneration of spermatogenesis, and autologous stem cell transplantation can rescue congenital defects of a self-renewing tissue.


Assuntos
Fertilidade/genética , Infertilidade/terapia , Espermatogônias/transplante , Transplante de Células-Tronco , Animais , Modelos Animais de Doenças , Fertilidade/fisiologia , Humanos , Infertilidade/genética , Infertilidade/patologia , Masculino , Camundongos , Espermatogênese/genética , Espermatogônias/crescimento & desenvolvimento , Espermatozoides/crescimento & desenvolvimento , Espermatozoides/transplante , Células-Tronco/citologia , Transplante Autólogo/métodos
10.
J Reprod Dev ; 68(6): 369-376, 2022 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-36223953

RESUMO

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.


Assuntos
Infertilidade Feminina , Camundongos , Feminino , Animais , Humanos , Infertilidade Feminina/genética , Infertilidade Feminina/terapia , Infertilidade Feminina/metabolismo , Adenoviridae/genética , Folículo Ovariano/metabolismo , Células da Granulosa/metabolismo , Oócitos/metabolismo , Fertilidade/genética
11.
Proc Natl Acad Sci U S A ; 116(33): 16404-16409, 2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31358627

RESUMO

Because spermatogonial stem cells (SSCs) are immortal by serial transplantation, SSC aging in intact testes is considered to be caused by a deteriorated microenvironment. Here, we report a cell-intrinsic mode of SSC aging by glycolysis activation. Using cultured SSCs, we found that aged SSCs proliferated more actively than young SSCs and showed enhanced glycolytic activity. Moreover, they remained euploid and exhibited stable androgenetic imprinting patterns with robust SSC activity despite having shortened telomeres. Aged SSCs showed increased Wnt7b expression, which was associated with decreased Polycomb complex 2 activity. Our results suggest that aberrant Wnt7b expression activated c-jun N-terminal kinase (JNK), which down-regulated mitochondria numbers by suppressing Ppargc1a Down-regulation of Ppargc1a probably decreased reactive oxygen species and enhanced glycolysis. Analyses of the Klotho-deficient aging mouse model and 2-y-old aged rats confirmed JNK hyperactivation and increased glycolysis. Therefore, not only microenvironment but also intrinsic activation of JNK-mediated glycolysis contributes to SSC aging.


Assuntos
Envelhecimento/genética , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Proteínas Proto-Oncogênicas/genética , Espermatogênese/genética , Proteínas Wnt/genética , Células-Tronco Germinativas Adultas/metabolismo , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Animais , Proliferação de Células/genética , Regulação da Expressão Gênica no Desenvolvimento , Glucuronidase/genética , Glicólise/genética , Proteínas Klotho , Masculino , Camundongos , Proteínas do Grupo Polycomb/genética , Ratos , Espécies Reativas de Oxigênio/metabolismo , Espermatogônias/crescimento & desenvolvimento , Espermatogônias/metabolismo , Nicho de Células-Tronco/genética , Testículo/crescimento & desenvolvimento , Testículo/metabolismo
12.
Biol Reprod ; 104(3): 706-716, 2021 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-33252132

RESUMO

Although reactive oxygen species (ROS) are required for spermatogonial stem cell (SSC) self-renewal, they induce DNA damage and are harmful to SSCs. However, little is known about how SSCs protect their genome during self-renewal. Here, we report that Ogg1 is essential for SSC protection against ROS. While cultured SSCs exhibited homologous recombination-based DNA double-strand break repair at levels comparable with those in pluripotent stem cells, they were significantly more resistant to hydrogen peroxide than pluripotent stem cells or mouse embryonic fibroblasts, suggesting that they exhibit high levels of base excision repair (BER) activity. Consistent with this observation, cultured SSCs showed significantly lower levels of point mutations than somatic cells, and showed strong expression of BER-related genes. Functional screening revealed that Ogg1 depletion significantly impairs survival of cultured SSCs upon hydrogen peroxide exposure. Thus, our results suggest increased expression of BER-related genes, including Ogg1, protects SSCs from ROS-induced damage.


Assuntos
Células-Tronco Germinativas Adultas/metabolismo , DNA Glicosilases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Quebras de DNA de Cadeia Dupla , DNA Glicosilases/genética , Reparo do DNA , Regulação da Expressão Gênica , Genoma , Peróxido de Hidrogênio/toxicidade , Masculino , Camundongos , Mutação
13.
Genes Dev ; 27(18): 1949-58, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-24029916

RESUMO

Spermatogonial stem cells (SSCs) present the potential to acquire pluripotency under specific culture conditions. However, the frequency of pluripotent cell derivation is low, and the mechanism of SSC reprogramming remains unknown. In this study, we report that induction of global DNA hypomethylation in germline stem (GS) cells (cultured SSCs) induces pluripotent cell derivation. When DNA demethylation was triggered by Dnmt1 depletion, GS cells underwent apoptosis. However, GS cells were converted into embryonic stem (ES)-like cells by double knockdown of Dnmt1 and p53. This treatment down-regulated Dmrt1, a gene involved in sexual differentiation, meiosis, and pluripotency. Dmrt1 depletion caused apoptosis of GS cells, but a combination of Dmrt1 and p53 depletion also induced pluripotency. Functional screening of putative Dmrt1 target genes revealed that Dmrt1 depletion up-regulates Sox2. Sox2 transfection up-regulated Oct4 and produced pluripotent cells. This conversion was enhanced by Oct1 depletion, suggesting that the balance of Oct proteins maintains SSC identity. These results suggest that spontaneous SSC reprogramming is caused by unstable DNA methylation and that a Dmrt1-Sox2 cascade is critical for regulating pluripotency in SSCs.


Assuntos
Células-Tronco Pluripotentes/fisiologia , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular , Reprogramação Celular/genética , Metilação de DNA , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fator 1 de Transcrição de Octâmero/genética , Fator 1 de Transcrição de Octâmero/metabolismo , Células-Tronco Pluripotentes/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Espermatogônias/metabolismo , Fatores de Transcrição/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
14.
Biol Reprod ; 102(1): 220-232, 2020 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-31403678

RESUMO

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.


Assuntos
Células-Tronco Germinativas Adultas/metabolismo , Receptores da Família Eph/metabolismo , Espermatogônias/metabolismo , Testículo/metabolismo , Células-Tronco Germinativas Adultas/citologia , Animais , Proliferação de Células/fisiologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Masculino , Camundongos , Fosforilação , Proto-Oncogene Mas , RNA Interferente Pequeno , Receptores da Família Eph/genética , Espermatogônias/citologia
15.
J Reprod Dev ; 66(4): 341-349, 2020 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-32213736

RESUMO

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.


Assuntos
Células-Tronco Germinativas Adultas/metabolismo , Antígenos CD2/metabolismo , Espermatogênese/fisiologia , Espermatogônias/metabolismo , Animais , Citometria de Fluxo , Masculino , Camundongos , Ratos
16.
Biol Reprod ; 100(2): 523-534, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30165393

RESUMO

Spermatogonial stem cells (SSCs) provide the foundation of spermatogenesis. However, because of their small number and slow self-renewal, transfection of SSCs has met with limited success. Although several viral vectors can infect SSCs, genome integration and an inability to maintain long-term gene expression have hampered studies on SSCs. Here we report successful SSC infection by Sendai virus (SV), an RNA virus in the Paramyxoviridae. The SV efficiently transduced germline stem (GS) cells, cultured spermatogonia with enriched SSC activity, and maintained gene expression for at least 5 months. It also infected freshly isolated SSCs from adult testes. The transfected GS cells reinitiated spermatogenesis following spermatogonial transplantation into seminiferous tubules of infertile mice, suggesting that SV transfection does not interfere with spermatogenesis progression. On the other hand, microinjection of SV into the seminiferous tubules of immature mice transduced SSCs and Sertoli cells, but did not transduce Leydig or peritubular cells by interstitial virus injection. SV-infected hamster GS cells, and freshly isolated rabbit or monkey SSC-like cells were identified following xenogeneic spermatogonial transplantation, suggesting that SV transduces SSCs from several mammalian species. Thus, SV is a useful vector that can transduce both SSCs and Sertoli cells and overcome problems associated with other viral vectors.


Assuntos
Células-Tronco Germinativas Adultas , Vírus Sendai/fisiologia , Transdução Genética/métodos , Animais , Cricetinae , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Transgênicos , Células de Sertoli , Espermatogênese/fisiologia , Espermatogônias/metabolismo
17.
J Reprod Dev ; 64(6): 511-522, 2018 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-30175719

RESUMO

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.


Assuntos
Barreira Hematotesticular/metabolismo , Células de Sertoli/metabolismo , Espermatogônias/metabolismo , Testículo/metabolismo , Animais , Barreira Hematotesticular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Claudina-5/metabolismo , Masculino , Camundongos , Morfolinas/farmacologia , Oligopeptídeos/farmacologia , Células de Sertoli/citologia , Células de Sertoli/efeitos dos fármacos , Espermatogênese/efeitos dos fármacos , Espermatogênese/fisiologia , Espermatogônias/citologia , Espermatogônias/efeitos dos fármacos , Nicho de Células-Tronco/efeitos dos fármacos , Testículo/efeitos dos fármacos , Triazinas/farmacologia
18.
Biol Reprod ; 97(6): 902-910, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29136097

RESUMO

Vast amounts of sperm are produced from spermatogonial stem cells (SSCs), which continuously undergo self-renewal. We examined the possible effect of laterality in male germline transmission efficiency of SSCs using a spermatogonial transplantation technique. We transplanted the same number of wild-type and Egfp transgenic SSCs in the same or different testes of individual recipient mice and compared the fertility of each type of recipient by natural mating. Transgenic mice were born within 3 months after transplantation regardless of the transplantation pattern. However, transgenic offspring were born at a significantly increased frequency when wild-type and transgenic SSCs were transplanted separately. In addition, this type of recipient sired significantly more litters that consisted exclusively of transgenic mice, which suggested that left and right testes have different time windows for fertilization. Thus, laterality plays an important role in germline transmission patterns from SSCs.


Assuntos
Células-Tronco Germinativas Adultas/transplante , Espermatogônias/transplante , Testículo/citologia , Animais , Feminino , Fertilidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
19.
Biol Reprod ; 96(1): 221-231, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28395324

RESUMO

Spermatogenesis is a complicated process that originates from spermatogonial stem cells (SSCs), which have self-renewal activity. Because SSCs are the only stem cells in the body that transmit genetic information to the next generation, they are an attractive target for germline modification. Although several virus vectors have been successfully used to transduce SSCs, cell toxicity or insertional mutagenesis of the transgene has limited their usage. Adeno-associated virus (AAV) is unique among virus vectors because of its target specificity and low toxicity in somatic cells, and clinical trials have shown that it has promise for gene therapy. However, there are conflicting reports on the possibility of germline integration of AAV into the genome of male germ cells, including SSCs. Here, we examined the usefulness of AAV vectors for exploring germline gene modification in SSCs. AAV1 infected cultured SSCs without apparent toxicity. Moreover, SSCs that were infected in fresh testis cells generated normal appearing spermatogenic colonies after spermatogonial transplantation. A microinsemination experiment produced offspring that underwent excision of the floxed target gene by AAV1-mediated Cre expression. Analysis of the offspring DNA showed no evidence of AAV integration, suggesting a low risk of germline integration by AAV infection. Although more extensive experiments are required to assess the risk of germline integration, our results show that AAV1 is useful for genetic manipulation of SSCs, and gene transduction by AAV will provide a useful approach to overcome potential problems associated with previous virus vector-mediated gene transduction.


Assuntos
Adenoviridae , Células-Tronco Germinativas Adultas , Técnicas de Transferência de Genes , Animais , Masculino , Camundongos Endogâmicos , Sorogrupo
20.
Proc Natl Acad Sci U S A ; 111(24): 8826-31, 2014 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-24879440

RESUMO

Spermatogonial stem cells (SSCs) undergo self-renewal divisions to support spermatogenesis throughout life. Although several positive regulators of SSC self-renewal have been discovered, little is known about the negative regulators. Here, we report that F-box and WD-40 domain protein 7 (FBXW7), a component of the Skp1-Cullin-F-box-type ubiquitin ligase, is a negative regulator of SSC self-renewal. FBXW7 is expressed in undifferentiated spermatogonia in a cell cycle-dependent manner. Although peptidyl-prolyl cis/trans isomerase NIMA-interacting 1 (PIN1), essential for spermatogenesis, is thought to destroy FBXW7, Pin1 depletion decreased FBXW7 expression. Spermatogonial transplantation showed that Fbxw7 overexpression compromised SSC activity whereas Fbxw7 deficiency enhanced SSC colonization and caused accumulation of undifferentiated spermatogonia, suggesting that the level of FBXW7 is critical for self-renewal and differentiation. Screening of putative FBXW7 targets revealed that Fbxw7 deficiency up-regulated myelocytomatosis oncogene (MYC) and cyclin E1 (CCNE1). Although depletion of Myc/Mycn or Ccne1/Ccne2 compromised SSC activity, overexpression of Myc, but not Ccne1, increased colonization of SSCs. These results suggest that FBXW7 regulates SSC self-renewal in a negative manner by degradation of MYC.


Assuntos
Proteínas F-Box/fisiologia , Espermatogônias/metabolismo , Células-Tronco/citologia , Ubiquitina-Proteína Ligases/fisiologia , Animais , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Proteínas F-Box/genética , Proteína 7 com Repetições F-Box-WD , Deleção de Genes , Proteínas de Fluorescência Verde/metabolismo , Homeostase , Cinética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-myc/metabolismo , Túbulos Seminíferos/metabolismo , Testículo/metabolismo , Ubiquitina-Proteína Ligases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA