Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
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.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
Biol Reprod ; 94(5): 112, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27053363

RESUMO

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.


Assuntos
Células-Tronco Germinativas Adultas/transplante , Fertilidade , Infertilidade Masculina/terapia , Transplante de Células-Tronco/métodos , Animais , Feminino , Infertilidade Masculina/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Gravidez , Espermatogênese/fisiologia , Espermatogônias/fisiologia , Testículo/patologia
10.
Biol Reprod ; 94(1): 13, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26607720

RESUMO

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.


Assuntos
Antracenos/química , Corantes Fluorescentes/química , Morfolinas/química , Espermatogônias/ultraestrutura , Células-Tronco/ultraestrutura , Animais , Proteínas Cdh1/metabolismo , Células Cultivadas , Criptorquidismo/patologia , Células Germinativas/ultraestrutura , Magnetismo , Masculino , Camundongos , Camundongos Transgênicos , Rodamina 123/química , Espermatogônias/transplante , Testículo/citologia , Tetraspanina 29/metabolismo
11.
Biol Reprod ; 92(6): 147, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25947060

RESUMO

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.


Assuntos
NADPH Oxidases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Espermatogênese/fisiologia , Espermatogônias/metabolismo , Células-Tronco/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Masculino , Camundongos , NADH NADPH Oxirredutases/genética , NADH NADPH Oxirredutases/metabolismo , NADPH Oxidase 1 , NADPH Oxidases/genética , Espermatogônias/citologia , Células-Tronco/citologia
12.
Biol Reprod ; 91(4): 88, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25210127

RESUMO

Spermatogonial stem cells (SSCs) undergo self-renewal division, which can be recapitulated in vitro. Attempts to establish serum-free culture conditions for SSCs have met with limited success. Although we previously reported that SSCs can be cultured without serum on laminin-coated plates, the growth rate and SSC concentration were relatively low, which made it inefficient for culturing large numbers of SSCs. In this study, we report on a novel culture medium that showed improved SSC maintenance. We used Iscove modified Dulbecco medium, supplemented with lipid mixture, fetuin, and knockout serum replacement. In the presence of glial cell line-derived neurotrophic factor (GDNF) and fibroblast growth factor 2 (FGF2), SSCs cultured on laminin-coated plates could proliferate for more than 5 mo and maintained normal karyotype and androgenetic DNA methylation patterns in imprinted genes. Germ cell transplantation showed that SSCs in the serum-free medium proliferated more actively than those in the serum-supplemented medium and that the frequency of SSCs was comparable between the two culture media. Cultured cells underwent germline transmission. Development of a new serum- and feeder-free culture method for SSCs will facilitate studies into the effects of microenvironments on self-renewal and will stimulate further improvements to derive SSC cultures from different animal species.


Assuntos
Células-Tronco Adultas/fisiologia , Técnicas de Cultura de Células/métodos , Meios de Cultura Livres de Soro , Meios de Cultura/química , Animais , Proliferação de Células , Transplante de Células , MAP Quinase Quinase 1/genética , MAP Quinase Quinase 1/metabolismo , Masculino , Camundongos
13.
Stem Cell Reports ; 19(4): 443-455, 2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38458191

RESUMO

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.


Assuntos
Sêmen , Espermatogônias , Humanos , Camundongos , Masculino , Animais , Espermatogônias/metabolismo , Testículo/metabolismo , Fertilidade , Transplante de Células-Tronco , Espermatogênese
14.
Stem Cell Reports ; 18(4): 985-998, 2023 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-36963391

RESUMO

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."


Assuntos
Transplante de Células-Tronco Hematopoéticas , Proteína Quinase 14 Ativada por Mitógeno , Masculino , Camundongos , Animais , Espermatogônias , Espécies Reativas de Oxigênio/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Proliferação de Células , Testículo , Espermatogênese/genética
15.
Dev Cell ; 13(6): 796-811, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18061563

RESUMO

Cultured adherent cells divide on the substratum, leading to formation of the cell monolayer. However, how the orientation of this anchorage-dependent cell division is regulated remains unknown. We have previously shown that integrin-dependent adhesion orients the spindle parallel to the substratum, which ensures this anchorage-dependent cell division. Here, we show that phosphatidylinositol-3,4,5-triphosphate (PtdIns(3,4,5)P3) is essential for this spindle orientation control. In metaphase, PtdIns(3,4,5)P3 is accumulated in the midcortex in an integrin-dependent manner. Inhibition of phosphatidylinositol-3-OH kinase (PI(3)K) reduces the accumulation of PtdIns(3,4,5)P3 and induces spindle misorientation. Introduction of PtdIns(3,4,5)P3 to these cells restores the midcortical accumulation of PtdIns(3,4,5)P3 and proper spindle orientation. PI(3)K inhibition causes dynein-dependent spindle rotations along the z-axis, resulting in spindle misorientation. Moreover, dynactin, a dynein-binding partner, is accumulated in the midcortex in a PtdIns(3,4,5)P3-dependent manner. We propose that PtdIns(3,4,5)P3 directs dynein/dynactin-dependent pulling forces on spindles to the midcortex, and thereby orients the spindle parallel to the substratum.


Assuntos
Actinas/metabolismo , Polaridade Celular , Metáfase/fisiologia , Proteínas Associadas aos Microtúbulos/metabolismo , Fosfatos de Fosfatidilinositol/fisiologia , Fuso Acromático/metabolismo , Adesão Celular/fisiologia , Células Cultivadas , Citoesqueleto/metabolismo , Complexo Dinactina , Dineínas/metabolismo , Células HeLa , Humanos , Immunoblotting , Integrinas/metabolismo , Córtex Renal/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , RNA Interferente Pequeno/farmacologia , Transfecção
16.
Biol Reprod ; 87(6): 139, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23053437

RESUMO

Spermatogonial stem cells (SSCs) provide the foundation of spermatogenesis, but studies are hampered by their scarcity. Although the cryptorchid operation is often used to obtain an enriched SSC population, making cryptorchid testes is time-consuming and the technique is not applicable to many animal species. In the present study, we screened for a new surface antigen on SSCs using germline stem (GS) cells (i.e., cultured SSCs). Germ cell transplantation experiments showed that SSCs express melanoma cell adhesion molecule (MCAM), which belongs to the immunoglobulin superfamily and mediates cation-independent adhesion. Although MCAM overexpression in GS cells did not influence SSC colony formation frequency or subsequent spermatogenesis after transplantation, MCAM knockdown in GS cells by short-interfering RNA treatment reduced colony numbers, suggesting that MCAM plays a role in sustaining SSC potential. Multiparameter selection of wild-type adult testis cells with a CD9⁺EPCAM(low)MCAM⁺KIT⁻ phenotype resulted in a 561-fold enrichment of SSCs. Development of a new strategy for SSC enrichment from mature adult testes will facilitate analyses of SSCs in the normal testicular microenvironment.


Assuntos
Membrana Celular/metabolismo , Espermatogênese , Espermatogônias/metabolismo , Células-Tronco/metabolismo , Animais , Antígenos de Neoplasias/metabolismo , Antígeno CD146/química , Antígeno CD146/genética , Antígeno CD146/metabolismo , Moléculas de Adesão Celular/metabolismo , Células Cultivadas , Molécula de Adesão da Célula Epitelial , Citometria de Fluxo , Separação Imunomagnética , Infertilidade Masculina/terapia , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Interferência de RNA , RNA Interferente Pequeno , Proteínas Recombinantes/metabolismo , Espermatogônias/citologia , Espermatogônias/transplante , Transplante de Células-Tronco/efeitos adversos , Células-Tronco/citologia , Tetraspanina 29/metabolismo
17.
Biol Reprod ; 86(5): 148, 1-11, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22357549

RESUMO

Germ cell tumors (GCTs) are unique in that they exhibit diverse biological characteristics and pathological features. Although several in vivo GCT models are available, studies on GCTs are hampered because in vivo development of GCTs is time consuming and prevents a detailed molecular analysis of the transformation process. Here we developed a novel strategy to transform mouse testis cells in vitro. Lentivirus-mediated transfection of dominant negative Trp53, Myc, and activated Hras1 into a CD9-expressing testis cells caused tumorigenic conversion in vitro. Although these cells resembled embryonic stem (ES) cells, they were aneuploid and lacked Nanog expression, which is involved in the maintenance of the undifferentiated state in ES cells. Euploid ES-like cells were produced by transfecting the Yamanaka factors (Pou5f1, Myc, Klf4, and Sox2) into the same cell population. Although these cells expressed Nanog, they were distinct from ES cells in that they expressed CD44, a cancer stem cell antigen. Both treatments induced similar changes in the DNA methylation patterns in differentially methylated regions of imprinted genes. Moreover, despite the differences in their phenotype and karyotype, both cell types similarly produced mixed GCTs on transplantation, which were composed of teratomas, seminomas, and embryonal carcinomas. Thus, in vitro testis cell transformation facilitates an analysis of the GCT formation process, and our results also suggest the close similarity between GCT formation and reprogramming.


Assuntos
Transformação Celular Neoplásica/genética , Neoplasias Embrionárias de Células Germinativas/genética , Oncogenes , Neoplasias Testiculares/genética , Testículo , Transfecção/métodos , Aneuploidia , Animais , Carcinoma Embrionário/genética , Metilação de DNA , Impressão Genômica , Proteínas de Homeodomínio/análise , Receptores de Hialuronatos/análise , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Infecções por Lentivirus , Masculino , Camundongos , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/genética , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Fatores de Transcrição SOXB1/genética , Seminoma/genética , Teratoma/genética , Tetraspanina 29/análise
18.
Biol Reprod ; 86(6): 178, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22441799

RESUMO

Somatic cell hybridization is widely used to study the control of gene regulation and the stability of differentiated states. In contrast, the application of this method to germ cells has been limited in part because of an inability to culture germ cells. In this study, we produced germ cell hybrids using germ-line stem (GS) cells and multipotent germ-line stem (mGS) cells. While GS cells are enriched for spermatogonial stem cell (SSC) activity, mGS cells are similar to embryonic stem (ES) cells and originally derived from GS cells. Hybrids were successfully obtained between GS cells and ES cells, between GS cells and mGS cells, and between mGS cells and thymocytes. All exhibited ES cell markers and a behavior similar to ES cells, formed teratomas, and differentiated into somatic cell tissues. However, none of the hybrid cells were able to reconstitute spermatogenesis after microinjection into seminiferous tubules. Analyses of the DNA methylation patterns of imprinted genes also showed that mGS cells do not possess a DNA demethylation ability, which was found in embryonic germ cells derived from primordial germ cells. However, mGS cells reactivated the X chromosome and induced Pou5f1 expression in female thymocytes in a manner similar to ES cells. These data show that mGS cells possess ES-like reprogramming potential, which predominates over-SSC activity.


Assuntos
Técnicas de Cultura de Células , Células-Tronco Embrionárias , Células Germinativas , Células Híbridas , Células-Tronco Multipotentes , Animais , Fusão Celular , Feminino , Impressão Genômica , Células Híbridas/citologia , Células Híbridas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fator 3 de Transcrição de Octâmero/metabolismo , Fenótipo , Teratoma/etiologia , Testículo/citologia , Cromossomo X/metabolismo
19.
Cell Rep Med ; 3(5): 100606, 2022 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-35584625

RESUMO

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.


Assuntos
Infertilidade Feminina , Ovário , Animais , Dependovirus/genética , Feminino , Fertilidade/genética , Humanos , Infertilidade Feminina/genética , Camundongos , Folículo Ovariano
20.
Stem Cell Reports ; 17(4): 924-935, 2022 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-35334214

RESUMO

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.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Testículo , Animais , Masculino , Camundongos , Organoides , Ratos , Espermatogênese/genética , Espermatogônias/transplante , Transplante de Células-Tronco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA