Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Biol Reprod ; 107(1): 148-156, 2022 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-35774031

RESUMEN

The prevalence of gestational diabetes mellitus (GDM) is increasing rapidly. In addition to the metabolic disease risks, GDM might increase the risks of cryptorchidism in children. However, its mechanism involved in abnormalities of the male reproductive system is still unclear. The purpose of this study was to study the effects of GDM on the development of mouse fetal Leydig cells (FLCs) and Sertoli cells (SCs). Pregnant mice were treated on gestational days 6.5 and 12.5 with streptozotocin (100 mg/kg) or vehicle (sodium citrate buffer). Leydig cell and SC development and functions were evaluated by investigating serum testosterone levels, cell number and distribution, genes, and protein expression. GDM decreased serum testosterone levels, the anogenital distance, and the level of desert hedgehog in SCs of testes of male offspring. FLC number was also decreased in testes of GDM offspring by delaying the commitment of stem Leydig cells into the Leydig cell lineage. RNA-seq showed that FOXL2, RSPO1/ß-catenin signaling was activated and Gsk3ß signaling was inhibited in GDM offspring testis. In conclusion, GDM disrupted reproductive tract and testis development in mouse male offspring via altering genes related to development.


Asunto(s)
Diabetes Gestacional , Testículo , Animales , Diabetes Gestacional/metabolismo , Femenino , Desarrollo Fetal , Humanos , Células Intersticiales del Testículo/metabolismo , Masculino , Ratones , Embarazo , Células de Sertoli/metabolismo , Testículo/metabolismo , Testosterona
2.
FASEB J ; 33(4): 5425-5439, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30759346

RESUMEN

Brown adipose tissue (BAT) is an exclusive tissue of nonshivering thermogenesis. It is fueled by lipids and glucose and involved in energy and metabolic homeostasis. Intrauterine exposure to hyperglycemia during gestational diabetes mellitus may result in abnormal fetal development and metabolic phenotypes in adulthood. However, whether intrauterine hyperglycemia influences the development of BAT is unknown. In this study, mouse embryos were exposed to the intrauterine hyperglycemia environment by injecting streptozocin into pregnant mice at 1 d post coitum (dpc). The structure of BAT was examined by hematoxylin and eosin staining and immunohistochemical analysis. The glucose uptake in BAT was measured in vivo by [18F]-fluoro-2-deoxyglucose-micro-positron emission tomography. The gene expression in BAT was determined by real-time PCR, and the 5'-C-phosphate-G-3' site-specific methylation was quantitatively analyzed. Intrauterine hyperglycemia exposure resulted in the impaired structure of BAT and decreased glucose uptake function in BAT in adulthood. The expressions of the genes involved in thermogenesis and mitochondrial respiratory chain in BAT, such as Ucp1, Cox5b, and Elovl3, were down-regulated by intrauterine hyperglycemia exposure at 18.5 dpc and at 16 wk of age. Furthermore, higher methylation levels of Ucp1, Cox5b, and Elovl3 were found in offspring of mothers with streptozotocin-induced diabetes. Our results provide the evidence for enduring inhibitory effects of intrauterine hyperglycemia on BAT development in offspring. Intrauterine hyperglycemia is associated with increased DNA methylation of the BAT specific genes in offspring, which support an epigenetic involvement.-Yu, D.-Q., Lv, P.-P., Yan, Y.-S., Xu, G.-X., Sadhukhan, A., Dong, S., Shen, Y., Ren, J., Zhang, X.-Y., Feng, C., Huang, Y.-T., Tian, S., Zhou, Y., Cai, Y.-T., Ming, Z.-H., Ding, G.-L., Zhu, H., Sheng, J.-Z., Jin, M., Huang, H.-F. Intrauterine exposure to hyperglycemia retards the development of brown adipose tissue.


Asunto(s)
Tejido Adiposo Pardo/fisiopatología , Hiperglucemia/fisiopatología , Útero/fisiopatología , Tejido Adiposo Pardo/metabolismo , Animales , Metilación de ADN/fisiología , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Diabetes Gestacional/inducido químicamente , Diabetes Gestacional/metabolismo , Diabetes Gestacional/fisiopatología , Transporte de Electrón/fisiología , Femenino , Expresión Génica/fisiología , Glucosa/metabolismo , Hiperglucemia/metabolismo , Ratones , Ratones Endogámicos ICR , Embarazo , Estreptozocina/farmacología , Termogénesis/fisiología , Útero/metabolismo
3.
Nutr Diabetes ; 14(1): 56, 2024 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-39043630

RESUMEN

BACKGROUND: Maternal diabetes mellitus can influence the development of offspring. Gestational diabetes mellitus (GDM) creates a short-term intrauterine hyperglycaemic environment in offspring, leading to glucose intolerance in later life, but the long-term effects and specific mechanism involved in skeletal muscle dysfunction in offspring remain to be clarified. METHODS: Pregnant mice were divided into two groups: The GDM group was intraperitoneally injected with 100 mg/kg streptozotocin on gestational days (GDs) 6.5 and 12.5, while the control (CTR) group was treated with vehicle buffer. Only pregnant mice whose random blood glucose level was higher than 16.8 mmol/L beginning on GD13.5 were regarded as the GDM group. The growth of the offspring was monitored, and the glucose tolerance test was performed at different time points. Body composition analysis and immunohistochemical methods were used to evaluate the development of lean mass at 8 weeks. The exercise capacity and grip strength of the male mouse offspring were assessed at the same period. Transmission electron microscopy was used to observe the morphology inside skeletal muscle at 8 weeks and as a foetus. The genes and proteins associated with mitochondrial biogenesis and oxidative metabolism were investigated. We also coanalyzed RNA sequencing and proteomics data to explore the underlying mechanism. Chromatin immunoprecipitation and bisulfite-converted DNA methylation detection were performed to evaluate this phenomenon. RESULTS: Short-term intrauterine hyperglycaemia inhibited the growth and reduced the lean mass of male offspring, leading to decreased endurance exercise capacity. The myofiber composition of the tibialis anterior muscle of GDM male offspring became more glycolytic and less oxidative. The morphology and function of mitochondria in the skeletal muscle of GDM male offspring were destroyed, and coanalysis of RNA sequencing and proteomics of foetal skeletal muscle showed that mitochondrial elements and lipid oxidation were consistently impaired. In vivo and in vitro myoblast experiments also demonstrated that high glucose concentrations impeded mitochondrial organisation and function. Importantly, the transcription of genes associated with mitochondrial biogenesis and oxidative metabolism decreased at 8 weeks and during the foetal period. We predicted Ppargc1α as a key upstream regulator with the help of IPA software. The proteins and mRNA levels of Ppargc1α in the skeletal muscle of GDM male offspring were decreased as a foetus (CTR vs. GDM, 1.004 vs. 0.665, p = 0.002), at 6 weeks (1.018 vs. 0.511, p = 0.023) and 8 weeks (1.006 vs. 0.596, p = 0.018). In addition, CREB phosphorylation was inhibited in GDM group, with fewer activated pCREB proteins binding to the CRE element of Ppargc1α (1.042 vs. 0.681, p = 0.037), Pck1 (1.091 vs. 0.432, p = 0.014) and G6pc (1.118 vs. 0.472, p = 0.027), resulting in their decreased transcription. Interestingly, we found that sarcopenia and mitochondrial dysfunction could even be inherited by the next generation. CONCLUSIONS: Short-term intrauterine hyperglycaemia significantly reduced lean mass in male offspring at 8 weeks, resulting in decreased exercise endurance and metabolic disorders. Disrupted organisation and function of the mitochondria in skeletal muscle were also observed among them. Foetal exposure to hyperglycaemia decreased the ratio of phosphorylated CREB and reduced the transcription of Ppargc1α, which inhibited the transcription of downstream genes involving in mitochondrial biogenesis and oxidative metabolism. Abnormal mitochondria, which might be transmitted through aberrant gametes, were also observed in the F2 generation.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico , Diabetes Gestacional , Hiperglucemia , Músculo Esquelético , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Efectos Tardíos de la Exposición Prenatal , Transducción de Señal , Animales , Femenino , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Embarazo , Ratones , Masculino , Músculo Esquelético/metabolismo , Diabetes Gestacional/metabolismo , Hiperglucemia/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Diabetes Mellitus Experimental/metabolismo , Mitocondrias/metabolismo , Glucemia/metabolismo
4.
Front Nutr ; 10: 1067282, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37255932

RESUMEN

Diabetes mellitus during pregnancy, which can be classified into pregestational diabetes and gestational diabetes, has become much more prevalent worldwide. Maternal diabetes fosters an intrauterine abnormal environment for fetus, which not only influences pregnancy outcomes, but also leads to fetal anomaly and development of diseases in later life, such as metabolic and cardiovascular diseases, neuropsychiatric outcomes, reproduction malformation, and immune dysfunction. The underlying mechanisms are comprehensive and ambiguous, which mainly focus on microbiota, inflammation, reactive oxygen species, cell viability, and epigenetics. This review concluded with the influence of intrauterine hyperglycemia on fetal structure development and organ function on later life and outlined potential mechanisms that underpin the development of diseases in adulthood. Maternal diabetes leaves an effect that continues generations after generations through gametes, thus more attention should be paid to the prevention and treatment of diabetes to rescue the pathological attacks of maternal diabetes from the offspring.

5.
Endocrine ; 75(2): 400-408, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34542801

RESUMEN

PURPOSE: Adulthood and childhood obesity are both associated with reproductive diseases and gynecological cancers in females. However, the causal factors associated with these observations have yet to be identified. Mendelian randomization is a process that is independent of inverse bias and confounding and can act as a random control trial in which genetic groups are settled during meiosis, thus representing an effective tool with which to investigate causality. METHODS: We carried out several Mendelian randomization trials based on the combined genetic scores of 75 adult-associated and 15 childhood-associated body mass index (BMI) single nucleotide polymorphisms (SNPs), databases for several gynecological cancers and reproductive diseases from the UK Biobank (with 194,153 participants), using the traditional inverse-variance weighted (IVW) method as the main method. RESULTS: Elevated adult-associated BMI scores (odds ratio [OR] = 1.003; 95% confidence interval [CI]: 1.001-1.004) and childhood-associated BMI scores (OR = 1.003; 95% CI: 1.001-1.004) were related to a higher risk of the polycystic ovarian syndrome (PCOS), as determined by the traditional IVW method. The random IVW method further revealed a nominal negative causal association between childhood-associated BMI and subsequent endometriosis (OR = 0.995; 95% CI: 0.991-0.999). CONCLUSIONS: Consistent with observational consequences, our findings indicated that adulthood obesity may play role in the development of PCOS and that childhood obesity can increase the risk of PCOS but may reduce the incidence of endometriosis in later life. Further research is now needed to validate our findings and identify the precise mechanisms involved.


Asunto(s)
Análisis de la Aleatorización Mendeliana , Obesidad Infantil , Adulto , Índice de Masa Corporal , Niño , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Obesidad Infantil/epidemiología , Obesidad Infantil/genética , Polimorfismo de Nucleótido Simple
6.
Front Cell Dev Biol ; 10: 748862, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35237591

RESUMEN

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.

7.
Front Endocrinol (Lausanne) ; 12: 683226, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34248845

RESUMEN

Objective: Sex steroids are thought to contribute to the pathogenesis of osteoarthritis (OA). This study investigated the causal role of sex steroids in site- and sex-specific OA and risk of joint replacement surgery using the Mendelian randomization (MR) method. Methods: Instrumental variables for estradiol, dehydroepiandrosterone sulfate, testosterone (T), and dihydrotestosterone (DHT) were selected. We used the inverse variance weighting (IVW) approach as the main MR method to estimate causal effects based on the summary-level data for OA and joint replacement surgery from genome-wide association studies (GWAS). Results: A positive causal association was observed between serum T level and risks of hip OA (odds ratio [OR]=1.558, 95% confidence interval [CI]: 1.193-2.034; P=0.001) and hip replacement (OR=1.013, 95% CI: 1.008-1.018; P=2.15×10-8). Serum DHT level was also positively associated with the risk of hip replacement (OR=1.011, 95% CI: 1.006-1.015; P=4.03×10-7) and had potential causality with hip OA (OR=1.398, 95% CI: 1.054-1.855; P=0.020). Conclusions: Serum T and DHT levels may play causal roles in the development of hip OA and contribute to the risk of hip replacement, although the underlying mechanisms require further investigation.


Asunto(s)
Hormonas Esteroides Gonadales/sangre , Osteoartritis de la Cadera , Osteoartritis de la Rodilla , Artroplastia de Reemplazo de Cadera , Artroplastia de Reemplazo de Rodilla , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Análisis de la Aleatorización Mendeliana , Osteoartritis de la Cadera/sangre , Osteoartritis de la Cadera/genética , Osteoartritis de la Cadera/cirugía , Osteoartritis de la Rodilla/sangre , Osteoartritis de la Rodilla/genética , Osteoartritis de la Rodilla/cirugía , Polimorfismo de Nucleótido Simple
8.
J Clin Endocrinol Metab ; 106(6): e2434-e2443, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33581692

RESUMEN

PURPOSE: Multiple risk factors have been implicated in the development of osteoporosis. This study examined potential associations between serum nutritional factors and bone mineral density (BMD). METHODS: Six nutritional factors were selected as exposures. Outcomes included total body BMD (n = 66 945); BMD at the forearm (FA), femoral neck (FN) and lumbar spine (LS) (n = 8143, n = 32 735, and n = 28 498, respectively); estimated heel BMD (HL eBMD) (n = 394 929); and HL eBMD stratified by sex (n = 206 496). A 2-sample Mendelian randomization approach was adopted to estimate the association between serum nutritional factors and BMD. The threshold for adjusted P value was 1.39 × 10-3. RESULTS: Serum calcium levels were inversely associated with LS BMD (effect = -0.55; 95% CI, -0.86 to -0.24; P = 0.001), whereas serum selenium levels were positively correlated with HL eBMD (effect = 0.22; 95% CI, 0.10 to 0.33; P = 1.70 × 10-4). Regarding nominal significance, there was a positive association between serum selenium levels and FA BMD. Nominally significant results were also obtained for serum retinol as well as vitamin E levels and HL eBMD. Moreover, sex-specific effects of serum retinol and vitamin E levels on BMD were observed in men. CONCLUSION: Serum calcium and selenium levels influence BMD at specific skeletal sites. This implies that these nutritional factors play crucial roles in bone metabolism.


Asunto(s)
Densidad Ósea/genética , Nutrientes/sangre , Calcio/sangre , Femenino , Interacción Gen-Ambiente , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Análisis de la Aleatorización Mendeliana , Herencia Multifactorial , Osteoporosis/sangre , Osteoporosis/epidemiología , Osteoporosis/etiología , Osteoporosis/genética , Polimorfismo de Nucleótido Simple , Factores de Riesgo , Población Blanca/genética
9.
Front Endocrinol (Lausanne) ; 12: 710221, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34531826

RESUMEN

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.


Asunto(s)
Biomarcadores/metabolismo , Diabetes Gestacional/fisiopatología , Feto/metabolismo , Intolerancia a la Glucosa/patología , Hiperglucemia/patología , Metaboloma , Útero/fisiopatología , Animales , Epigénesis Genética , Femenino , Feto/patología , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Hiperglucemia/genética , Hiperglucemia/metabolismo , Masculino , Páncreas/metabolismo , Páncreas/patología , Embarazo , Factores Sexuales
10.
Mol Cell Endocrinol ; 529: 111264, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33811969

RESUMEN

Over the past four decades, the global prevalence of obesity has increased rapidly in all age ranges. Emerging evidence suggests that paternal lifestyle and environmental exposure have a crucial role in the health of offspring. Therefore, the current study investigated the impact of paternal obesity on the metabolic profile of offspring in a male mouse model of obesity. Female offspring of obese fathers fed a high-fat diet (HFD) (60% kcal fat) showed hyperglycemia because of enhanced gluconeogenesis and elevated expression of phosphoenolpyruvate carboxykinase (PEPCK), which is a key enzyme involved in the regulation of gluconeogenesis. Methylation of the Igf2/H19 imprinting control region (ICR) was dysregulated in the liver of offspring, and the sperm, of HFD fathers, suggesting that epigenetic changes in germ cells contribute to this father-offspring transmission. In addition, we explored whether H19 might regulate hepatic gluconeogenesis. Our results showed that overexpression of H19 in Hepa1-6 cells enhanced the expression of PEPCK and gluconeogenesis by promoting nuclear retention of forkhead box O1 (FOXO1), which is involved in the transcriptional regulation of Pepck. Thus, the current study suggests that paternal exposure to HFD impairs the gluconeogenesis of offspring via altered Igf2/H19 DNA methylation.


Asunto(s)
Epigénesis Genética , Hiperglucemia/genética , Factor II del Crecimiento Similar a la Insulina/genética , Obesidad/genética , Fosfoenolpiruvato Carboxiquinasa (ATP)/genética , ARN Largo no Codificante/genética , Animales , Línea Celular , Metilación de ADN , Dieta Alta en Grasa/efectos adversos , Femenino , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Impresión Genómica , Gluconeogénesis/genética , Hepatocitos/metabolismo , Hepatocitos/patología , Hiperglucemia/etiología , Hiperglucemia/metabolismo , Hiperglucemia/patología , Patrón de Herencia , Factor II del Crecimiento Similar a la Insulina/metabolismo , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/metabolismo , Obesidad/patología , Fosfoenolpiruvato Carboxiquinasa (ATP)/metabolismo , Procesamiento Proteico-Postraduccional , ARN Largo no Codificante/metabolismo , Espermatozoides/metabolismo
11.
Diabetes ; 68(4): 696-708, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30696708

RESUMEN

Gestational diabetes mellitus (GDM) is associated with an increased risk of metabolic disorders in offspring in later life. Although mounting evidence suggests that therapy for GDM could improve neonatal health, whether the therapy confers long-term metabolic benefits to offspring in their later adult lives is not known. Here, using a mouse model of diabetes in the latter half of pregnancy to mimic human GDM, we find that the efficient insulin therapy for GDM confers significant protection against glucose intolerance and obesity in offspring fed a normal chow diet. However, the therapy fails to protect offspring when challenged with a high-fat diet, especially for male offspring. Genome-wide DNA methylation profiling of pancreatic islets from male offspring identified hypermethylated regions in several genes that regulate insulin secretion, including Abcc8, Cav1.2, and Cav2.3 that encode KATP or Ca2+ channels, which are associated with reduced gene expression and impaired insulin secretion. This finding suggests a methylation-mediated epigenetic mechanism for GDM-induced intergenerational glucose intolerance. It highlights that even efficient insulin therapy for GDM is insufficient to fully protect adult offspring from diet-induced metabolic disorders.


Asunto(s)
Diabetes Gestacional/tratamiento farmacológico , Epigénesis Genética , Hipoglucemiantes/uso terapéutico , Insulina/uso terapéutico , Obesidad/etiología , Animales , Metilación de ADN , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Femenino , Ratones , Obesidad/prevención & control , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA