Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 161
Filtrar
1.
Hum Reprod ; 28(6): 1635-46, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23482336

RESUMO

STUDY QUESTION: What human tissues and cell types express the X-linked reproductive homeobox (RHOX) gene cluster? SUMMARY ANSWER: The RHOX homeobox genes and proteins are selectively expressed in germ cells in both the ovary and testis. WHAT IS KNOWN ALREADY: The RHOX homeobox transcription factors are encoded by an X-linked gene cluster whose members are selectively expressed in the male and female reproductive tract of mice and rats. The Rhox genes have undergone strong selection pressure to rapidly evolve, making it uncertain whether they maintain their reproductive tissue-centric expression pattern in humans, an issue we address in this report. STUDY DESIGN, SIZE, DURATION: We examined the expression of all members of the human RHOX gene cluster in 11 fetal and 8 adult tissues. The focus of our analysis was on fetal testes, where we evaluated 16 different samples from 8 to 20 weeks gestation. We also analyzed fixed sections from fetal testes, adult testes and adult ovaries to determine the cell type-specific expression pattern of the proteins encoded by RHOX genes. PARTICIPANTS/MATERIALS, SETTING, METHODS: We used quantitative reverse transcription-polymerase chain reaction analysis to assay human RHOX gene expression. We generated antisera against RHOX proteins and used them for western blotting, immunohistochemical and immunofluorescence analyses of RHOXF1 and RHOXF2/2B protein expression. MAIN RESULTS AND THE ROLE OF CHANCE: We found that the RHOXF1 and RHOXF2/2B genes are highly expressed in the testis and exhibit low or undetectable expression in most other organs. Using RHOXF1- and RHOXF2/2B-specific antiserum, we found that both RHOXF1 and RHOXF2/2B are primarily expressed in germ cells in the adult testis. Early stage germ cells (spermatogonia and early spermatocytes) express RHOXF2/2B, while later stage germ cells (pachytene spermatocytes and round spermatids) express RHOXF1. Both RHOXF1 and RHOXF2/2B are expressed in prespermatogonia in human fetal testes. Consistent with this, RHOXF1 and RHOXF2/2B mRNA expression increases in the second trimester during fetal testes development when gonocytes differentiate into prespermatogonia. In the human adult ovary, we found that RHOXF1 and RHOXF2/2B are primarily expressed in oocytes. LIMITATIONS, REASONS FOR CAUTION: While the average level of expression of RHOX genes was low or undetectable in all 19 human tissues other than testes, it is still possible that RHOX genes are highly expressed in a small subset of cells in some of these non-testicular tissues. As a case in point, we found that RHOX proteins are highly expressed in oocytes within the human ovary, despite low levels of RHOX mRNA in the whole ovary. WIDER IMPLICATIONS OF THE FINDINGS: The cell type-specific and developmentally regulated expression pattern of the RHOX transcription factors suggests that they perform regulatory functions during human fetal germ cell development, spermatogenesis and oogenesis. Our results also raise the possibility that modulation of RHOX gene levels could correct some cases of human infertility and that their encoded proteins are candidate targets for contraceptive drug design.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Oócitos/metabolismo , Espermatozoides/metabolismo , Adulto , Sequência de Aminoácidos , Western Blotting , Feminino , Proteínas de Homeodomínio/metabolismo , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Família Multigênica , Placenta/metabolismo , Gravidez , Primeiro Trimestre da Gravidez , Segundo Trimestre da Gravidez , Testículo/metabolismo
2.
Hum Reprod Update ; 17(5): 667-83, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21546379

RESUMO

BACKGROUND: The increase in the incidence of obesity has a substantial societal health impact. Contrasting reports have been published on whether overweight and obesity affect male fertility. To clarify this, we have reviewed published data on the relation between overweight/obesity, semen parameters, endocrine status and human male fertility. Subsequently, we have used results obtained in animal models of obesity to explain the human data. METHODS: Pubmed, Scopus, Web of Science and Google Scholar databases were searched between September 2009 and October 2010 for a comprehensive publication record. Available studies on adult human males were examined. The included animal studies examined obesity and fertility, and focused on leptin, leptin receptor signaling, kisspeptins and/or NPY. RESULTS: Most overweight/obese men do not experience significant fertility problems, despite the presence of reduced testosterone alongside normal gonadotrophin levels. Only a subgroup of subjects suffers from hypogonadotropic hypogonadism. Animal models offer several explanations and show that reduced leptin signaling leads to reduced GnRH neuronal activity. This may be due to decreased hypothalamic Kiss1 expression, a potent regulator of GnRH/LH/FSH release. As the Kiss1 neurons express leptin receptors, the Kiss1 system may participate in transmitting metabolic information to the GnRH neurons, thus providing a bridge between metabolic regulation and fertility. CONCLUSIONS: Infertility in overweight/obese males may be explained by leptin insensitivity. This implies a possible role for the KISS1 system in human obesity-related male infertility. If substantiated, it will pave the way for methods to restore fertility in these subjects.


Assuntos
Infertilidade Masculina/complicações , Obesidade/complicações , Adulto , Animais , Modelos Animais de Doenças , Humanos , Masculino , Obesidade/metabolismo , Análise do Sêmen , Transdução de Sinais
3.
Hum Reprod ; 25(1): 158-67, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19815622

RESUMO

BACKGROUND: Given the significant drawbacks of using human embryonic stem (hES) cells for regenerative medicine, the search for alternative sources of multipotent cells is ongoing. Studies in mice have shown that multipotent ES-like cells can be derived from neonatal and adult testis. Here we report the derivation of ES-like cells from adult human testis. METHODS: Testis material was donated for research by four men undergoing bilateral castration as part of prostate cancer treatment. Testicular cells were cultured using StemPro medium. Colonies that appeared sharp edged and compact were collected and subcultured under hES-specific conditions. Molecular characterization of these colonies was performed using RT-PCR and immunohistochemistry. (Epi)genetic stability was tested using bisulphite sequencing and karyotype analysis. Directed differentiation protocols in vitro were performed to investigate the potency of these cells and the cells were injected into immunocompromised mice to investigate their tumorigenicity. RESULTS: In testicular cell cultures from all four men, sharp-edged and compact colonies appeared between 3 and 8 weeks. Subcultured cells from these colonies showed alkaline phosphatase activity and expressed hES cell-specific genes (Pou5f1, Sox2, Cripto1, Dnmt3b), proteins and carbohydrate antigens (POU5F1, NANOG, SOX2 and TRA-1-60, TRA-1-81, SSEA4). These ES-like cells were able to differentiate in vitro into derivatives of all three germ layers including neural, epithelial, osteogenic, myogenic, adipocyte and pancreatic lineages. The pancreatic beta cells were able to produce insulin in response to glucose and osteogenic-differentiated cells showed deposition of phosphate and calcium, demonstrating their functional capacity. Although we observed small areas with differentiated cell types of human origin, we never observed extensive teratomas upon injection of testis-derived ES-like cells into immunocompromised mice. CONCLUSIONS: Multipotent cells can be established from adult human testis. Their easy accessibility and ethical acceptability as well as their non-tumorigenic and autogenic nature make these cells an attractive alternative to human ES cells for future stem cell therapies.


Assuntos
Técnicas de Cultura de Células , Células-Tronco Embrionárias/citologia , Células-Tronco Multipotentes/citologia , Testículo/citologia , Idoso , Idoso de 80 Anos ou mais , Animais , Diferenciação Celular , Proliferação de Células , Células-Tronco Embrionárias/metabolismo , Humanos , Cariotipagem , Masculino , Camundongos , Pessoa de Meia-Idade , Células-Tronco Multipotentes/metabolismo
4.
Reproduction ; 133(4): 743-51, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17504918

RESUMO

Phosphoprotein enriched in astrocytes (PEA-15) is a 15 kDa acidic serine-phosphorylated protein expressed in different cell types, especially in the CN. We initially detected the expression of PEA-15 in primary cultures of Sertoli cells. To assess the presence and localization of PEA-15 in the mouse testis, we studied the expression pattern of the PEA-15 protein by immunohistochemistry and mRNA by in situ hybridization. Both the protein and the mRNA of PEA-15 were localized in the cytoplasm of Sertoli cells, all types of spermatogonia, and spermatocytes up till zygotene phase of the meiotic prophase. Subsequently, with ongoing development of the spermatocytes, the expression decreased and was very low in the cytoplasm of diplotene spermatocytes. To analyze the possible role of PEA-15 in the developing testis, null mutants for PEA-15 were examined. As the PEA-15 C terminus contains residues for ERK binding, we studied possible differences between the localization of the ERK2 protein in wild type (WT) and PEA-15(-/-)mice. In the WT testis, ERK2 was localized in the cytoplasm of Sertoli cells, B spermatogonia, preleptotene, leptotene, and zygotene spermatocytes, whereas in the KO testis, ERK2 was primarily localized in the nuclei of these cells and only little staining remained in the cytoplasm. Moreover, in PEA-15-deficient mice, significantly increased numbers of apoptotic spermatocytes were found, indicating an anti-apoptotic role of PEA-15 during the meiotic prophase. The increased numbers of apoptotic spermatocytes were not found at a specific step in the meiotic prophase.


Assuntos
Fosfoproteínas/fisiologia , Espermatócitos/citologia , Testículo/metabolismo , Animais , Apoptose , Proteínas Reguladoras de Apoptose , Western Blotting/métodos , Linhagem Celular , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno , Proteína Quinase 3 Ativada por Mitógeno , Fosfoproteínas/análise , Fosfoproteínas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células de Sertoli/química , Células de Sertoli/metabolismo , Espermatócitos/química , Testículo/química
5.
Mol Hum Reprod ; 13(3): 155-63, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17208930

RESUMO

Testis expressed gene 18 (Tex18) is a small gene with one exon of 240 bp, which is specifically expressed in male germ cells. The gene encodes for a protein of 80 amino acids with unknown domain. To investigate the function of (Tex18) gene, we generated mice with targeted disruption of the (Tex18) gene by homologous recombination. Homozygous mutant males on a mixed genetic background (C57BL/6J x 129/Sv) are fertile, while they are subfertile on the 129/Sv background, although mating is normal. We showed that Tex18(-/-) males are subfertile because of abnormal sperm morphology and reduced motility, which is called asthenoteratozoospermia, suggesting that (Tex18) affects sperm characteristics. Maturation of spermatids is unsynchronized and partially impaired in seminiferous tubules of Tex18(-/-) mice. Electron microscopical examination demonstrated abnormal structures of sperm head. In vivo experiments with sperm of Tex18(-/-) 129/Sv mice revealed that the migration of spermatozoa from the uterus into the oviduct is reduced. This result is supported by the observation that sperm motility, as determined by the computer-assisted semen analysis system, is significantly affected, compared to wild-type spermatozoa. Generation of transgenic mice containing Tex18-EGFP fusion construct revealed a high transcriptional activity of (Tex18) during spermiogenesis, a process with morphological changes of haploid germ cells and development to mature spermatozoa. These results indicate that (Tex18) is expressed predominantly during spermatid differentiation and subfertility of the male Tex18(-/-) mice on the 129/Sv background is due to the differentiation arrest, abnormal sperm morphology and reduced sperm motility.


Assuntos
Astenozoospermia/metabolismo , Espermatozoides/ultraestrutura , Testículo/metabolismo , Reação Acrossômica , Animais , Astenozoospermia/genética , Astenozoospermia/patologia , Astenozoospermia/fisiopatologia , Diferenciação Celular , Movimento Celular , Modelos Animais de Doenças , Feminino , Proteínas de Fluorescência Verde , Tamanho da Ninhada de Vivíparos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas Recombinantes de Fusão/metabolismo , Cabeça do Espermatozoide/ultraestrutura , Espermatogênese , Testículo/patologia , Transcrição Gênica
6.
Mol Reprod Dev ; 73(11): 1361-6, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16894550

RESUMO

Phthalate esters are considered endocrine disruptors that interfere with the endocrine balance and development of the mammalian testis. Mono-2-ethylhexyl phthalate (MEHP), the active metabolite of the ubiquitously used plasticizer di-2-ethylhexyl phthalate (DEHP), acts upon Sertoli cells as initial target. By subtractive cDNA libraries we identified genes deregulated as response to MEHP in primary cultures of mouse Sertoli cells. The expression of mouse stress inducible protein 1 (Stip1) was detected as upregulated as a result of MEHP exposure. Stip1 is a cochaperone protein that is homologous to the human heat shock cognate protein 70 (hsc70)/heat shock protein 90 (hsp90)-organizing protein (Hop). To assess the presence and localization of Stip1 in mouse testis and its potential role in stress defense, we studied the expression pattern of the Stip1 protein by immunohistochemistry and of the mRNA by in situ hybridization. Both the protein and the mRNA of Stip1 were mainly found in the cytoplasm of all types of spermatogonia and spermatocytes up till zygotene, the expression decreased during late pachytene and was very weak in diplotene spermatocytes and round spermatids. Interestingly, this expression pattern resembled the pattern of stress sensitivity of spermatogenic cells in that the most sensitive cell types show the weakest expression of Stip1. This suggests an important role for Stip1 in the ability of germ cells to survive in stress conditions including high temperatures.


Assuntos
Proteínas de Choque Térmico/metabolismo , Testículo/metabolismo , Animais , Citoplasma/química , Citoplasma/metabolismo , Proteínas de Choque Térmico/análise , Proteínas de Choque Térmico/genética , Masculino , Camundongos , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Células de Sertoli/química , Células de Sertoli/metabolismo , Espermatócitos/química , Espermatócitos/metabolismo , Espermatogônias/química , Espermatogônias/metabolismo , Testículo/química , Testículo/citologia
7.
Cytogenet Genome Res ; 107(3-4): 256-62, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15467370

RESUMO

ATM, the protein product of the gene mutated in the human autosomal recessive disorder ataxia telangiectasia, is involved in detection of double strand breaks (DSBs) and is a key component of the damage surveillance network of cell cycle proteins. In somatic cells ATM phosphorylates many other proteins including p53, an important regulator of cell cycle control. Mice deficient for Atm are male sterile with arrest and apoptosis occurring at testis epithelial stage IV, which in normal spermatocytes corresponds to mid-pachynema. Unlike the situation in somatic cells, we find no evidence that disruption of the Trp53 (p53) gene, or its down-stream target Cdkn1a (p21/Cip1) results in even a partial rescue of the Atm defect.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas de Ligação a DNA/genética , Meiose , Mutação/genética , Proteínas Serina-Treonina Quinases/genética , Espermatócitos/citologia , Espermatócitos/metabolismo , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/genética , Animais , Apoptose , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/metabolismo , Inibidor de Quinase Dependente de Ciclina p21 , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/metabolismo , Feminino , Masculino , Camundongos , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/metabolismo , Rad51 Recombinase , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/metabolismo
8.
Cytogenet Genome Res ; 105(1): 36-46, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15218256

RESUMO

We have developed a protocol for the identification of aberrant chromosome behavior during human male meiosis up to metaphase of the secondary spermatocyte. Histological evaluation by the Johnsen score of a testicular biopsy was combined with immunofluorescence of first meiotic prophase spermatocytes, using antibodies against synaptonemal complex protein 3 (SYCP3) and the product of the ataxia telangiectasia and rad3-related gene (ATR). This combination enables accurate meiotic prophase substaging and the identification of pachytene spermatocytes with asynapsis. Furthermore, we also investigated the competence of late pachytene primary spermatocytes to complete the first meiotic division up to metaphase and of secondary spermatocytes to transform into metaphase by an in vitro challenge with okadaic acid (OA). We tested this protocol on five males with normal Johnsen scores that presented with obstructive azoospermia, five males with low Johnsen scores and non-obstructive azoospermia and six vasectomized control males of proven fertility and normal Johnsen scores. In all azoospermics, the profiling of meiotic prophase stages by immunofluorescence increases the resolving power of the Johnsen score. In both obstructive and non-obstructive azoospermic patients, relatively more leptotene meiotic prophase stages were counted compared to the controls. In non-obstructive azoospermics, a marked heterogeneity in spermatogenesis was found, after combining the results of all three approaches, pointing at functional mosaicism of the germinal epithelium. Asynaptic pachytene spermatocytes were rarely encountered. Also, when first meiotic metaphase could be induced by OA, chiasma counts were normal. In none of the non-obstructive azoospermic males did the pattern of spermatogenesis resemble that of knock-out mouse azoospermics. We conclude that this combined histological and cytological approach enables a detailed phenotypic classification of infertile males, at a level comparable to that applied for male-sterile knock-out mice with a meiotic defect. This may facilitate the identification of candidate genes for human male infertility.


Assuntos
Meiose , Oligospermia/fisiopatologia , Animais , Biópsia , Pareamento Cromossômico , Protocolos Clínicos , Fertilidade , Imunofluorescência , Humanos , Masculino , Camundongos , Camundongos Knockout , Prófase , Cromossomos Sexuais , Espermatócitos/citologia , Espermatogênese
9.
Cytogenet Genome Res ; 103(3-4): 267-76, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-15051947

RESUMO

In naturally occurring mutant mice but also in mice genetically modified for the study of other organs, relatively often a spermatogenic arrest is seen. In a number of cases the arrests appear to be very specific causing apoptosis of germ cells at a particular step in their development, while before this step cells progress normally. These steps include: proliferation/migration of primordial germ cells, the production of differentiating spermatogonia by gonocytes, the regulation of stem cell renewal/differentiation, the differentiation of A(al) into A1 spermatogonia, proliferation of A1-A4 spermatogonia, germ cell density regulation, start of meiosis, epithelial stage IV checkpoint of pachytene spermatocytes, the first meiotic division, the formation of the acrosomic vesicle in spermatids and several other steps in spermatid development. In addition, there are many mice that have not been studied in enough detail for a proper categorization. In this review an overview is given of the various mutations and genetically modified mice showing a direct effect on specific spermatogenic cell types. In addition, the relevance of these models to our understanding of the spermatogenic process is discussed.


Assuntos
Espermatogênese , Espermatozoides/citologia , Animais , Linhagem da Célula , Masculino , Meiose , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Prófase , Ratos , Espermátides/citologia , Espermatogônias/citologia , Espermatozoides/crescimento & desenvolvimento
10.
Reproduction ; 126(6): 765-74, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14748695

RESUMO

The aim of this study was to develop a method for spermatogonial stem cell transplantation into the bovine testis. Five-month-old Holstein-Friesian calves were used and half of the calves were hemicastrated to allow autologous transplantation and the other half were used for homologous transplantation. Approximately 20 g of each testis was used for cell isolation. On average 106 cells per gram of testis containing about 70% type A spermatogonia were isolated. The cells were frozen in liquid nitrogen until transplantation. Testes were irradiated locally with 10-14 Gy of X-rays to deplete endogenous spermatogenesis. At 2 months after irradiation, cells (approximately 10 x 10(6) were injected into the rete testis through a long injection needle (18 gauge), using ultrasonography and an ultrasound contrast solution. At 2.5 months after transplantation, calves were castrated and samples of testes were taken for histological examination. After 2.5 months in the irradiated non-transplanted control testes, only 45% of the tubules contained type A spermatogonia. However, after autologous spermatogonial transplantation, >80% of the tubule cross-sections contained type A spermatogonia. In addition, only 20% of the tubules of the control testes contained spermatocytes and, except for a few tubules (5%) with round spermatids, no more advanced germ cells were found. After autologous spermatogonial transplantation, about 60% of the tubules contained spermatocytes; 30% contained spermatids and in about 15% of tubules spermatozoa were found. No improvement in spermatogonial repopulation was found after homologous transplantation. The results of this study demonstrate, for the first time, successful autologous transplantation of bovine spermatogonial stem cells resulting in a complete regeneration of spermatogenesis.


Assuntos
Bovinos , Espermatogênese , Espermatogônias/transplante , Testículo/cirurgia , Animais , Masculino , Orquiectomia , Túbulos Seminíferos , Espermatogênese/efeitos da radiação , Testículo/patologia , Testículo/efeitos da radiação , Coleta de Tecidos e Órgãos/métodos , Transplante Autólogo , Transplante Homólogo
11.
Reproduction ; 124(1): 85-94, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12090922

RESUMO

The aim of this study was to isolate and purify bovine type A spermatogonia. Testes from 5-7-month-old calves were used to isolate germ cells using a two-step enzymatic digestion. During the isolation and purification steps, the viability of cells was determined using live/dead staining. The identity of type A spermatogonia during isolation and purification was determined under a light microscope equipped with a Nomarski lens. Isolated cells were characterized further by using specific markers for type A spermatogonia, including Dolichos biflorus agglutinin (DBA) and c-kit. The cell suspension was transplanted into immunodeficient recipient mouse testes and the colonization was assessed 1-3 months after transplantation, to assess the stem cell population among the isolated cells. After isolation, a cell suspension was obtained containing about 25% type A spermatogonia, which was enriched further by differential plating and separation on a discontinuous Percoll gradient. Finally, fractions containing 65-87% pure type A spermatogonia were obtained. Large and small type A spermatogonia with different numbers and sizes of nucleoli were found. DBA stained both large and small type A spermatogonia and its application in fluorescence-activated cell sorting (FACS) resulted in comparable percentages of type A spermatogonia to those determined by morphological examination under a light microscope equipped with a Nomarski lens. Nearly all of the large type A spermatogonia showed strong c-kit immunoreactivity, indicating that these cells had undergone at least an initial differentiation step. In contrast, approximately half of the small type A spermatogonia were negative for c-kit, indicating the presence of the spermatogonial stem cells in this population. At 3 months after transplantation, groups of bovine type A spermatogonia were found in most tubule cross-sections of the recipient mouse testes, showing the presence of spermatogonial stem cells among the isolated cells.


Assuntos
Separação Celular/métodos , Lectinas de Plantas , Espermatogônias , Animais , Bovinos , Sobrevivência Celular , Citometria de Fluxo , Imuno-Histoquímica/métodos , Lectinas , Masculino , Proteínas Proto-Oncogênicas c-kit/análise , Espermatogônias/citologia , Espermatogônias/transplante , Testículo
12.
Biol Reprod ; 66(6): 1579-84, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12021034

RESUMO

With a novel method of eliminating spermatogenesis in host animals, male germ cells isolated from mice with targeted overexpression of glial cell line-derived neurotrophic factor (GDNF) were transplanted to evaluate their ability to reproduce the phenotype previously found in the transgenic animals. Successful depletion of endogenous spermatogenesis was achieved using fractionated ionizing irradiation. A dose of 1.5 Gy followed by a dose of 12 Gy after 24 h reduced the percentage of tubule cross-sections displaying endogenous spermatogenesis to approximately 3% and 10% as evidenced by histologic evaluation of testes at 12 and 21 wk, respectively, after irradiation. At this dose, no apparent harmful side effects were noted in the animals. Upon transplantation, GDNF-overexpressing germ cells were found to be able to repopulate the irradiated testes and to form clusters of spermatogonia-like cells resembling those found in the overexpressing donor mice. The cluster cells in transplanted host testes expressed human GDNF, as had been shown previously for clusters in donor animals, and both were strongly positive for the tyrosine kinase receptor Ret. Thus, we devised an efficient method for depleting the seminiferous epithelium of host mice without appreciable adverse effects. In these host mice, GDNF-overexpressing cells reproduced the aberrant phenotype found in the donor transgenic mice.


Assuntos
Proteínas de Drosophila , Expressão Gênica , Fatores de Crescimento Neural/genética , Espermatogênese/efeitos da radiação , Espermatozoides/transplante , Testículo/citologia , Animais , Feminino , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial , Humanos , Hibridização In Situ , Masculino , Camundongos , Camundongos Nus , Camundongos Transgênicos , Fenótipo , Proteínas Proto-Oncogênicas/análise , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-ret , Receptores Proteína Tirosina Quinases/análise , Receptores Proteína Tirosina Quinases/genética , Proteínas Recombinantes , Epitélio Seminífero/fisiologia , Epitélio Seminífero/efeitos da radiação , Espermatozoides/metabolismo , Testículo/efeitos da radiação
13.
Reproduction ; 124(6): 791-9, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12530917

RESUMO

The culture of spermatogonial cells under well-defined conditions would be an important method for elucidating the mechanisms involved in spermatogenesis and in establishing tissue regeneration in vivo. In this study, a serum-free culture system was established, with type A spermatogonia isolated from adult vitamin A-deficient mice. At days 1, 3 and 7 of culture, the viability and proliferation of cells were monitored. The viability of the cells decreased by day 7 to 10% of the cells present. Proliferation occurred mainly during day 1, when 1% of the germ cells was proliferating. Co-labelling for a germ cell marker (heat shock protein-90alpha, Hsp90alpha), and a marker used to detect dividing cells (bromodeoxyuridine, BrdU), showed that this proliferation was restricted to germ cells. In an attempt to improve these parameters, medium containing fetal calf serum (FCS) was used. Viability was not influenced by serum, but proliferation was markedly enhanced. However, after day 7 of incubation with FCS, co-immunolocalization for Hsp90alpha and BrdU showed a preferential proliferation of somatic cells. Comparison of cultures of adult cells with cultures of prepubertal germ cells, commonly used in studies of spermatogenesis, showed that prepubertal germ cells are twice as viable. In addition, a different proliferation profile was observed, with a peak at day 3. Here, a distinct proliferation of somatic cells was also noted. The results from the present study indicate that the origin of isolated germ cells partly determines culture outcome and that cultures of prepubertal germ cells may not be representative for adult spermatogenesis. Moreover, adding FCS to the culture medium invokes the risk of profound and undesirable effects on cell composition, also underlining the need for identification of germ cells during culture.


Assuntos
Técnicas de Cultura de Células/métodos , Maturidade Sexual , Espermatogônias , Animais , Bromodesoxiuridina/análise , Divisão Celular , Sobrevivência Celular , Meios de Cultura Livres de Soro , Substâncias de Crescimento , Proteínas de Choque Térmico HSP90/análise , Imuno-Histoquímica/métodos , Masculino , Camundongos , Camundongos Endogâmicos , Fatores de Tempo , Deficiência de Vitamina A
14.
Mol Cell Endocrinol ; 184(1-2): 33-9, 2001 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-11694339

RESUMO

Two related ligands, glial cell line-derived neurotrophic factor (GDNF) and neurturin (NRTN), are expressed by Sertoli cells, but their cognate ligand-binding co-receptors, GDNF family receptor alpha1 and alpha2, are displayed by different germ cells suggesting different targets for the ligands. GDNF regulates cell fate decision of undifferentiated spermatogonia 'Science 287 (2000) 1489'. The role of NRTN was now approached by targeted overexpression in mouse testis. Between 3 and 5 weeks of age, transient degeneration of spermatogenic cells was observed in approximately 20% of all five transgenic lines generated. Spermatids and pachytene spermatocytes underwent segmental degeneration, if the rete testis was undilated. When it was dilated, the spermatids and spermatocytes were more generally depleted. After 5 weeks of age, spermatogenic defects were no more observed and the NRTN overexpressing mice were fertile. The data suggest that NRTN might regulate survival and differentiation of spermatocytes and spermatids, but the low penetrance indicates that either the transgene expression has not been high enough or NRTN is not as essential as GDNF for spermatogenesis.


Assuntos
Proteínas de Drosophila , Fatores de Crescimento Neural/farmacologia , Espermatogênese/efeitos dos fármacos , Testículo/efeitos dos fármacos , Animais , Regulação da Expressão Gênica , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Neurturina , Fenótipo , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-ret , RNA Mensageiro/metabolismo , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Espermatogênese/genética , Espermatogônias/citologia , Espermatogônias/efeitos dos fármacos , Testículo/citologia , Testículo/metabolismo
15.
Biol Reprod ; 65(3): 771-6, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11514340

RESUMO

Dazl encodes an RNA-binding protein essential for spermatogenesis. Mice that are deficient for Dazl are infertile, lacking any formation of spermatozoa, and the only germ cells present are spermatogonia and a few spermatocytes. To gain more insight regarding the timing of the spermatogenic arrest in Dazl -/- mice, we studied the spermatogonial cell types present in testis sections and in seminiferous tubular whole mounts. Most of the seminiferous tubular cross-sections contained A spermatogonia as the most advanced cell type, with only very few containing cells up to pachytene spermatocytes. Both 5-bromodeoxy-uridine incorporation and mitotic index indicated that the remaining A spermatogonia were actively proliferating. C-kit immunohistochemical studies showed that most of the A spermatogonia were positively stained for the c-Kit protein ( approximately 80%). The clonal composition of the A spermatogonia in tubular whole mounts indicated these cells to be A(single) (A(s)), A(paired) (A(pr)), and A(aligned) (A(al)) spermatogonia. It is concluded that the prime spermatogenic defect in the Dazl -/- mice is a failure of the great majority of the A(al) spermatogonia to differentiate into A(1) spermatogonia. As a result, most seminiferous tubules of Dazl -/- mice only contain actively proliferating A(s), A(pr), and A(al) spermatogonia, with cell production being equaled by apoptosis of these cells.


Assuntos
Infertilidade Masculina/genética , Proteínas/genética , Proteínas de Ligação a RNA , Espermatogênese/genética , Animais , Apoptose , Imuno-Histoquímica , Infertilidade Masculina/patologia , Masculino , Camundongos , Camundongos Knockout , Índice Mitótico , Proteínas/fisiologia , Proteínas Proto-Oncogênicas c-kit/análise , Túbulos Seminíferos/patologia , Células de Sertoli/patologia , Espermatócitos/patologia , Espermatogônias/patologia , Testículo/patologia
16.
Oncogene ; 20(32): 4298-304, 2001 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-11466610

RESUMO

p53 plays a central role in the induction of apoptosis of spermatogonia in response to ionizing radiation. In p53(-/-) testes, however, spermatogonial apoptosis still can be induced by ionizing radiation, so p53 independent apoptotic pathways must exist in spermatogonia. Here we show that the p53 homologues p63 and p73 are present in the testis and that p73, but not p63, is localized in the cytoplasm of spermatogonia. Unlike p53, neither p63 nor p73 protein levels were found to increase after a dose of 4 Gy of X-rays. Although p73 protein levels did not increase, its interaction with the non-receptor tyrosine kinase c-Abl and its phosphorylation on tyrosine residues did. c-Abl and p73 co-localize in the cytoplasm of spermatogonia and spermatocytes and in the residual bodies. Furthermore, c-Abl protein levels increase after irradiation. p63 was not found to co-localize or interact with c-Abl neither before nor after irradiation. In conclusion, in the testis ionizing radiation elevates cytoplasmic c-Abl that in turn interacts with p73. This may represent an additional, cytoplasmic, apoptotic pathway. Although less efficient than the p53 route, this pathway may cause spermatogonial apoptosis as observed in p53 deficient mice.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Proteínas de Membrana , Proteínas Nucleares/fisiologia , Proteínas Proto-Oncogênicas c-abl/fisiologia , Radiação Ionizante , Espermatozoides/efeitos da radiação , Transativadores , Animais , Citoplasma/metabolismo , Proteínas de Ligação a DNA/metabolismo , Genes Supressores de Tumor , Masculino , Camundongos , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Fosfoproteínas/fisiologia , Fosfotirosina/metabolismo , Proteínas Proto-Oncogênicas c-abl/metabolismo , Espermatócitos/metabolismo , Espermatócitos/efeitos da radiação , Espermatogônias/metabolismo , Espermatogônias/efeitos da radiação , Espermatozoides/metabolismo , Testículo/metabolismo , Proteína Tumoral p73 , Proteínas Supressoras de Tumor , Raios X
17.
Nat Genet ; 28(3): 216-7, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11431689

RESUMO

Mutations in SOX9 are associated with male-to-female sex reversal in humans. To analyze Sox9 function during sex determination, we ectopically expressed this gene in XX gonads. Here, we show that Sox9 is sufficient to induce testis formation in mice, indicating that it can substitute for the sex-determining gene Sry.


Assuntos
Transtornos do Desenvolvimento Sexual , Proteínas de Grupo de Alta Mobilidade/genética , Fatores de Transcrição/genética , Cromossomo X/genética , Animais , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Fenótipo , Fatores de Transcrição SOX9
18.
Cancer Res ; 61(8): 3267-71, 2001 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-11309277

RESUMO

We show with transgenic mice that targeted overexpression of glial cell line-derived neurotrophic factor (GDNF) in undifferentiated spermatogonia promotes malignant testicular tumors, which express germ-cell markers. The tumors are invasive and contain aneuploid cells, but no distant metastases have been found. By several histological, molecular, and histochemical characteristics, the GDNF-induced tumors mimic classic seminomas in men, representing a useful experimental model for testicular germ-cell tumors. The data also show that a deregulated stimulation of a normal proto-oncogene by its ligand can be an initiative event in carcinogenesis.


Assuntos
Proteínas do Tecido Nervoso/biossíntese , Seminoma/etiologia , Seminoma/metabolismo , Neoplasias Testiculares/etiologia , Neoplasias Testiculares/metabolismo , Aneuploidia , Animais , Modelos Animais de Doenças , Citometria de Fluxo , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Hibridização In Situ , Masculino , Camundongos , Camundongos Transgênicos , Fatores de Crescimento Neural/biossíntese , Fatores de Crescimento Neural/genética , Proteínas do Tecido Nervoso/genética , Seminoma/genética , Espermatogônias/metabolismo , Neoplasias Testiculares/genética
19.
J Cell Sci ; 114(Pt 4): 685-93, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11171374

RESUMO

Mammalian meiotic progression, like mitotic cell cycle progression, is regulated by cyclins and cyclin dependent kinases (CDKs). However, the unique requirements of meiosis (homologous synapsis, reciprocal recombination and the dual divisions that segregate first homologues, then sister chromatids) have led to different patterns of CDK expression. Here we show that Cdk4 colocalizes with replication protein A (RPA) on the synaptonemal complexes (SCs) of newly synapsed axes of homologously pairing bivalents, but disappears from these axes by mid-pachynema. The switch from the mitotic pattern of expression occurs during the last two spermatogonial divisions. Cdk2 colocalizes with MLH1, a mismatch repair protein at sites of reciprocal recombination in mid-late pachynema. In addition Cdk2 localizes to the telomeres of chromosomal bivalents throughout meiotic prophase. The mitotic pattern of expression of Cdk2 remains unchanged throughout the spermatogonial divisions, but is altered in meiosis of the spermatocytes.


Assuntos
Quinases Ciclina-Dependentes/metabolismo , Meiose , Proteínas Proto-Oncogênicas , Animais , Quinase 4 Dependente de Ciclina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oócitos/enzimologia , Espermatócitos/enzimologia
20.
Reproduction ; 121(2): 287-96, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11226053

RESUMO

Testicular tumours in dogs are of Sertoli cell, Leydig cell or germinal origin and mixed tumours are also frequently observed. The cellular components of mixed tumours are usually identified by histological examination but sometimes this is difficult. In this study, a panel of specific antibodies was used to identify the different cell types in testicular tumours by immunohistochemistry. Leydig cells were identified using an antibody against the LH receptor and an antibody against the steroidogenic enzyme 3beta-hydroxysteroid dehydrogenase (3beta-HSD), both of which are characteristic of Leydig cells in testes. Sertoli cells were identified using an antibody against the intermediate filament vimentin. Seminoma cells did not stain with any of these antibodies. Vimentin was used only in histologically complex cases. Eighty-six tumours, diagnosed histologically as 29 Sertoli cell tumours, 25 Leydig cell tumours, 19 seminomas and 13 mixed tumours, were studied. Feminization was observed in 17 dogs. Leydig cell tumours stained positively with the antibodies against the LH receptor and 3beta-HSD, whereas seminomas and Sertoli cell tumours were negative (unstained). The antibody against vimentin stained both Sertoli and Leydig cells, and tumours arising from these cells, but not seminomas. Immunohistochemistry revealed that three tumours identified histologically as Sertoli cell tumours were actually Leydig cell tumours. In 14 dogs the histological diagnosis appeared to be incomplete, as mixed tumours instead of pure types of tumours were identified in 11 dogs, and in three dogs mixed tumours appeared to be pure types. Hence, the histological diagnosis was insufficient in approximately 20% of dogs. Furthermore, immunohistochemical analysis of testis tumours revealed that feminization occurred in dogs with Sertoli cell tumours or Leydig cell tumours and their combinations, but not in dogs with a seminoma. In conclusion, incubation with antibodies against LH receptor and 3beta-HSD proved to be a consistently reliable method for identification of Leydig cell tumours in dogs. Vimentin can be used to discriminate between Sertoli cell tumours and seminomas. Overall, this panel of antibodies can be very useful for determination of the identity of testicular tumours in which histological characterization is complicated and the pathogenesis of feminization is not clear.


Assuntos
3-Hidroxiesteroide Desidrogenases/imunologia , Doenças do Cão/patologia , Imuno-Histoquímica/métodos , Receptores do LH/imunologia , Neoplasias Testiculares/veterinária , Vimentina/imunologia , Animais , Anticorpos Monoclonais , Doenças do Cão/metabolismo , Cães , Tumor de Células de Leydig/patologia , Tumor de Células de Leydig/veterinária , Masculino , Receptores do LH/metabolismo , Seminoma/patologia , Seminoma/veterinária , Tumor de Células de Sertoli/patologia , Tumor de Células de Sertoli/veterinária , Neoplasias Testiculares/patologia , Vimentina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...