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.
Anim Reprod Sci ; 247: 107091, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36228424

RESUMO

During the reproduction stage of poultry, a single follicle is selected from a cohort of 6-8 mm small yellow follicles to initiate rapid growth and final ovulation almost daily. In the process, follicle-stimulating hormone (FSH) plays a pivotal role by interacting with intraovarian factors, including insulin-like growth Factor 1 (IGF1). The objective of this study was to analyze whether IGF1 coordinates with FSH to affect the characteristics of granulosa cells from prehierarchical follicles. After treating granulosa cells with 50 ng/mL FSH and 200 ng/mL IGF1, we detected the proliferation and apoptosis of granulosa cells using flow cytometry. The percentage of G1 phase granulosa cells was increased, and the percentage of mitotic cells and apoptotic cells was reduced under IGF1 treatment. The expression levels of the steroidogenic acute regulatory protein gene, cytochrome P450 side-chain cleavage gene and 3ß-hydroxysteroid dehydrogenase gene, which are related to steroidogenic synthesis, were reduced by cotreatment with FSH and IGF1. The expression of the cell proliferation- or apoptosis-related genes cyclin dependent kinase 2, cyclin D2, B-cell leukemia/lymphoma 2, and BCL2 like 1 and the ratio of B-cell leukemia/lymphoma 2/BCL2-associated X were increased by treatment with IGF1. There was a decrease in the expression of caspase3 after treatment with FSH and IGF1. All these results showed that IGF1 reduced the expression of genes involved in progesterone synthesis, stimulated proliferation and inhibited apoptosis in granulosa cells. Thus, IGF1 may be one of the factors involved in affecting FSH responsiveness and maintaining the undifferentiated state of prehierarchical follicles before follicle selection.


Assuntos
Galinhas , Fator de Crescimento Insulin-Like I , Feminino , Animais , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/farmacologia , Fator de Crescimento Insulin-Like I/metabolismo , Galinhas/fisiologia , Células da Granulosa/fisiologia , Hormônio Foliculoestimulante/farmacologia , Hormônio Foliculoestimulante/metabolismo , Apoptose/fisiologia , Proliferação de Células , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Células Cultivadas
2.
Front Cell Dev Biol ; 10: 748862, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35237591

RESUMO

Growing evidence suggests that adverse intrauterine environments could affect the long-term health of offspring. Recent evidence indicates that gestational diabetes mellitus (GDM) is associated with neurocognitive changes in offspring. However, the mechanism remains unclear. Using a GDM mouse model, we collected hippocampi, the structure critical to cognitive processes, for electron microscopy, methylome and transcriptome analyses. Reduced representation bisulfite sequencing (RRBS) and RNA-seq in the GDM fetal hippocampi showed altered methylated modification and differentially expressed genes enriched in common pathways involved in neural synapse organization and signal transmission. We further collected fetal mice brains for metabolome analysis and found that in GDM fetal brains, the metabolites displayed significant changes, in addition to directly inducing cognitive dysfunction, some of which are important to methylation status such as betaine, fumaric acid, L-methionine, succinic acid, 5-methyltetrahydrofolic acid, and S-adenosylmethionine (SAM). These results suggest that GDM affects metabolites in fetal mice brains and further affects hippocampal DNA methylation and gene regulation involved in cognition, which is a potential mechanism for the adverse neurocognitive effects of GDM in offspring.

3.
BMC Endocr Disord ; 21(1): 240, 2021 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-34856980

RESUMO

BACKGROUND: Pentraxin 3 (PTX3) - a crucial humoral innate immunity component - is related to obesity and cardiovascular complications in women who suffer from polycystic ovary syndrome (PCOS). However, the circulating PTX3 level in PCOS is still debated. In this study, we aimed to evaluate PTX3 plasma levels in PCOS women of childbearing age, and find possible endocrine/metabolic factors that could affect this level. METHODS: A total of 360 women were enrolled: 120 PCOS women and 240 body mass index (BMI) matched normally ovulating women. Blood samples were collected on the third day of natural menstrual cycle or from the bleeding after progesterone withdrawal. The PTX3 concentration was measured by immunoassay. RESULTS: The PTX3 plasma level was significantly higher in PCOS women compared to controls. There was a positive correlation between PTX3 plasma level and PCOS diagnosis, overweight, cycle length, serum LH to FSH ratio, estradiol, total testosterone (TT) on the third day of menstrual cycle, antral follicle count (AFC), as well as uric acid. Multivariant linear regression analysis indicated that participants' serum PTX3 levels were proportional to the circulating TT level, existence of PCOS, basal estradiol level and AFC. CONCLUSIONS: Overall, the circulating PTX3 level was elevated in PCOS women and significantly associated with the presence of hyperandrogenism. This study provided the basis for further in-depth researches regarding PTX3 role in PCOS pathophysiology.


Assuntos
Proteína C-Reativa/análise , Hiperandrogenismo/sangue , Síndrome do Ovário Policístico/sangue , Componente Amiloide P Sérico/análise , Adulto , Índice de Massa Corporal , Estudos de Casos e Controles , Estradiol/sangue , Feminino , Humanos , Folículo Ovariano/patologia , Sobrepeso/sangue , Síndrome do Ovário Policístico/patologia , Testosterona/sangue
4.
Front Endocrinol (Lausanne) ; 12: 710221, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34531826

RESUMO

Mounting evidence has shown that intrauterine hyperglycemia exposure during critical stages of development may be contributing to the increasing prevalence of diabetes. However, little is known about the mechanisms responsible for offspring metabolic disorder. In this present study, we explored intrauterine hyperglycemia exposure on fetal pancreatic metabolome, and its potential link to impaired glucose tolerance in adult offspring. Here, using a GDM mouse model, we found the metabolome profiling of pancreas from male and female fetus showing altered metabolites in several important pathways, including 5-methylcytosine, α-KG, branched-chain amino acids, and cystine, which are associated with epigenetic modification, insulin secretion, and intracellular redox status, respectively. This finding suggests that intrauterine exposure to hyperglycemia could cause altered metabolome in pancreas, which might be a metabolism-mediated mechanism for GDM-induced intergenerational diabetes predisposition.


Assuntos
Biomarcadores/metabolismo , Diabetes Gestacional/fisiopatologia , Feto/metabolismo , Intolerância à Glucose/patologia , Hiperglicemia/patologia , Metaboloma , Útero/fisiopatologia , Animais , Epigênese Genética , Feminino , Feto/patologia , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Hiperglicemia/genética , Hiperglicemia/metabolismo , Masculino , Pâncreas/metabolismo , Pâncreas/patologia , Gravidez , Fatores Sexuais
5.
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
6.
Mol Genet Genomic Med ; 8(3): e1130, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31971667

RESUMO

BACKGROUND: Germline mutations in PTEN are associated with the PTEN hamartoma tumor syndrome (PHTS), an umbrella term used to describe a spectrum of autosomal-dominant disorders characterized by variable phenotypic manifestations associated with cell or tissue overgrowth. We report a boy who developed severe progressive abdominal distention due to a dramatic adipose mass from the age of 7 months and developed recurrent hypoinsulinemic hypoglycemia that led to seizures at the age of 4 years. METHODS: Trio-based whole-genome sequencing was performed by using blood DNA from the child and his parents. The possible pathogenic variants were verified by Sanger sequencing. Functional characterization of the identified variant was completed by western blot. RESULTS: The child inherited a single-nucleotide deletion NM_000314.6:c.849delA (p.Glu284Argfs) in the tumor suppressor gene PTEN from his father. The paternal family members have a history of cancer. It is conceivable that PTEN loss-of-function induced the adipose tumor growth and hypoglycemia, although the proband did not meet the usual diagnosis criteria of Cowden syndrome or Bannayan-Riley-Ruvalcaba syndrome that are characterized by germline mutations of PTEN. CONCLUSION: This case underlines the variability of phenotypes associated with PTEN germline mutations and provides useful information for diagnosis and genetic counseling of PTEN-related diseases for pediatric patients.


Assuntos
Neoplasias Abdominais/genética , Hipoglicemia/genética , Neoplasias Lipomatosas/genética , PTEN Fosfo-Hidrolase/genética , Fenótipo , Convulsões/genética , Neoplasias Abdominais/patologia , Pré-Escolar , Humanos , Hipoglicemia/patologia , Mutação com Perda de Função , Masculino , Neoplasias Lipomatosas/patologia , Linhagem , Convulsões/patologia , Síndrome
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA