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

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Assist Reprod Genet ; 38(1): 33-40, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32926298

RESUMO

OBJECTIVE: To study the effect of aging on ovarian circadian rhythm. DESIGN: Human and animal study. SETTING: University hospital and research laboratory. PATIENTS/ANIMALS: Human granulosa cells were obtained by follicular aspiration from women undergoing in vitro fertilization (IVF), and ovarian and liver tissues were obtained from female C57BL/6 mice. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): Expression of circadian genes in young and older human granulosa cells and circadian rhythm in ovaries and livers of young and older mice. RESULT(S): All examined circadian clock genes in human granulosa cells showed a downward trend in expression with aging, and their mRNA expression levels were negatively correlated with age (P < 0.05). Older patients (≥ 40 years of age) had significantly reduced serum anti-Müllerian hormone (AMH) levels. Except for Rev-erbα, all other examined circadian clock genes were positively correlated with the level of AMH (P < 0.05). The circadian rhythm in the ovaries of older mice (8 months) was changed significantly relative to that in ovaries of young mice (12 weeks), although the circadian rhythm in the livers of older mice was basically consistent with that of young mice. CONCLUSION(S): Lower ovarian reserve in older women is partially due to ovarian circadian dysrhythmia as a result of aging.


Assuntos
Envelhecimento/genética , Ritmo Circadiano/genética , Fígado/metabolismo , Ovário/metabolismo , Envelhecimento/patologia , Animais , Hormônio Antimülleriano/sangue , Feminino , Fertilização in vitro , Células da Granulosa/metabolismo , Humanos , Camundongos , Reserva Ovariana/genética , Ovário/crescimento & desenvolvimento , Ovário/patologia , RNA Mensageiro/genética
2.
Artigo em Inglês | MEDLINE | ID: mdl-32081430

RESUMO

Polycystic ovary syndrome (PCOS) is a complicated reproductive endocrine disease characterized by hyperandrogenism, polycystic ovaries, and anovulation. Previous studies have revealed that androgen receptors (ARs) are strongly associated with hyperandrogenism and abnormalities in folliculogenesis in patients with PCOS. However, the kinases responsible for androgen receptor activity, especially in granulosa cells, and the role of casein kinase 2α (CK2α) specifically in the pathogenesis of PCOS, remain unknown. Here, we show that both CK2α protein and mRNA levels were higher in luteinized granulosa cells of patients with PCOS compared with non-PCOS, as well as in the ovarian tissues of mice with a dehydroepiandrosterone-induced PCOS-like phenotype, compared with controls. In addition, CK2α not only interacted with AR in vivo and in vitro, but it also phosphorylated and stabilized AR, triggering AR and ovulation related genes excessive expression. CK2α also promoted cell proliferation in the KGN cell line and inhibited apoptosis. Collectively, the finding highlighted that the CK2α-AR axis probably caused the etiology of the PCOS. Thus, CK2α might be a promising clinical therapeutic target for PCOS treatment.

3.
J Mol Cell Cardiol ; 128: 1-10, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30641088

RESUMO

High circulating androgen in women with polycystic ovary syndrome (PCOS) may increase the risk of cardiovascular disease in offspring. The aim of the present study is to investigate whether maternal androgen excess in the rat PCOS model would lead to cardiac hypertrophy in offspring. Maternal testosterone propionate (maternal-TP)-treated adult female offspring displayed cardiac hypertrophy associated with local high cardiac dihydrotestosterone (DHT). The molecular markers of cardiac hypertrophy along with androgen receptor (AR) and PKCδ, were increased in the Maternal-TP group. Treatment of primary neonatal rat ventricular cardiomyocytes (NRCMs) and H9c2 cells with DHT significantly increased cell size and upregulated PKCδ expression, which could be attenuated by AR antagonist. Treatment with phorbol 12-myristate 13-acetate (PMA), a PKC activator, significantly increased cell size and upregulated myh7 level. Rottlerin, that may inhibit PKCδ, significantly reduced the hypertrophic effect of DHT and PMA on NRCMs and H9c2 cells. Chromatin immunoprecipitation revealed that AR could bind to Pkcδ promoter. Our results indicate that prenatal exposure to testosterone may induce cardiac hypertrophy in adult female rats through enhanced Pkcδ expression in cardiac myocytes.


Assuntos
Cardiomegalia/genética , Síndrome do Ovário Policístico/genética , Proteína Quinase C-delta/genética , Receptores Androgênicos/genética , Acetofenonas/farmacologia , Androgênios/genética , Androgênios/metabolismo , Animais , Animais Recém-Nascidos , Benzopiranos/farmacologia , Cardiomegalia/etiologia , Cardiomegalia/patologia , Di-Hidrotestosterona/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Síndrome do Ovário Policístico/complicações , Síndrome do Ovário Policístico/patologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/patologia , Proteína Quinase C-delta/antagonistas & inibidores , Ratos , Transdução de Sinais/efeitos dos fármacos , Testosterona/farmacologia , Acetato de Tetradecanoilforbol/farmacologia
4.
Front Physiol ; 11: 1108, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33013474

RESUMO

Women with polycystic ovarian syndrome (PCOS) often have a history of infertility and poor pregnancy outcome. The character of the lower genital tract (LGT) microbiome of these patients is still unknown. We collected both vaginal and cervical canal swabs from 47 PCOS patients (diagnosed by the Rotterdam Criteria) and 50 healthy reproductive-aged controls in this study. Variable regions 3-4 (V3-4) were sequenced and analyzed. Operational taxonomic unit (OTU) abundance was noted for all samples. Taxa that discriminated between PCOS and healthy women was calculated by linear discriminant analysis effect size (LEFSe). Results from 97 paired vaginal and cervical canal samples collected from 97 women [mean age 30 (±4 years)] were available for analysis. Using the Rotterdam Criteria, 47 women were diagnosed with PCOS (PCOS, n = 47; control, n = 50). There was no significant difference between cervical canal microbiome and vaginal microbiome from the same individual, however, Lactobacillus spp. was less abundant in both vaginal and cervical canal microbiome of PCOS patients. Several non-Lactobacillus taxa including Gardnerella_vaginalis_00703mash, Prevotella_9_other, and Mycoplasma hominis, were more abundant in the LGT microbiota of PCOS patients. There is a difference between the microorganism in the LGT of patients with PCOS and healthy reproductive-aged women.

6.
EBioMedicine ; 61: 103058, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33096483

RESUMO

BACKGROUND: Disordered folliculogenesis is a core characteristic of polycystic ovary syndrome (PCOS) and androgen receptors (ARs) are closely associated with hyperandrogenism and abnormalities in folliculogenesis in PCOS. However, whether the new AR binding partner phosphoglycerate kinase 1 (PGK1) in granulosa cells (GCs) plays a key role in the pathogenesis of PCOS remains unclear. METHODS: We identified the new AR binding partner PGK1 by co-IP (co-immunoprecipitation) in luteinized GCs, and reconfirmed by co-IP, co-localization and GST pull down assay, and checked PGK1 expression levels with qRT-PCR and western blotting. Pharmaceuticals rescue assays in mice, and metabolism assay, AR protein stability and RNA-seq of PGK1 targets in cells proved the function in PCOS. FINDINGS: PGK1 and AR are highly expressed in PCOS luteinized GCs and PCOS-like mouse ovarian tissues. PGK1 regulated glucose metabolism and deteriorated PCOS-like mouse metabolic disorder, and paclitaxel rescued the phenotype of PCOS-like mice and reduced ovarian PGK1 and AR protein levels. PGK1 inhibited AR ubiquitination levels and increased AR stability in an E3 ligase SKP2-dependent manner. Additionally, PGK1 promoted AR nuclear translocation, and RNA-seq data showed that critical ovulation-related genes were regulated by the PGK1-AR axis. INTERPRETATION: PGK1 regulated GCs metabolism and interacted with AR to regulate the expression of key ovulation genes, and also mediated cell proliferation and apoptosis, which resulted in the etiology of PCOS. This work highlights the pathogenic mechanism and represents a novel therapeutic target for PCOS. FUNDING: National Key Research and Development Program of China; National Natural Science Foundation of China grant.


Assuntos
Glucose/metabolismo , Células da Granulosa/metabolismo , Ovulação , Fosfoglicerato Quinase/metabolismo , Receptores Androgênicos/metabolismo , Proteínas Quinases Associadas a Fase S/metabolismo , Adulto , Animais , Apoptose/genética , Biomarcadores , Metabolismo dos Carboidratos , Linhagem Celular , Modelos Animais de Doenças , Suscetibilidade a Doenças , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Doenças Metabólicas/etiologia , Doenças Metabólicas/metabolismo , Camundongos , Modelos Biológicos , Ovulação/genética , Fosfoglicerato Quinase/genética , Síndrome do Ovário Policístico/diagnóstico , Síndrome do Ovário Policístico/etiologia , Síndrome do Ovário Policístico/metabolismo , Ligação Proteica , Estabilidade Proteica , Transporte Proteico , Proteínas Quinases Associadas a Fase S/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA