Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 119(15): e2116826119, 2022 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-35377789

RESUMO

During spermatogenesis, spermatogonia undergo a series of mitotic and meiotic divisions on their path to spermatozoa. To achieve this, a succession of processes requiring high proteolytic activity are in part orchestrated by the proteasome. The spermatoproteasome (s20S) is specific to the developing gametes, in which the gamete-specific α4s subunit replaces the α4 isoform found in the constitutive proteasome (c20S). Although the s20S is conserved across species and was shown to be crucial for germ cell development, its mechanism, function, and structure remain incompletely characterized. Here, we used advanced mass spectrometry (MS) methods to map the composition of proteasome complexes and their interactomes throughout spermatogenesis. We observed that the s20S becomes highly activated as germ cells enter meiosis, mainly through a particularly extensive 19S activation and, to a lesser extent, PA200 binding. Additionally, the proteasome population shifts from c20S (98%) to s20S (>82 to 92%) during differentiation, presumably due to the shift from α4 to α4s expression. We demonstrated that s20S, but not c20S, interacts with components of the meiotic synaptonemal complex, where it may localize via association with the PI31 adaptor protein. In vitro, s20S preferentially binds to 19S and displays higher trypsin- and chymotrypsin-like activities, both with and without PA200 activation. Moreover, using MS methods to monitor protein dynamics, we identified significant differences in domain flexibility between α4 and α4s. We propose that these differences induced by α4s incorporation result in significant changes in the way the s20S interacts with its partners and dictate its role in germ cell differentiation.


Assuntos
Complexo de Endopeptidases do Proteassoma , Espermatogênese , Espermatogônias , Humanos , Masculino , Complexo de Endopeptidases do Proteassoma/química , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Espermatogônias/enzimologia
2.
Int J Toxicol ; 40(4): 344-354, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33866838

RESUMO

Phosphatidylinositol 3-kinase (PI3K) δ is a lipid kinase primarily found in leukocytes, which regulates important cell functions. AMG2519493 was a PI3K δ-specific inhibitor in development for treatment of various inflammatory diseases. AMG2519493-related changes in the male and/or female reproductive organs were observed in the 1- and 3-month oral repeat dose toxicology studies in the rat and cynomolgus monkey. Hemorrhagic corpora lutea cysts and increased incidence of corpora lutea cysts without hemorrhage were observed in the ovaries at supra pharmacological doses in the rat. A decrease in seminiferous germ cells in the testis, indicative of spermatogenesis maturation arrest, was observed in both the rat and cynomolgus monkey. Although the characteristics were comparable, the drug systemic exposures associated with the testicular changes were very different between the 2 species. In the rat, the testicular change was only observed at supra pharmacologic exposure. Isotype assessment of PI3K signaling in rat spermatogonia in vitro indicated a role for PI3K ß, but not δ, in the c Kit/PI3K/protein kinase B signaling pathway. Therefore, changes in both the ovary and testis of the rat were considered due to off target effect as they only occurred at suprapharmacologic exposure. In contrast, the testicular changes in the cynomolgus monkey (decrease in seminiferous germ cells) occurred at very low doses associated with PI3K δ-specific inhibition, indicating that the PI3K δ isoform may be important in spermatogenesis maturation in the cynomolgus monkey. Our results suggest species-related differences in PI3K isoform-specific control on reproductive organs.


Assuntos
Ovário/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase/farmacologia , Pirimidinas/farmacologia , Quinolinas/farmacologia , Testículo/efeitos dos fármacos , Animais , Feminino , Macaca fascicularis , Masculino , Camundongos , Ovário/enzimologia , Ratos , Ratos Sprague-Dawley , Espermatogônias/enzimologia , Testículo/enzimologia
3.
Sci Rep ; 10(1): 10796, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32612133

RESUMO

Germ cell depletion in recipient testes is indispensable for successful transplantation of spermatogonial stem cells. However, we found that such treatment had an adverse effect on spermatogenesis of orthotopically transplanted donor testis tissues. In the donor tissue, the frequency of stimulated by retinoic acid (RA) 8 (STRA8) expression was reduced in germ cells, suggesting that RA signalling indispensable for spermatogenesis was attenuated in germ cell-depleted recipient testes. In this context, germ cell depletion diminished expression of testicular Aldh1a2, which is responsible for testicular RA synthesis, while Cyp26b1, which is responsible for testicular RA metabolism, was still expressed even after germ cell depletion, suggesting an alteration of the RA synthesis/metabolism ratio. These observations suggested that RA insufficiency was one of the causes of the defective donor spermatogenesis. Indeed, repetitive RA administrations significantly improved donor spermatogenesis to produce fertile offspring without any side effects. These findings may contribute to improving fertility preservation techniques for males, especially to prevent iatrogenic infertility induced by chemotherapy in prepubertal cancer patients.


Assuntos
Transplante de Órgãos , Espermatogênese , Espermatogônias/enzimologia , Testículo , Tretinoína/metabolismo , Família Aldeído Desidrogenase 1/biossíntese , Animais , Regulação Enzimológica da Expressão Gênica , Humanos , Masculino , Camundongos , Retinal Desidrogenase/biossíntese , Ácido Retinoico 4 Hidroxilase/biossíntese , Testículo/enzimologia , Testículo/transplante
4.
Ecotoxicol Environ Saf ; 190: 110063, 2020 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-31846860

RESUMO

Arsenic is a toxic metalloid that can cause male reproductive malfunctions and is widely distributed in the environment. The aim of this study was to investigate the cytotoxicity of arsenic trioxide (ATO) induced GC-1 spermatogonial (spg) cells. Our results found that ATO increased the levels of catalase (CAT) and malonaldehyde (MDA) and reactive oxygen species (ROS), while decreasing glutathione (GSH) and the total antioxidant capacity (T-AOC). Therefore, ATO triggered oxidative stress in GC-1 spg cells. In addition, ATO also caused severe mitochondrial dysfunction that included an increase in residual oxygen consumption (ROX), and decreased the routine respiration, maximal and ATP-linked respiration (ATP-L-R), as well as spare respiratory capacity (SRC), and respiratory control rate (RCR); ATO also damaged the mitochondrial structure, including mitochondrial cristae disordered and dissolved, mitochondrial vacuolar degeneration. Moreover, degradation of p62, LC3 conversion, increasing the number of acidic vesicle organelles (AVOs) and autophagosomes and autolysosomes are demonstrated that the cytotoxicity of ATO may be associated with autophagy. Meanwhile, the metabolomics analysis results showed that 20 metabolites (10 increased and 10 decreased) were significantly altered with the ATO exposure, suggesting that maybe there are the perturbations in amino acid metabolism, lipid metabolism, glycan biosynthesis and metabolism, metabolism of cofactors and vitamins. We concluded that ATO was toxic to GC-1 spg cells via inducing oxidative stress, mitochondrial dysfunction and autophagy as well as the disruption of normal metabolism. This study will aid our understanding of the mechanisms behind ATO-induced spermatogenic toxicity.


Assuntos
Trióxido de Arsênio/toxicidade , Autofagia/efeitos dos fármacos , Poluentes Ambientais/toxicidade , Mitocôndrias/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Espermatogônias/efeitos dos fármacos , Animais , Antioxidantes/metabolismo , Glutationa/metabolismo , Lisossomos/metabolismo , Masculino , Metabolômica , Camundongos , Mitocôndrias/ultraestrutura , Espécies Reativas de Oxigênio/metabolismo , Espermatogônias/enzimologia , Espermatogônias/metabolismo
5.
Endocrinology ; 160(12): 2929-2945, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31621880

RESUMO

Premature overexposure to thyroid hormone causes profound effects on testis growth, spermatogenesis, and male fertility. We used genetic mouse models of type 3 deiodinase (DIO3) deficiency to determine the genetic programs affected by premature thyroid hormone action and to define the role of DIO3 in regulating thyroid hormone economy in testicular cells. Gene expression profiling in the neonatal testis of DIO3-deficient mice identified 5699 differentially expressed genes. Upregulated and downregulated genes were, respectively, involved according to DAVID analysis with cell differentiation and proliferation. They included anti-Müllerian hormone and genes involved in the formation of the blood-testis barrier, which are specific to Sertoli cells (SCs). They also included steroidogenic genes, which are specific to Leydig cells. Comparison with published data sets of genes enriched in SCs and spermatogonia, and responsive to retinoic acid (RA), identified a subset of genes that were regulated similarly by RA and thyroid hormone. This subset of genes showed an expression bias, as they were downregulated when enriched in spermatogonia and upregulated when enriched in SCs. Furthermore, using a genetic approach, we found that DIO3 is not expressed in SCs, but spermatogonia-specific inactivation of DIO3 led to impaired testis growth, reduced SC number, decreased cell proliferation and, especially during neonatal development, altered gene expression specific to somatic cells. These findings indicate that spermatogonial DIO3 protects testicular cells from untimely thyroid hormone signaling and demonstrate a mechanism of cross-talk between somatic and germ cells in the neonatal testis that involves the regulation of thyroid hormone availability and action.


Assuntos
Regulação da Expressão Gênica , Iodeto Peroxidase/deficiência , Espermatogônias/enzimologia , Testículo/enzimologia , Hormônios Tireóideos/metabolismo , Animais , Animais Recém-Nascidos , Proliferação de Células , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Espermatogênese , Testículo/crescimento & desenvolvimento
6.
Zygote ; 27(6): 432-435, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31422785

RESUMO

Long-term heat stress (HS) induced by testicular insulation generates oxidative stress (OS) on the testicular environment; consequently activating antioxidant enzymes such as superoxide dismutase (SOD), glutathione reductase (GR) and glutathione peroxidase (GPx). The aim of this work was to immunolocalize antioxidant enzymes present in different cells within the seminiferous tubule when rams were submitted to HS. Rams were divided into control (n = 6) and treated group (n = 6), comprising rams subjected to testicular insulation for 240 h. After the testicular insulation period, rams were subjected to orchiectomy. Testicular fragments were submitted to immunohistochemistry for staining against SOD, GR and GPx enzymes. We observed immunolocalization of GPx in more cell types of the testis after HS and when compared with other enzymes. In conclusion, GPx is the main antioxidant enzyme identified in testicular cells in an attempt to maintain oxidative balance when HS occurs.


Assuntos
Glutationa Peroxidase/metabolismo , Glutationa Redutase/metabolismo , Resposta ao Choque Térmico/fisiologia , Túbulos Seminíferos/enzimologia , Superóxido Dismutase/metabolismo , Testículo/enzimologia , Animais , Antioxidantes/metabolismo , Imuno-Histoquímica/métodos , Masculino , Orquiectomia , Estresse Oxidativo/fisiologia , Túbulos Seminíferos/citologia , Ovinos , Espermátides/citologia , Espermátides/enzimologia , Espermatócitos/citologia , Espermatócitos/enzimologia , Espermatogônias/citologia , Espermatogônias/enzimologia , Testículo/citologia , Fatores de Tempo
7.
Artigo em Inglês | MEDLINE | ID: mdl-30590176

RESUMO

Identification of germ cell markers is important for investigating reproduction biology in fish. Vasa is one of the most studied germ cell markers in mammals and lower vertebrates including fish. Here, we characterized a vasa homologue from the fish marbled goby (Oxyeleotris marmorata), termed omvasa. The full length of omvasa cDNA is 2344 bp and encodes 658 amino acids, sharing high identities with Vasa homologues of other vertebrates. OmVasa protein contains 15 RG/RGG repeats at N-terminus, 2 ATPase motifs, as well as RNA unwinding and RNA binding domains at C-terminus. Phylogenetic tree showed that omVasa had the closest relationship with the Vasa homologue from the fish Boleophthalmus pectinirostris, the great blue-spotted mudskipper. qRT-PCR analysis indicated that omvasa was specifically transcribed in gonads, and the transcription level was gradually increased during oocyte development. The germ cell-specific distribution of omvasa mRNA was revealed by fluorescent in situ hybridization. In ovary, the signal of omvasa RNA displayed strong-weak-strong dynamics from oogonia over pre-vitellogenic oocytes to vitellogenic oocytes. In testis, omvasa signal was strong in spermatogonia, modest in spermatocytes but undetectable in spermatids and somatic cells. During embryogenesis, the transcription of omvasa mRNA was high at blastula stage, gradually decreased from gastrula stage and maintained at a low level in later developmental stages. Whole mount in situ hybridization indicated that omvasa mRNA was specific to primordial germ cells (PGCs). In summary, marbled goby vasa is a germ cell-specific transcript during gametogenesis, and can be used as an ideal marker for tracing PGC formation and migration, which is pivotal to germ cell manipulation in this species.


Assuntos
Blástula/enzimologia , RNA Helicases DEAD-box , Desenvolvimento Embrionário/fisiologia , Proteínas de Peixes , Peixes , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Oogônios/enzimologia , Espermatogônias/enzimologia , Animais , RNA Helicases DEAD-box/biossíntese , RNA Helicases DEAD-box/genética , Feminino , Proteínas de Peixes/biossíntese , Proteínas de Peixes/genética , Peixes/embriologia , Peixes/genética , Masculino
8.
Elife ; 72018 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-29985130

RESUMO

Exhaustion of stem cells is a hallmark of aging. In the Drosophila testis, dedifferentiated germline stem cells (GSCs) derived from spermatogonia increase during lifespan, leading to the model that dedifferentiation counteracts the decline of GSCs in aged males. To test this, we blocked dedifferentiation by mis-expressing the differentiation factor bag of marbles (bam) in spermatogonia while lineage-labeling these cells. Strikingly, blocking bam-lineage dedifferentiation under normal conditions in virgin males has no impact on the GSC pool. However, in mated males or challenging conditions, inhibiting bam-lineage dedifferentiation markedly reduces the number of GSCs and their ability to proliferate and differentiate. We find that bam-lineage derived GSCs have significantly higher proliferation rates than sibling GSCs in the same testis. We determined that Jun N-terminal kinase (JNK) activity is autonomously required for bam-lineage dedifferentiation. Overall, we show that dedifferentiation provides a mechanism to maintain the germline and ensure fertility under chronically stressful conditions.


Assuntos
Desdiferenciação Celular , Drosophila melanogaster/citologia , Sistema de Sinalização das MAP Quinases , Espermatogônias/enzimologia , Espermatogônias/patologia , Células-Tronco/enzimologia , Células-Tronco/patologia , Estresse Fisiológico , Testículo/patologia , Envelhecimento/metabolismo , Animais , Linhagem da Célula , Ativação Enzimática , Masculino , Modelos Biológicos , Espermatogênese
9.
Biochem Biophys Res Commun ; 503(1): 51-55, 2018 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-29842884

RESUMO

Male germ cells are transformed from undifferentiated stem cells into spermatozoa through a series of highly regulated steps together termed spermatogenesis. Spermatogonial stem cells undergo mitosis and differentiation followed by two rounds of meiotic division and then proceed through a series of dramatic cell shape changes to form highly differentiated spermatozoa. Using indirect immunofluorescence, we investigated a role for the mitotic kinase, Aurora A (AURKA), in these events through localization of this protein in mouse testis and spermatozoa. AURKA is expressed in several cell types in the testis. Spermatogonia and spermatocytes express AURKA as expected based on the known role of this kinase in cell division. Surprisingly, we also found AURKA localized to spermatids and the flagellum of spermatozoa. Total AURKA and activated AURKA are expressed in different compartments of the sperm flagellum with total AURKA found in the principal piece and its phosphorylated and activated form found in the sperm midpiece. In addition, active AURKA is enriched in the flagellum of motile sperm isolated from cauda epididymis. These results provide evidence for a unique role for AURKA in spermatogenesis and sperm motility. Defining the signaling mechanisms that govern spermatogenesis and sperm cell function is crucial to understanding and treating male infertility as well as for development of new contraceptive strategies.


Assuntos
Aurora Quinase A/metabolismo , Espermatogênese/fisiologia , Testículo/citologia , Testículo/enzimologia , Animais , Epididimo/citologia , Epididimo/enzimologia , Técnica Indireta de Fluorescência para Anticorpo , Infertilidade Masculina/enzimologia , Masculino , Camundongos , Transdução de Sinais , Motilidade dos Espermatozoides/fisiologia , Cauda do Espermatozoide/enzimologia , Espermátides/enzimologia , Espermatócitos/enzimologia , Espermatogônias/enzimologia , Espermatozoides/enzimologia
10.
Cell Cycle ; 17(2): 225-239, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29169284

RESUMO

We have previously shown that the transcript levels of Vegfc and its receptor Vegfr3 were high in spermatogonia and extremely low in spermatocytes and spermatids. However, it remains unknown about the functions and the mechanisms of VEGFC/VEGFR3 signaling in regulating the fate determinations of spermatogonia. To this end, here we explored the role and signaling pathways of VEGFC/VEGFR3 by using a cell line derived from immortalized mouse spermatogonia retaining markers of mitotic germ cells, namely GC-1 cells. VEGFR3 was expressed in mouse primary spermatogonia and GC-1 cells. VEGFC stimulated the proliferation and DNA synthesis of GC-1 cells and enhanced the phosphorylation of PI3K-AKT and MAPK, whereas LY294002 (an inhibitor for AKT) and CI-1040 (an inhibitor for MAPK) blocked the effect of VEGFC on GC-1 cell proliferation. Furthermore, VEGFC increased the transcripts of c-fos and Egr1 and protein levels of cyclin D1, PCNA and Bcl-2. Conversely, the blocking of VEGFC/VEGFR3 signaling by VEGFR3 knockdown reduced the phosphorylation of AKT/MAPK and decreased the levels of cyclin D1 and PCNA. Additionally, VEGFR3 knockdown not only resulted in more apoptosis of GC-1 cells but also led to a decrease of Bcl-2 and promoted the cleavage of Caspase-3/9 and PARP. Collectively, these data suggested that VEGFC/VEGFR3 signaling promotes the proliferation of GC-1 cells via the AKT /MAPK and cyclin D1 pathway and it inhibits the cell apoptosis through Caspase-3/9, PARP and Bcl-2. Thus, this study sheds a novel insight to the molecular mechanisms underlying the fate decisions of mammalian spermatogonia.


Assuntos
Proliferação de Células , Transdução de Sinais , Espermatogônias/metabolismo , Fator C de Crescimento do Endotélio Vascular/fisiologia , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Apoptose , Caspase 3/metabolismo , Caspase 9/metabolismo , Linhagem Celular , Ciclina D1/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/biossíntese , Proteína 1 de Resposta de Crescimento Precoce/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Indóis/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Naftalenos/farmacologia , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-fos/biossíntese , Proteínas Proto-Oncogênicas c-fos/genética , Espermatogônias/citologia , Espermatogônias/efeitos dos fármacos , Espermatogônias/enzimologia , Fator C de Crescimento do Endotélio Vascular/genética , Fator C de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/fisiologia
11.
Reproduction ; 154(2): 135-143, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28559472

RESUMO

USP9X (ubiquitin-specific peptidase 9, X chromosome) is the mammalian orthologue of Drosophila deubiquitinase fat facets that was previously shown to regulate the maintenance of the germ cell lineage partially through stabilizing Vasa, one of the widely conserved factors crucial for gametogenesis. Here, we demonstrate that USP9X is expressed in the gonocytes and spermatogonia in mouse testes from newborn to adult stages. By using Vasa-Cre mice, germ cell-specific conditional deletion of Usp9x from the embryonic stage showed no abnormality in the developing testes by 1 week and no appreciable defects in the undifferentiated and differentiating spermatogonia at postnatal and adult stages. Interestingly, after 2 weeks, Usp9x-null spermatogenic cells underwent apoptotic cell death at the early spermatocyte stage, and then, caused subsequent aberrant spermiogenesis, which resulted in a complete infertility of Usp9x conditional knockout male mice. These data provide the first evidence of the crucial role of the spermatogonial USP9X during transition from the mitotic to meiotic phases and/or maintenance of early meiotic phase in Usp9x conditional knockout testes.


Assuntos
Endopeptidases/metabolismo , Fertilidade , Infertilidade Masculina/enzimologia , Espermatogênese , Espermatogônias/enzimologia , Testículo/enzimologia , Fatores Etários , Animais , Apoptose , Endopeptidases/deficiência , Endopeptidases/genética , Genótipo , Infertilidade Masculina/genética , Infertilidade Masculina/fisiopatologia , Masculino , Meiose , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Transdução de Sinais , Espermatogônias/patologia , Testículo/patologia , Testículo/fisiopatologia , Ubiquitina Tiolesterase
12.
Science ; 354(6314): 909-912, 2016 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-27856912

RESUMO

DNA methylation is prevalent in mammalian genomes and plays a central role in the epigenetic control of development. The mammalian DNA methylation machinery is thought to be composed of three DNA methyltransferase enzymes (DNMT1, DNMT3A, and DNMT3B) and one cofactor (DNMT3L). Here, we describe the discovery of Dnmt3C, a de novo DNA methyltransferase gene that evolved via a duplication of Dnmt3B in rodent genomes and was previously annotated as a pseudogene. We show that DNMT3C is the enzyme responsible for methylating the promoters of evolutionarily young retrotransposons in the male germ line and that this specialized activity is required for mouse fertility. DNMT3C reveals the plasticity of the mammalian DNA methylation system and expands the scope of the mechanisms involved in the epigenetic control of retrotransposons.


Assuntos
DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , Epigênese Genética , Mutagênese/genética , Regiões Promotoras Genéticas , Retroelementos , Espermatogônias/enzimologia , Animais , Linhagem Celular , DNA (Citosina-5-)-Metiltransferases/classificação , DNA (Citosina-5-)-Metiltransferases/genética , Etilnitrosoureia/farmacologia , Técnicas de Inativação de Genes , Hipogonadismo/induzido quimicamente , Hipogonadismo/genética , Hipogonadismo/patologia , Masculino , Camundongos , Filogenia , Espermatogônias/efeitos dos fármacos , Testículo/efeitos dos fármacos , Testículo/patologia
13.
Genes Dev ; 29(23): 2420-34, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26584619

RESUMO

Telomerase inactivation causes loss of the male germline in worms, fish, and mice, indicating a conserved dependence on telomere maintenance in this cell lineage. Here, using telomerase reverse transcriptase (Tert) reporter mice, we found that very high telomerase expression is a hallmark of undifferentiated spermatogonia, the mitotic population where germline stem cells reside. We exploited these high telomerase levels as a basis for purifying undifferentiated spermatogonia using fluorescence-activated cell sorting. Telomerase levels in undifferentiated spermatogonia and embryonic stem cells are comparable and much greater than in somatic progenitor compartments. Within the germline, we uncovered an unanticipated gradient of telomerase activity that also enables isolation of more mature populations. Transcriptomic comparisons of Tert(High) undifferentiated spermatogonia and Tert(Low) differentiated spermatogonia by RNA sequencing reveals marked differences in cell cycle and key molecular features of each compartment. Transplantation studies show that germline stem cell activity is confined to the Tert(High) cKit(-) population. Telomere shortening in telomerase knockout strains causes depletion of undifferentiated spermatogonia and eventual loss of all germ cells after undifferentiated spermatogonia drop below a critical threshold. These data reveal that high telomerase expression is a fundamental characteristic of germline stem cells, thus explaining the broad dependence on telomerase for germline immortality in metazoans.


Assuntos
Células-Tronco Adultas/enzimologia , Regulação Enzimológica da Expressão Gênica , Espermatogônias/enzimologia , Telomerase/genética , Telomerase/metabolismo , Animais , Diferenciação Celular/genética , Células-Tronco Embrionárias/enzimologia , Citometria de Fluxo , Técnicas de Introdução de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas/genética
14.
FASEB J ; 29(11): 4402-16, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26243864

RESUMO

Little is known of the fundamental processes governed by epigenetic mechanisms in the supplier cells of spermatogenesis, the spermatogonial stem cells (SSCs). The histone H3 lysine demethylase KDM1A is expressed in spermatogonia. We hypothesized that KDM1A serves in transcriptional regulation of SSCs and fertility. Using a conditional deletion of Kdm1a [conditional knockout (cKO)] in mouse spermatogonia, we determined that Kdm1a is essential for spermatogenesis as adult cKO males completely lack germ cells. Analysis of postnatal testis development revealed that undifferentiated and differentiating spermatogonial populations form in Kdm1a-cKO animals, yet the majority fail to enter meiosis. Loss of germ cells in the cKO was rapid with none remaining by postnatal day (PND) 21. To gain insight into the mechanistic implications of Kdm1a ablation, we isolated PND 6 spermatogonia enriched for SSCs and analyzed their transcriptome by RNA sequencing. Loss of Kdm1a was associated with altered transcription of 1206 genes. Importantly, differentially expressed genes between control and Kdm1a-cKO animals included those that are essential for SSC and progenitor maintenance and spermatogonial differentiation. The complete loss of fertility and failure to establish spermatogenesis indicate that Kdm1a is a master controller of gene transcription in spermatogonia and is required for SSC and progenitor maintenance and differentiation.


Assuntos
Histona Desmetilases/metabolismo , Espermatogênese/fisiologia , Espermatogônias/enzimologia , Células-Tronco/enzimologia , Transcrição Gênica/fisiologia , Animais , Histona Desmetilases/genética , Masculino , Camundongos , Camundongos Knockout , Espermatogônias/citologia , Células-Tronco/citologia
15.
Fertil Steril ; 104(2): 302-11.e3, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26056924

RESUMO

OBJECTIVE: To compare mechanical dissociation, employing the Medimachine system, and enzymatic digestion of human testicular tissues with respect to the proportion of spermatogonia and somatic cells, with the long-term objective of establishing human spermatogonial cultures. DESIGN: Experimental basic science study. SETTING: Reproductive biology laboratory. PATIENT(S): Testicular tissues were obtained from patients with gender dysphoria on the day of sex reassignment surgery. On the basis of the histological evaluation, tissue samples with complete spermatogenesis (fresh, n = 6; cryopreserved, n = 7) and with meiotic arrest (cryopreserved, n = 4) were selected. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): The composition of testicular cell suspensions was assessed performing quantitative real-time polymerase chain reaction (qPCR) analyses for germ cell-specific (FGFR3, SALL4, UTF1, MAGE-A4) and somatic marker genes (ACTA2 and VIM). Additionally, flow-cytometric analyses were used to evaluate the percentage of SALL4-and vimentin-positive cells. RESULT(S): While Medimachine dissociation yielded higher cell numbers in all patient groups, viability of cells was highly variable and correlated with the histological status of the tissue. Interestingly, qPCR analysis revealed a significantly decreased expression of the somatic marker genes ACTA2 and VIM and an increased expression of the spermatogonial marker genes FGFR3 and SALL4 after Medimachine dissociation. These findings were corroborated by flow-cytometric analyses that demonstrated that the proportion of SALL4-positive cells was up to 4 times higher after mechanical dissociation. CONCLUSION(S): Medimachine dissociation of human testicular tissues is comparably fast and leads to an enrichment of SALL4-positive spermatogonia. The use of this method may therefore constitute an advantage for the establishment of human spermatogonial cell cultures.


Assuntos
Ensaios Enzimáticos/métodos , Citometria de Fluxo/métodos , Testículo/enzimologia , Sobrevivência Celular/fisiologia , Humanos , Masculino , Reação em Cadeia da Polimerase em Tempo Real/métodos , Espermatogônias/enzimologia , Testículo/citologia
16.
Toxicol Lett ; 234(2): 120-30, 2015 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-25680692

RESUMO

In this study, DNA arrays have been employed to monitor gene expression patterns in testis of mice exposed to tobacco smoke for 24 weeks and compared to control animals. The results of the analysis revealed significant changes in expression of several genes that may have a role in spermatogenesis. Cdk14 was chosen for further characterization because of a suggested role in the testis and in regulation of Wnt signaling. RT-PCR analysis confirmed down regulation of Cdk14 in mice exposed to cigarette smoke (CS). Cdk14 is expressed in all testicular cells; spermatogonia- and Sertoli-derived cell lines treated with cigarette smoke extract (CSE) in vitro showed down-regulation of CDK14 mRNA and protein levels as well as down-regulation of ß-catenin levels. CS-induced down-regulation of CDK14 mRNA and protein levels was also observed in several lung epithelium-derived cell lines including primary normal human bronchial epithelial cells (NHBE), suggesting that the effect is not restricted to the testis. Similar to testicular cells, CS-induced down-regulation of CDK14 in lung cells correlated with decreased levels of ß-catenin, a finding suggesting impaired Wnt signaling. In the lungs, CDK14 was localized to the alveolar and bronchial epithelium.


Assuntos
Quinases Ciclina-Dependentes/metabolismo , Proteínas Quinases/metabolismo , Fumaça/efeitos adversos , Fumar/efeitos adversos , Testículo/efeitos dos fármacos , Animais , Quinases Ciclina-Dependentes/genética , Regulação para Baixo , Perfilação da Expressão Gênica , Humanos , Exposição por Inalação/efeitos adversos , Masculino , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Quinases/genética , Células de Sertoli/efeitos dos fármacos , Células de Sertoli/enzimologia , Fumar/genética , Fumar/metabolismo , Espermatogônias/efeitos dos fármacos , Espermatogônias/enzimologia , Testículo/enzimologia , Fatores de Tempo , Via de Sinalização Wnt/efeitos dos fármacos , beta Catenina/genética , beta Catenina/metabolismo
17.
Cell Death Dis ; 6: e1610, 2015 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-25611385

RESUMO

G-quadruplex (G4) DNA and G4 DNA resolvase are involved in a variety of biological processes. To understand the biological function of G4 DNA structures and their resolvases in spermatogenesis, we investigated the distribution of G4 structures in mouse testis and identified their alterations during spermatogenesis. Meanwhile, we studied the function of RNA helicase associated with AU-rich element (RHAU), a G4 DNA resolvase, in spermatogenesis with a germ-cell-specific knockout mouse model. The results showed that the ablation of RHAU in germ cells caused the increase of G4 structures and thus resulted in the decrease of spermatogonial differentiation. c-kit, a spermatogonia differentiation-related gene, contains two G4 DNA motifs on its promoter. We found its expression was significantly downregulated in RHAU conditional knockout testis. A further analysis demonstrated that RHAU directly bound to the G4 structures to activate c-kit expression. We concluded that RHAU regulates spermatogonia differentiation by promoting c-kit expression via directly binding to the G4 DNA motifs c-kit promoter.


Assuntos
Diferenciação Celular , RNA Helicases DEAD-box/metabolismo , DNA/química , Quadruplex G , Recombinases/metabolismo , Espermatogônias/citologia , Espermatogônias/enzimologia , Trifosfato de Adenosina/metabolismo , Animais , Apoptose , Azoospermia/enzimologia , Azoospermia/patologia , Sequência de Bases , Proliferação de Células , RNA Helicases DEAD-box/deficiência , DNA/metabolismo , Deleção de Genes , Regulação da Expressão Gênica , Masculino , Meiose , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Motivos de Nucleotídeos/genética , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Testículo/citologia , Testículo/enzimologia
18.
Stem Cells ; 32(3): 741-53, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24123360

RESUMO

SHP2 is a widely expressed protein tyrosine phosphatase required for signal transduction from multiple cell surface receptors. Gain and loss of function SHP2 mutations in humans are known to cause Noonan and LEOPARD syndromes, respectively, that are characterized by numerous pathological conditions including male infertility. Using conditional gene targeting in the mouse, we found that SHP2 is required for maintaining spermatogonial stem cells (SSCs) and the production of germ cells required for male fertility. After deleting SHP2, spermatogenesis was halted at the initial step during which transit-amplifying undifferentiated spermatogonia are produced from SSCs. In the absence of SHP2, proliferation of SSCs and undifferentiated spermatogonia was inhibited, thus germ cells cannot be replenished and SSCs cannot undergo renewal. However, germ cells beyond the undifferentiated spermatogonia stage of development at the time of SHP2 knockout were able to complete their maturation to become sperm. In cultures of SSCs and their progeny, inhibition of SHP2 activity reduced growth factor-mediated intracellular signaling that regulates SSC proliferation and cell fate. Inhibition of SHP2 also decreased the number of SSCs present in culture and caused SSCs to detach from supporting cells. Injection of mice with an SHP2 inhibitor blocked the production of germ cells from SSCs. Together, our studies show that SHP2 is essential for SSCs to maintain fertility and indicates that the pathogenesis of infertility in humans with SHP2 mutations is due to compromised SSC functions that block spermatogenesis.


Assuntos
Fertilidade , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Espermatogônias/citologia , Espermatogônias/enzimologia , Células-Tronco/citologia , Células-Tronco/enzimologia , Envelhecimento , Animais , Adesão Celular , Contagem de Células , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Deleção de Genes , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Masculino , Camundongos , Camundongos Knockout , Proteína Tirosina Fosfatase não Receptora Tipo 11/antagonistas & inibidores , Transdução de Sinais , Espermatogênese
19.
Biochem Biophys Res Commun ; 441(1): 120-5, 2013 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-24129193

RESUMO

The function of protease during male meiosis has not been well studied. We previously cloned and characterized four testis-specific serine proteases in the mouse testis. One of the proteases, Prss41/Tessp-1, was expressed in the germ and Sertoli cell. This time, to examine the involvement of Prss41/Tessp-1 in spermatogenesis, we conducted the organ culture of testis fragments in the presence of the anti-Prss41/Tessp-1 antibody. Because in the Sertoli cell, the Prss41/Tessp-1 protein was mostly associated with the membrane of intracellular organelles by glycosylphosphatidylinositol, the antibody was expected to affect Prss41/Tessp-1 at the plasma membrane of spermatogonia. By adding the antibody, the number of germ cells was decreased in some seminiferous tubules. The marker genes expression strongly suggested that meiosis was arrested at spermatogonia, and the number of apoptotic germ cells increased by terminal deoxynucleotidyl transferase dUTP nick end labeling assay. These data indicated that Prss41/Tessp-1 was necessary for the progression of meiosis at the stage of spermatogonia during in vitro spermatogenesis. Together with our previous study, the current results suggest that the Prss/Tessp proteases are important for the progression of meiosis at each stage.


Assuntos
Meiose , Serina Endopeptidases/metabolismo , Espermatogênese , Testículo/citologia , Testículo/enzimologia , Animais , Anticorpos/metabolismo , Apoptose , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Técnicas de Cultura de Órgãos , Especificidade de Órgãos , Transporte Proteico , Células de Sertoli/citologia , Células de Sertoli/enzimologia , Espermatogônias/citologia , Espermatogônias/enzimologia , Frações Subcelulares/enzimologia
20.
Stem Cells ; 31(11): 2517-27, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23897718

RESUMO

Coordination of stem cell fate is regulated by extrinsic niche signals and stem cell intrinsic factors. In mammalian testes, spermatogonial stem cells maintain constant production of abundant spermatozoa by alternating between self-renewal and differentiation at regular intervals according to a periodical program known as the seminiferous epithelial cycle. Although retinoic acid (RA) signaling has been suggested to direct the cyclical differentiation of spermatogonial stem cells, it remains largely unclear how their cycle-dependent self-renewal/proliferation is regulated. Here, we show that MEK/ERK signaling contributes to the cyclical activity of spermatogonial stem cells. We found that ERK1/2 is periodically activated in Sertoli cells during the stem cell self-renewal/proliferation phase, and that MEK/ERK signaling is required for the stage-related expression of the critical niche factor GDNF. In addition, ERK1/2 is activated in GFRα1-positive spermatogonial stem cells under the control of GDNF and prevent them from being differentiated. These results suggest that MEK/ERK signaling directly and indirectly maintains spermatogonial stem cells by mediating a signal that promotes their periodical self-renewal/proliferation. Conversely, RA signaling directly and indirectly induces differentiation of spermatogonial stem cells. We propose that temporally regulated activations of RA signaling and a signal regulating MEK/ERK antagonistically coordinates the cycle-related activity of spermatogonial stem cells.


Assuntos
Sistema de Sinalização das MAP Quinases/fisiologia , Espermatogônias/citologia , Espermatogônias/enzimologia , Células-Tronco/citologia , Células-Tronco/enzimologia , Animais , Diferenciação Celular/fisiologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células de Sertoli/citologia , Células de Sertoli/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...