Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nature ; 626(7997): 151-159, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38233525

RESUMO

Enhancers control the location and timing of gene expression and contain the majority of variants associated with disease1-3. The ZRS is arguably the most well-studied vertebrate enhancer and mediates the expression of Shh in the developing limb4. Thirty-one human single-nucleotide variants (SNVs) within the ZRS are associated with polydactyly4-6. However, how this enhancer encodes tissue-specific activity, and the mechanisms by which SNVs alter the number of digits, are poorly understood. Here we show that the ETS sites within the ZRS are low affinity, and identify a functional ETS site, ETS-A, with extremely low affinity. Two human SNVs and a synthetic variant optimize the binding affinity of ETS-A subtly from 15% to around 25% relative to the strongest ETS binding sequence, and cause polydactyly with the same penetrance and severity. A greater increase in affinity results in phenotypes that are more penetrant and more severe. Affinity-optimizing SNVs in other ETS sites in the ZRS, as well as in ETS, interferon regulatory factor (IRF), HOX and activator protein 1 (AP-1) sites within a wide variety of enhancers, cause gain-of-function gene expression. The prevalence of binding sites with suboptimal affinity in enhancers creates a vulnerability in genomes whereby SNVs that optimize affinity, even slightly, can be pathogenic. Searching for affinity-optimizing SNVs in genomes could provide a mechanistic approach to identify causal variants that underlie enhanceropathies.


Assuntos
Elementos Facilitadores Genéticos , Extremidades , Polidactilia , Proteínas Proto-Oncogênicas c-ets , Humanos , Elementos Facilitadores Genéticos/genética , Extremidades/embriologia , Extremidades/patologia , Mutação com Ganho de Função , Proteínas de Homeodomínio/metabolismo , Fatores Reguladores de Interferon/metabolismo , Especificidade de Órgãos/genética , Penetrância , Fenótipo , Polidactilia/embriologia , Polidactilia/genética , Polidactilia/patologia , Polimorfismo de Nucleotídeo Único , Ligação Proteica , Proteínas Proto-Oncogênicas c-ets/metabolismo , Fator de Transcrição AP-1/metabolismo
2.
Endocrinology ; 162(12)2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34529765

RESUMO

Polycystic ovary syndrome (PCOS) is a common reproductive disorder characterized by elevated androgens and antimüllerian hormone (AMH). These hormones remain elevated throughout pregnancy, and potential effects of hormone exposure on offspring from women with PCOS remain largely unexplored. Expanding on recent reports of prenatal AMH exposure in mice, we have fully characterized the reproductive consequences of prenatal AMH (pAMH) exposure throughout the lifespan of first- and second-generation offspring of both sexes. We also sought to elucidate mechanisms underlying pAMH-induced reproductive effects. There is a known reciprocal relationship between AMH and androgens, and in PCOS and PCOS-like animal models, androgen feedback is dysregulated at the level of the hypothalamus. Kisspeptin neurons express androgen receptors and play a critical role in sexual development and function. We therefore hypothesized that pAMH-induced reproductive phenotypes would be mediated by androgen signaling at the level of kisspeptin cells. We tested the pAMH model in kisspeptin-specific androgen receptor knockout (KARKO) mice and found that virtually all pAMH-induced phenotypes assayed are eliminated in KARKO offspring compared to littermate controls. By demonstrating the necessity of androgen receptor in kisspeptin cells to induce pAMH phenotypes, we have advanced understanding of the interactions between AMH and androgens in the context of prenatal exposure, which could have significant implications for children of women with PCOS.


Assuntos
Hormônio Antimülleriano/farmacologia , Efeitos Tardios da Exposição Pré-Natal , Receptores Androgênicos/fisiologia , Reprodução/efeitos dos fármacos , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Feminino , Gônadas/efeitos dos fármacos , Gônadas/metabolismo , Kisspeptinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Receptores Androgênicos/metabolismo
3.
Mol Cell Endocrinol ; 530: 111286, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-33872733

RESUMO

Within pituitary gonadotropes, the gonadotropin-releasing hormone receptor (GnRHR) receives hypothalamic input from GnRH neurons that is critical for reproduction. Previous studies have suggested that androgens may regulate GnRHR, although the mechanisms remain unknown. In this study, we demonstrated that androgens positively regulate Gnrhr mRNA in mice. We then investigated the effects of androgens and androgen receptor (AR) on Gnrhr promoter activity in immortalized mouse LßT2 cells, which represent mature gonadotropes. We found that AR positively regulates the Gnrhr proximal promoter, and that this effect requires a hormone response element (HRE) half site at -159/-153 relative to the transcription start site. We also identified nonconsensus, full-length HREs at -499/-484 and -159/-144, which are both positively regulated by androgens on a heterologous promoter. Furthermore, AR associates with the Gnrhr promoter in ChIP. Altogether, we report that GnRHR is positively regulated by androgens through recruitment of AR to the Gnrhr proximal promoter.


Assuntos
Androgênios/farmacologia , Gonadotrofos/citologia , Receptores Androgênicos/metabolismo , Receptores LHRH/genética , Animais , Linhagem Celular , Sequenciamento de Cromatina por Imunoprecipitação , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Gonadotrofos/efeitos dos fármacos , Gonadotrofos/metabolismo , Masculino , Camundongos , Regiões Promotoras Genéticas , Receptores LHRH/metabolismo , Análise de Sequência de DNA
4.
Wiley Interdiscip Rev Syst Biol Med ; 12(2): e1467, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31808313

RESUMO

Successful development depends on the precise tissue-specific regulation of genes by enhancers, genetic elements that act as switches to control when and where genes are expressed. Because enhancers are critical for development, and the majority of disease-associated mutations reside within enhancers, it is essential to understand which sequences within enhancers are important for function. Advances in sequencing technology have enabled the rapid generation of genomic data that predict putative active enhancers, but functionally validating these sequences at scale remains a fundamental challenge. Herein, we discuss the power of genome-wide strategies used to identify candidate enhancers, and also highlight limitations and misconceptions that have arisen from these data. We discuss the use of massively parallel reporter assays to test enhancers for function at scale. We also review recent advances in our ability to study gene regulation during development, including CRISPR-based tools to manipulate genomes and single-cell transcriptomics to finely map gene expression. Finally, we look ahead to a synthesis of complementary genomic approaches that will advance our understanding of enhancer function during development. This article is categorized under: Physiology > Mammalian Physiology in Health and Disease Developmental Biology > Developmental Processes in Health and Disease Laboratory Methods and Technologies > Genetic/Genomic Methods.


Assuntos
Elementos Facilitadores Genéticos/genética , Genômica , Animais , Desenvolvimento Embrionário/genética , Edição de Genes , Regulação da Expressão Gênica , Genes Reporter , Humanos , Análise de Célula Única
5.
Endocrinology ; 159(4): 1734-1747, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29471436

RESUMO

Polycystic ovary syndrome (PCOS), the most common endocrinopathy in women of reproductive age, is characterized by hyperandrogenism, anovulation, and polycystic ovaries. Although its etiology is unknown, excess androgens are thought to be a critical factor driving the pathology of PCOS. We previously demonstrated that continuous exposure to the aromatase inhibitor letrozole (LET) in mice produces many hallmarks of PCOS, including elevated testosterone (T) and luteinizing hormone, anovulation, and obesity. In the current study, we sought to determine whether androgen receptor (AR) actions are responsible for any of the phenotypes observed in LET mice. C57BL/6 female mice were subcutaneously implanted with LET or placebo control and subsequently treated with the nonsteroidal AR antagonist flutamide or vehicle control. Flutamide treatment in LET females reversed elevated T levels and restored ovarian expression of Cyp17a1 (critical for androgen synthesis) to normal levels. Pituitary expression of Lhb was decreased in LET females that received flutamide treatment, with no changes in expression of Fshb or Gnrhr. Flutamide treatment also restored estrous cycling and reduced the number of ovarian cyst-like follicles in LET females. Furthermore, body weight and adipocyte size were decreased in flutamide-treated LET females. Altogether, our findings provide strong evidence that AR signaling is responsible for many key reproductive and metabolic PCOS phenotypes and further establish the LET mouse model as an important tool for the study of androgen excess.


Assuntos
Antagonistas de Androgênios/uso terapêutico , Ciclo Estral/efeitos dos fármacos , Flutamida/uso terapêutico , Ovário/efeitos dos fármacos , Hipófise/efeitos dos fármacos , Síndrome do Ovário Policístico/tratamento farmacológico , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Antagonistas de Androgênios/farmacologia , Animais , Inibidores da Aromatase , Peso Corporal/efeitos dos fármacos , Modelos Animais de Doenças , Ciclo Estral/metabolismo , Feminino , Flutamida/farmacologia , Hormônio Foliculoestimulante/metabolismo , Letrozol , Hormônio Luteinizante/metabolismo , Camundongos , Nitrilas , Ovário/metabolismo , Fenótipo , Hipófise/metabolismo , Síndrome do Ovário Policístico/induzido quimicamente , Síndrome do Ovário Policístico/metabolismo , Receptores LHRH/metabolismo , Reprodução/efeitos dos fármacos , Esteroide 17-alfa-Hidroxilase/metabolismo , Testosterona/sangue , Triazóis
6.
Biol Reprod ; 93(3): 69, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26203175

RESUMO

Polycystic ovary syndrome (PCOS) pathophysiology is poorly understood, due partly to lack of PCOS animal models fully recapitulating this complex disorder. Recently, a PCOS rat model using letrozole (LET), a nonsteroidal aromatase inhibitor, mimicked multiple PCOS phenotypes, including metabolic features absent in other models. Given the advantages of using genetic and transgenic mouse models, we investigated whether LET produces a similar PCOS phenotype in mice. Pubertal female C57BL/6N mice were treated for 5 wk with LET, which resulted in increased serum testosterone and normal diestrus levels of estradiol, similar to the hyperandrogenemia and follicular phase estrogen levels of PCOS women. As in PCOS, ovaries from LET mice were larger, polycystic, and lacked corpora lutea versus controls. Most LET females were acyclic, and all were infertile. LET females displayed elevated serum LH levels and higher Lhb mRNA in the pituitary. In contrast, serum FSH and Fshb were significantly reduced in LET females, demonstrating differential effects on gonadotropins, as in PCOS. Within the ovary, LET females had higher Cyp17, Cyp19, and Fsh receptor mRNA expression. In the hypothalamus, LET females had higher kisspeptin receptor mRNA expression but lower progesterone receptor mRNA levels. LET females also gained more weight than controls, had increased abdominal adiposity and adipocyte size, elevated adipose inflammatory mRNA levels, and impaired glucose tolerance, mirroring the metabolic phenotype in PCOS women. This is the first report of a LET paradigm in mice that recapitulates both reproductive and metabolic PCOS phenotypes and will be useful to genetically probe the PCOS condition.


Assuntos
Inibidores Enzimáticos/toxicidade , Nitrilas/toxicidade , Síndrome do Ovário Policístico/induzido quimicamente , Síndrome do Ovário Policístico/patologia , Reprodução/efeitos dos fármacos , Triazóis/toxicidade , Animais , Corpo Lúteo/metabolismo , Diestro/metabolismo , Ciclo Estral/efeitos dos fármacos , Feminino , Hiperandrogenismo/sangue , Hiperandrogenismo/induzido quimicamente , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Kisspeptinas/biossíntese , Kisspeptinas/genética , Letrozol , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Síndrome do Ovário Policístico/metabolismo , Gravidez , Testosterona/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...