Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.739
Filtrar
1.
Cell Host Microbe ; 32(7): 1048-1049, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38991502

RESUMEN

Gestational diabetes mellitus (GDM) is associated with increased risk of metabolic and neurodevelopmental disorders in offspring. In this issue of Cell Host & Microbe, Wang et al. provide evidence that changes in the gut microbiome of mothers with GDM may lead to dysbiosis in their infants and altered development in a sex-dependent manner.


Asunto(s)
Diabetes Gestacional , Disbiosis , Microbioma Gastrointestinal , Diabetes Gestacional/microbiología , Diabetes Gestacional/metabolismo , Embarazo , Microbioma Gastrointestinal/fisiología , Humanos , Femenino , Disbiosis/microbiología , Lactante , Masculino , Recién Nacido
2.
Nutr Diabetes ; 14(1): 56, 2024 Jul 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
3.
Front Endocrinol (Lausanne) ; 15: 1415630, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38938519

RESUMEN

Objective: Endocannabinoids and their N-acyl-ethanolamines (NAEs) and 2monoacyl-glycerols (2-MAGs) congeners are involved in the central and peripheral regulation of energy homeostasis, they are present in human milk and are associated with obesity. Infants exposed in utero to gestational diabetes mellitus (GDM) are more likely to develop obesity. The objective of this cross-sectional study is to compare the profile of eCBome mediators in milk of women with gestational diabetes (GDM+) and without (GDM-) and to assess the association with offspring growth. The hypothesis is that the eCBome of GDM+ human milk is altered and associated with a difference in infant growth. Methods: Circulating eCBome mediators were measured by LC-MS/MS in human milk obtained at 2 months postpartum from GDM+ (n=24) and GDM- (n=29) women. Infant weight and height at 2 months were obtained from the child health record. Z-scores were calculated. Results: Circulating Npalmitoylethanolamine (PEA) was higher in human milk of GDM+ women than in GDM- women (4.9 ± 3.2 vs. 3.3 ± 1.7, p=0.04). Higher levels were also found for several 2monoacyl-glycerols (2-MAGs) (p<0.05). The levels of NAEs (ß=-4.6, p=0.04) and especially non-omega-3 NAEs (B=-5.6, p=0.004) in human milk were negatively correlated with weight-for-age z-score of GDM+ offspring. Conclusion: The profile of eCBome mediators in human milk at 2 months postpartum was different in GDM+ compared to GDM- women and was associated with GDM+ offspring growth at 2 months. Clinical trial registration: ClinicalTrials.gov, identifier (NCT04263675 and NCT02872402).


Asunto(s)
Diabetes Gestacional , Endocannabinoides , Leche Humana , Adulto , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Embarazo , Desarrollo Infantil/fisiología , Estudios Transversales , Diabetes Gestacional/metabolismo , Diabetes Gestacional/sangre , Endocannabinoides/sangre , Endocannabinoides/metabolismo , Leche Humana/química , Leche Humana/metabolismo
4.
Clin Lab ; 70(6)2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38868866

RESUMEN

BACKGROUND: The goal was to analyze serums of GDM patients and healthy pregnant women using HPLC-MS and preliminarily screen differential metabolites by metabolomics. METHOD: Sixty pregnant women who underwent elective cesarean section at term in Dongguan Dalang Hospital from January 2023 to April 2023 were selected and divided into the GDM group and healthy pregnancy group. Pre-pregnancy and pregnancy examination information, such as age, BMI, OGTT results, triglyceride, total cholesterol, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, and other clinical data were col-lected for statistical analysis. Non-targeted metabolomics of serum from 30 GDM patients and 30 healthy pregnant women were studied by HPLC-MS, and different ions were searched. The structures of differential metabolites were identified by HMDB database. The metabolic pathways of differential metabolites were analyzed by KEGG database. RESULTS: The OGTT result, pCO2, pO2, HCO3, BE, Apgar score, and bilirubin levels in the GDM group were higher than those in the healthy pregnancy group (p < 0.05). However, there were no significant differences in age, triglyceride, total cholesterol, newborn birth weight, newborn birth blood glucose, and blood gas pH between the two groups (all p > 0.05). Using p < 0.05 as the screening standard, 55 differential metabolites were identified in serum, mainly including fatty acyl, carboxylic acids and their derivatives, steroids and their derivatives, ketoacids and their derivatives, and pyrimidine nucleosides, etc., all of which were up-regulated or down-regulated to varying degrees. The 55 metabolites were mainly involved in the metabolism of pyrimidine, pyruvate, alanine, aspartic acid, glutamic acid, and arachidonic acid, glycolysis, and biosynthesis of unsaturated fatty acids. CONCLUSIONS: The discovery of these metabolites provides a theoretical basis for an indepth understanding of GDM pathogenesis. Non-targeted metabonomics analysis of blood metabonomics research technology has shown great potential value in the early diagnosis of obstetric diseases and the study of disease mechanisms.


