Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Genes Dev ; 28(12): 1351-62, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24939937

RESUMO

The maintenance of cycling cell lineages relies on undifferentiated subpopulations consisting of stem and progenitor pools. Features that delineate these cell types are undefined for many lineages, including spermatogenesis, which is supported by an undifferentiated spermatogonial population. Here, we generated a transgenic mouse line in which spermatogonial stem cells are marked by expression of an inhibitor of differentiation 4 (Id4)-green fluorescent protein (Gfp) transgene. We found that Id4-Gfp(+) cells exist primarily as a subset of the type A(single) pool, and their frequency is greatest in neonatal development and then decreases in proportion during establishment of the spermatogenic lineage, eventually comprising ∼ 2% of the undifferentiated spermatogonial population in adulthood. RNA sequencing analysis revealed that expression of 11 and 25 genes is unique for the Id4-Gfp(+)/stem cell and Id4-Gfp(-)/progenitor fractions, respectively. Collectively, these findings provide the first definitive evidence that stem cells exist as a rare subset of the A(single) pool and reveal transcriptome features distinguishing stem cell and progenitor states within the mammalian male germline.


Assuntos
Células Germinativas/citologia , Proteínas Inibidoras de Diferenciação/metabolismo , Células-Tronco/citologia , Testículo/citologia , Animais , Diferenciação Celular , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas Inibidoras de Diferenciação/genética , Masculino , Camundongos , Camundongos Transgênicos , Espermatogênese/genética , Espermatogônias/metabolismo , Células-Tronco/metabolismo , Testículo/metabolismo , Transcriptoma
2.
Development ; 140(2): 280-90, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23221369

RESUMO

Continuity of cycling cell lineages relies on the activities of undifferentiated stem cell-containing subpopulations. Transition to a differentiating state must occur periodically in a fraction of the population to supply mature cells, coincident with maintenance of the undifferentiated state in others to sustain a foundational stem cell pool. At present, molecular mechanisms regulating these activities are poorly defined for most cell lineages. Spermatogenesis is a model process that is supported by an undifferentiated spermatogonial population and transition to a differentiating state involves attained expression of the KIT receptor. We found that impaired function of the X chromosome-clustered microRNAs 221 and 222 (miR-221/222) in mouse undifferentiated spermatogonia induces transition from a KIT(-) to a KIT(+) state and loss of stem cell capacity to regenerate spermatogenesis. Both Kit mRNA and KIT protein abundance are influenced by miR-221/222 function in spermatogonia. Growth factors that promote maintenance of undifferentiated spermatogonia upregulate miR-221/222 expression; whereas exposure to retinoic acid, an inducer of spermatogonial differentiation, downregulates miR-221/222 abundance. Furthermore, undifferentiated spermatogonia overexpressing miR-221/222 are resistant to retinoic acid-induced transition to a KIT(+) state and are incapable of differentiation in vivo. These findings indicate that miR-221/222 plays a crucial role in maintaining the undifferentiated state of mammalian spermatogonia through repression of KIT expression.


Assuntos
Células Germinativas/citologia , MicroRNAs/fisiologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Animais , Apoptose , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Citometria de Fluxo/métodos , Humanos , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Cromossomos Sexuais , Espermatogênese , Espermatogônias/patologia , Células-Tronco , Tretinoína/farmacologia
3.
Biol Reprod ; 95(1): 14, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27251094

RESUMO

Continual and robust spermatogenesis relies on the actions of an undifferentiated spermatogonial population that contains stem cells. A remarkable feature of spermatogonial stem cells (SSCs) is the capacity to regenerate spermatogenesis following isolation from a donor testis and transplantation into a permissive recipient testis. This capacity has enormous potential as a tool for enhancing the reproductive capacity of livestock, which can improve production efficiency. Because SSCs are a rare subset of the undifferentiated spermatogonial population, a period of in vitro amplification in number following isolation from donor testicular tissue is essential. Here, we describe methodology for isolation of a cell fraction from prepubertal bull testes that is enriched for undifferentiated spermatogonia and long-term maintenance of the cells in both the feeder cell coculture and the feeder-free format. To achieve this method, we derived bovine fetal fibroblasts (BFF) to serve as feeders for optimizing medium conditions that promote maintenance of bovine undifferentiated spermatogonia for at least 2 mo. In addition, we devised a feeder-free system with BFF-conditioned medium that sustained bovine undifferentiated spermatogonia for at least 1 mo in vitro. The methodologies described could be optimized to provide platforms for exponential expansion of bovine SSCs that will provide the numbers needed for transplantation into recipient testes.


Assuntos
Técnicas de Cultura de Células/métodos , Espermatogênese/fisiologia , Espermatogônias/citologia , Células-Tronco/citologia , Testículo/citologia , Animais , Bovinos , Células Cultivadas , Meios de Cultura , Masculino
4.
Biol Reprod ; 95(6): 117, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27733379

RESUMO

Precise separation of spermatogonial stem cells (SSCs) from progenitor spermatogonia that lack stem cell activity and are committed to differentiation remains a challenge. To distinguish between these spermatogonial subtypes, we identified genes that exhibited bimodal mRNA levels at the single-cell level among undifferentiated spermatogonia from Postnatal Day 6 mouse testes, including Tspan8, Epha2, and Pvr, each of which encode cell surface proteins useful for cell selection. Transplantation studies provided definitive evidence that a TSPAN8-high subpopulation is enriched for SSCs. RNA-seq analyses identified genes differentially expressed between TSPAN8-high and -low subpopulations that clustered into multiple biological pathways potentially involved in SSC renewal or differentiation, respectively. Methyl-seq analysis identified hypomethylated domains in the promoters of these genes in both subpopulations that colocalized with peaks of histone modifications defined by ChIP-seq analysis. Taken together, these results demonstrate functional heterogeneity among mouse undifferentiated spermatogonia and point to key biological characteristics that distinguish SSCs from progenitor spermatogonia.


Assuntos
Células-Tronco Germinativas Adultas/citologia , Testículo/citologia , Tetraspaninas/metabolismo , Células-Tronco Germinativas Adultas/metabolismo , Animais , Biomarcadores/metabolismo , Ciclo Celular/fisiologia , Perfilação da Expressão Gênica , Masculino , Camundongos , Receptor EphA2/genética , Receptor EphA2/metabolismo , Espermatogênese , Testículo/metabolismo , Tetraspaninas/genética
5.
J Cell Sci ; 126(Pt 4): 1009-20, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23239029

RESUMO

Continual spermatogenesis relies on the activities of a tissue-specific stem cell population referred to as spermatogonial stem cells (SSCs). Fate decisions of stem cells are influenced by their niche environments, a major component of which is soluble factors secreted by support cells. At present, the factors that constitute the SSC niche are undefined. We explored the role of chemokine (C-X-C motif) ligand 12 (CXCL12) signaling via its receptor C-X-C chemokine receptor type 4 (CXCR4) in regulation of mouse SSC fate decisions. Immunofluorescent staining for CXCL12 protein in cross sections of testes from both pup and adult mice revealed its localization at the basement membrane of seminiferous tubules. Within the undifferentiated spermatogonial population of mouse testes, a fraction of cells were found to express CXCR4 and possess stem cell capacity. Inhibition of CXCR4 signaling in primary cultures of mouse undifferentiated spermatogonia resulted in SSC loss, in part by reducing proliferation and increasing the transition to a progenitor state primed for differentiation upon stimulation by retinoic acid. In addition, CXCL12-CXCR4 signaling in mouse SSCs was found to be important for colonization of recipient testes following transplantation, possibly by influencing homing to establish stem-cell niches. Furthermore, inhibition of CXCR4 signaling in testes of adult mice impaired SSC maintenance, leading to loss of the germline. Collectively, these findings indicate that CXCL12 is an important component of the growth factor milieu of stem cells in mammalian testes and that it signals via the CXCR4 to regulate maintenance of the SSC pool.


Assuntos
Quimiocina CXCL12/metabolismo , Receptores CXCR4/metabolismo , Espermatogônias/citologia , Animais , Western Blotting , Proliferação de Células , Células Cultivadas , Quimiocina CXCL12/genética , Citometria de Fluxo , Imuno-Histoquímica , Masculino , Camundongos , Receptores CXCR4/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espermatogênese/genética , Espermatogênese/fisiologia
6.
Biol Reprod ; 86(5): 164, 1-11, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22378757

RESUMO

Spermatogenesis relies on coordinated differentiation of stem and progenitor spermatogonia, and the transcription factor STAT3 is essential for this process in mammals. Here we studied the THY1+ spermatogonial population in mouse testes, which contains spermatogonial stem cells (SSC) and non-stem cell progenitor spermatogonia, to further define the downstream mechanism regulating differentiation. Transcript abundance for the bHLH transcription factor Neurog3 was found to be significantly reduced upon transient inhibition of STAT3 signaling in these cells and exposure to GDNF, a key growth factor regulating self-renewal of SSCs, suppressed activation of STAT3 and in accordance Neurog3 gene expression. Moreover, STAT3 was found to bind the distal Neurog3 promoter/enhancer region in THY1+ spermatogonia and regulate transcription. Transient inhibition of Neurog3 expression in cultures of proliferating THY1+ spermatogonia increased stem cell content after several self-renewal cycles without effecting overall proliferation of the cells, indicating impaired differentiation of SSCs to produce progenitor spermatogonia. Furthermore, cultured THY1+ spermatogonia with induced deficiency of Neurog3 were found to be incapable of differentiation in vivo following transplantation into testes of recipient mice. Collectively, these results establish a mechanism by which activation of STAT3 regulates the expression of NEUROG3 to subsequently drive differentiation of SSC and progenitor spermatogonia in the mammalian germline.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Fator de Transcrição STAT3/fisiologia , Espermatogênese/fisiologia , Espermatogônias/fisiologia , Células-Tronco/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/farmacologia , Masculino , Camundongos , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Regiões Promotoras Genéticas , Espermatogênese/efeitos dos fármacos , Espermatogônias/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Antígenos Thy-1/análise
7.
Biol Reprod ; 85(2): 347-56, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21543770

RESUMO

Continual spermatogenesis at a quantitatively normal level is required to sustain male fertility. The foundation of this process relies on maintenance of an undifferentiated spermatogonial population consisting of spermatogonial stem cells (SSCs) that self-renew as well as transient amplifying progenitors produced by differentiation. In mammals, type A(single) spermatogonia form the SSC population, but molecular markers distinguishing these from differentiating progenitors are undefined and knowledge of mechanisms regulating their functions is limited. We show that in the mouse male germline the transcriptional repressor ID4 is expressed by a subpopulation of undifferentiated spermatogonia and selectively marks A(single) spermatogonia. In addition, we found that ID4 expression is up-regulated in isolated SSC-enriched fractions by stimulation from GDNF, a key growth factor driving self-renewal. In mice lacking ID4 expression, quantitatively normal spermatogenesis was found to be impaired due to progressive loss of the undifferentiated spermatogonial population during adulthood. Moreover, reduction of ID4 expression by small interfering RNA treatment abolished the ability of wild-type SSCs to expand in vitro during long-term culture without affecting their survival. Collectively, these results indicate that ID4 is a distinguishing marker of SSCs in the mammalian germline and plays an important role in the regulation of self-renewal.


Assuntos
Regulação da Expressão Gênica/fisiologia , Proteínas Inibidoras de Diferenciação/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Biomarcadores , Inativação Gênica , Proteínas Inibidoras de Diferenciação/genética , Masculino , Camundongos , RNA Interferente Pequeno , Espermatogônias/metabolismo , Testículo/metabolismo
8.
Biol Reprod ; 83(3): 427-33, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20505165

RESUMO

Homeostasis of many tissues is maintained by self-renewal and differentiation of stem cells. Spermatogenesis is one such system relying on the activity of spermatogonial stem cells (SSCs). Several key regulators of SSC self-renewal have been identified, yet knowledge of molecules that control SSC differentiation is undefined. In this study, we found that transient impairment of STAT3 signaling enhances SSC self-renewal in vitro without affecting general spermatogonial proliferation, indicating an alteration in the balance of SSC fate decisions that inhibited differentiation. Confirming this observation, short hairpin RNA-mediated stable reduction of STAT3 expression in cultured SSCs abolished their ability to differentiate beyond the undifferentiated spermatogonial stage following transplantation into recipient testes. Collectively, these results demonstrate that STAT3 promotes the differentiation of SSCs. In contrast, STAT3 plays a central role in maintaining self-renewal of mouse embryonic stem cells, and STAT signaling is essential for self-renewal of male germline stem cells in Drosophila.


Assuntos
Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/fisiologia , Espermatogênese/fisiologia , Espermatogônias/metabolismo , Análise de Variância , Animais , Western Blotting , Proliferação de Células , Células Cultivadas , Citometria de Fluxo , Imuno-Histoquímica , Masculino , Camundongos , Fosforilação/fisiologia , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espermatogônias/transplante , Transfecção
9.
Biol Reprod ; 82(6): 1103-11, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20181621

RESUMO

Continual spermatogenesis relies on a pool of spermatogonial stem cells (SSCs) that possess the capacity for self-renewal and differentiation. Maintenance of this pool depends on survival of SSCs throughout the lifetime of a male. Response to extrinsic stimulation from glial cell line-derived neurotrophic factor (GDNF), mediated by the PIK3/AKT signaling cascade, is a key pathway of SSC survival. In this study, we found that expression of the POU domain transcription factor POU3F1 in cultured SSCs is up-regulated via this mechanism. Reduction of Pou3f1 gene expression by short interfering RNA (siRNA) treatment induced apoptosis in cultured germ cell populations, and transplantation analyses revealed impaired SSC maintenance in vitro. POU3F1 expression was localized to spermatogonia in cross-sections of prepubertal and adult testes, implying a similar role in vivo. Through comparative analyses, we found that expression of POU5F1, another POU transcription factor implicated as essential for SSC self-renewal, is not regulated by GDNF in cultured SSCs. Transplantation analyses following siRNA treatment showed that POU5F1 expression is not essential for SSC maintenance in vitro. Additionally, expression of NODAL, a putative autocrine regulator of POU5F1 expression in mouse germ cells, could not be detected in SSCs isolated from testes or cultured SSCs. Collectively, these results indicate that POU3F1, but not POU5F1, is an intrinsic regulator of GDNF-induced survival and self-renewal of mouse SSCs.


Assuntos
Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Fator 6 de Transcrição de Octâmero/metabolismo , Espermatogênese , Espermatogônias/metabolismo , Células-Tronco/metabolismo , Animais , Apoptose , Comunicação Autócrina , Divisão Celular , Sobrevivência Celular , Masculino , Camundongos , Proteína Nodal/análise , Testículo/citologia , Testículo/metabolismo
10.
J Anim Sci ; 97(4): 1468-1477, 2019 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-30576512

RESUMO

Male reproductive capacity is a critical component of cattle production and the majority of genetic gain is made via selective utilization of gametes from desirable sires. Thus, strategies that enhance sperm production increase the availability of elite genetics for use in improving production characteristics of populations on a worldwide scale. In all mammals, the amount of sperm produced is strongly correlated to the number of Sertoli cells in testes. Studies with rodents showed that the size of the Sertoli cell population is set during prepubertal development via signaling from thyroid hormones. Here, we devised a strategy to increase Sertoli cell number in bulls via induction of a transient hypothyroidic state just prior to and extending beyond the period of Sertoli cell proliferation that we found to normally cease between 4.5 and 5 mo of age. In adulthood, these bulls produced a significantly greater number of sperm compared to age-matched controls and their testes contained nearly 2 times more Sertoli cells. Importantly, sperm motility, morphology, fertilizing ability, and viability after cryopreservation were found to be no different for treated bulls compared to untreated control bulls. This strategy of transient induction of hypothyroidism during a defined period of prepubertal development in bulls could prove to be an efficacious approach for enhancing daily sperm production in genetically desirable sires that will, in turn, provide an avenue for improving the efficiency of commercial cattle production.

11.
Contraception ; 97(4): 363-369, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29407362

RESUMO

OBJECTIVE: The objective was to develop a method to simultaneously quantify five commonly used hormonal contraceptives (HCs) and two endogenous sex steroids by liquid chromatography-tandem triple quadrupole mass spectrometry (LC-MS/MS) and apply this method to human serum samples. STUDY DESIGN: We developed a method to simultaneously analyze ethinyl estradiol (EE2), etonogestrel (ENG), levonorgestrel (LNG), medroxyprogesterone acetate (MPA) and norethisterone (NET), along with estradiol (E2) and progesterone (P4), in human serum for a Shimadzu Nexera-LCMS-8050 LC-MS/MS platform. We analyzed serum collected from women self-reporting use of oral contraceptives, contraceptive implants or injectable contraceptives (n=14) and normally cycling women using no HC (n=15) as well as pooled samples from women administered various HCs (ENG, n=6; LNG, n=14; MPA, n=7; NET, n=5). RESULTS: Limits of quantitation were 0.010ng/mL for E2, EE2 and P4; 0.020ng/mL for ENG, LNG and MPA; and 0.040ng/mL for NET. Precisions for all assays, as indicated by coefficient of variation, were less than or equal to 12.1%. Accuracies for all assays were in the range of 95%-108%. Endogenous hormone values obtained from analysis of human serum samples are in agreement with levels previously reported in the literature for normally cycling women as well as for women taking the appropriate HC. CONCLUSIONS: We have developed a robust, accurate and sensitive method for simultaneously analyzing commonly used contraceptive steroids and endogenous sex steroids in human serum. IMPLICATIONS: This analytical method can be used for quantitating contraceptive steroid levels in women for monitoring systemic exposure to determine drug interactions, nonadherence, misreporting and proper dosing.


Assuntos
Anticoncepcionais Orais Combinados/sangue , Anticoncepcionais Orais/sangue , Estradiol/sangue , Progesterona/sangue , Adulto , Cromatografia Líquida , Feminino , Humanos , Esteroides/sangue , Espectrometria de Massas em Tandem
12.
Sci Rep ; 7: 40176, 2017 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-28071690

RESUMO

Genome editing tools have revolutionized the generation of genetically modified animals including livestock. In particular, the domestic pig is a proven model of human physiology and an agriculturally important species. In this study, we utilized the CRISPR/Cas9 system to edit the NANOS2 gene in pig embryos to generate offspring with mono-allelic and bi-allelic mutations. We found that NANOS2 knockout pigs phenocopy knockout mice with male specific germline ablation but other aspects of testicular development are normal. Moreover, male pigs with one intact NANOS2 allele and female knockout pigs are fertile. From an agriculture perspective, NANOS2 knockout male pigs are expected to serve as an ideal surrogate for transplantation of donor spermatogonial stem cells to expand the availability of gametes from genetically desirable sires.


Assuntos
Animais Geneticamente Modificados , Técnicas de Inativação de Genes , Proteínas de Ligação a RNA/genética , Sus scrofa/genética , Animais , Sistemas CRISPR-Cas , Fertilidade , Infertilidade Masculina , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA