Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Obstet Gynecol ; 126(1): 90-2, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25923021

RESUMO

BACKGROUND: Heterozygous gene mutations in fumarate hydratase can result in a syndrome characterized by hereditary (cutaneous and uterine) leiomyomatosis and renal cell cancer. This disorder has been described in more than 200 families, but the prevalence of the disease is unknown. CASE: A 22 year-old woman of Bangladeshi lineage presented with menorrhagia and pelvic pain secondary to uterine leiomyomas and underwent an abdominal myomectomy. Because of a family history of renal cell cancer, she was tested for fumarate hydratase mutations and found to be a carrier. As a result of the risk of renal cell cancer associated with this mutation, an annual surveillance plan was initiated. CONCLUSION: Fumarate hydratase gene mutations should be considered in women presenting with leiomyomas and a family history of renal cancer.


Assuntos
Carcinoma de Células Renais/genética , Códon sem Sentido , Fumarato Hidratase/genética , Predisposição Genética para Doença , Neoplasias Renais/genética , Leiomiomatose/genética , Neoplasias Uterinas/genética , Feminino , Marcadores Genéticos , Heterozigoto , Humanos , Leiomiomatose/diagnóstico , Masculino , Linhagem , Neoplasias Uterinas/diagnóstico , Adulto Jovem
2.
J Clin Invest ; 124(9): 3929-44, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25133429

RESUMO

Spermatogenesis is a complex, multistep process that maintains male fertility and is sustained by rare germline stem cells. Spermatogenic progression begins with spermatogonia, populations of which express distinct markers. The identity of the spermatogonial stem cell population in the undisturbed testis is controversial due to a lack of reliable and specific markers. Here we identified the transcription factor PAX7 as a specific marker of a rare subpopulation of A(single) spermatogonia in mice. PAX7+ cells were present in the testis at birth. Compared with the adult testis, PAX7+ cells constituted a much higher percentage of neonatal germ cells. Lineage tracing in healthy adult mice revealed that PAX7+ spermatogonia self-maintained and produced expanding clones that gave rise to mature spermatozoa. Interestingly, in mice subjected to chemotherapy and radiotherapy, both of which damage the vast majority of germ cells and can result in sterility, PAX7+ spermatogonia selectively survived, and their subsequent expansion contributed to the recovery of spermatogenesis. Finally, PAX7+ spermatogonia were present in the testes of a diverse set of mammals. Our data indicate that the PAX7+ subset of A(single) spermatogonia functions as robust testis stem cells that maintain fertility in normal spermatogenesis in healthy mice and mediate recovery after severe germline injury, such as occurs after cancer therapy.


Assuntos
Fator de Transcrição PAX7/fisiologia , Células-Tronco/química , Testículo/citologia , Animais , Infertilidade Masculina/etiologia , Masculino , Camundongos , Fator de Transcrição PAX7/análise , Espermatogênese , Espermatogônias/fisiologia , Testículo/metabolismo
3.
Biol Reprod ; 88(4): 103, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23486915

RESUMO

The Foxos are key effectors of the PI3K/Akt signaling pathway and regulate diverse physiologic processes. Two of these factors, Foxo1 and Foxo3, serve specific roles in reproduction in the mouse. Foxo3 is required for suppression of primordial follicle activation in females, while Foxo1 regulates spermatogonial stem cell maintenance in males. In the mouse ovary, Foxo1 is highly expressed in somatic cells (but not in oocytes), suggesting an important functional role for Foxo1 in these cells. Given that invertebrate model species such as Caenorhabditis elegans and Drosophila melanogaster harbor a single ancestral Foxo homolog, these observations suggest that gene duplication conferred a selective advantage by permitting the Foxos to adopt distinct roles in oogenesis and spermatogenesis. Our objective was to determine if the remarkably specific expression patterns of Foxo1 and Foxo3 in mouse gonads (and, by inference, Foxo function) are conserved in diverse mammalian species. Western blotting was used to validate isoform-specific antibodies in rodents, companion animals, farm animals, nonhuman primates, and humans. Following validation of each antibody, immunohistochemistry was performed to ascertain Foxo1 and Foxo3 gonadal expression patterns. While Foxo1 expression in spermatogonia and granulosa cells was conserved in each species evaluated, Foxo3 expression in oocytes was not. Our findings suggest that Foxo3 is not uniquely required for primordial follicle maintenance in nonrodent species and that other Foxos, particularly Foxo1, may contribute to oocyte maintenance in a functionally redundant manner.


Assuntos
Fatores de Transcrição Forkhead/genética , Gônadas/metabolismo , Mamíferos/genética , Animais , Caenorhabditis elegans , Gatos , Cães , Drosophila melanogaster , Evolução Molecular , Feminino , Fatores de Transcrição Forkhead/metabolismo , Expressão Gênica , Especiação Genética , Humanos , Masculino , Mamíferos/metabolismo , Camundongos , Muridae , Primatas , Ratos , Especificidade da Espécie , Peixe-Zebra/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...