Asunto(s)
Diabetes Gestacional , Metabolómica , Humanos , Femenino , Diabetes Gestacional/sangre , Diabetes Gestacional/diagnóstico , Diabetes Gestacional/metabolismo , Embarazo , Metabolómica/métodos , Adulto , Recién Nacido , Estudios de Casos y Controles , Cromatografía Líquida de Alta Presión/métodos , Biomarcadores/sangre
5.
Front Endocrinol (Lausanne) ; 15: 1375771, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38883605

RESUMEN

Introduction: The incidence of infertility is significantly higher in women with diseases linked to impaired glucose homeostasis, such as insulin resistance. Defective glucose metabolism interferes with fertilization; however, the molecular mechanism underlying this interference is unclear. Smoothelin-like protein 1 (SMTNL1) was isolated from muscle and steroid hormone-responsive tissues and regulates the contractile functions of various cell types through the inhibition of myosin phosphatase (MP) holoenzyme. In addition, SMTNL-1 after phosphorylation at Ser301 by protein kinase A translocates to the nucleus and functions as a transcriptional co-activator of the progesterone receptor-B. SMTNL1 null mice exhibit reduced reproductive fitness and are more prone to type 2 diabetes mellitus. However, the role of SMTNL1 in endometrial epithelial cells is not known. Methods: The effect of SMTNL1 overexpression was investigated in pregnancy and in gestational diabetic endometrial epithelial cell models by immunofluorescent staining, cell migration, and semi quantitative Western blot analysis and glucose uptake assay. Results: We show that SMTNL1 promotes the differentiation of endometrial epithelial cells in a progesterone-dependent manner to attenuate insulin resistance. Furthermore, SMTNL1 hampers the migration capacity of epithelial cells in a gestational diabetes model by inhibiting the expression of MYPT1, the regulatory subunit of MP, and the activity of the holoenzyme, resulting in increased phosphorylation of the 20 kDa regulatory myosin light chain. SMTNL1 also acts as an insulin-sensitizing agent by increasing the gene expression of PP2A and DUPS9 protein phosphatases, resulting in decreased ERK1/2 activity and, hence, decreasing the phosphorylation of IRS-1 at Ser612 under gestational diabetes conditions. Conclusion: SMTNL1 may have therapeutic relevance to the progesterone-dependent inhibition of endometrial epithelial cell migration under hyperglycemic conditions and insulin sensitivity in the endometrium in gestational diabetes or other metabolic disorders.


Asunto(s)
Endometrio , Células Epiteliales , Resistencia a la Insulina , Proteínas Musculares , Femenino , Endometrio/metabolismo , Humanos , Células Epiteliales/metabolismo , Proteínas Musculares/metabolismo , Proteínas Musculares/genética , Embarazo , Animales , Diabetes Gestacional/metabolismo , Ratones , Fosforilación , Movimiento Celular , Péptidos y Proteínas de Señalización Intracelular
6.
Sci Rep ; 14(1): 14621, 2024 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-38918525

RESUMEN

Gestational diabetes mellitus (GDM) is associated with increased postpartum risk for metabolic dysfunction-associated steatotic liver disease (MASLD). GDM-related MASLD predisposes to advanced liver disease, necessitating a better understanding of its development in GDM. This preclinical study evaluated the MASLD development in a lean GDM mouse model with impaired insulin secretion capacity. Lean GDM was induced by short-term 60% high-fat diet and low-dose streptozotocin injections (60 mg/kg for 3 days) before mating in C57BL/6N mice. The control dams received only high-fat diet or low-fat diet. Glucose homeostasis was assessed during pregnancy and postpartum, whereas MASLD was assessed on postpartum day 30 (PP30). GDM dams exhibited a transient hyperglycemic phenotype during pregnancy, with hyperglycaemia reappearing after lactation. Lower insulin levels and impaired glucose-induced insulin response were observed in GDM mice during pregnancy and postpartum. At PP30, GDM dams displayed higher hepatic triglyceride content compared controls, along with increased MAS (MASLD) activity scores, indicating lipid accumulation, inflammation, and cell turnover indices. Additionally, at PP30, GDM dams showed elevated plasma liver injury markers. Given the absence of obesity in this double-hit GDM model, the results clearly indicate that impaired insulin secretion driven pregnancy hyperglycaemia has a distinct contribution to the development of postpartum MASLD.


Asunto(s)
Diabetes Gestacional , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Periodo Posparto , Animales , Diabetes Gestacional/metabolismo , Embarazo , Femenino , Ratones , Periodo Posparto/metabolismo , Hígado Graso/metabolismo , Hígado Graso/patología , Hígado Graso/etiología , Insulina/metabolismo , Insulina/sangre , Dieta Alta en Grasa/efectos adversos , Hígado/metabolismo , Hígado/patología , Glucemia/metabolismo , Triglicéridos/metabolismo , Triglicéridos/sangre
7.
Int J Mol Sci ; 25(11)2024 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-38892323

RESUMEN

The placenta plays a key role in several adverse obstetrical outcomes, such as preeclampsia, intrauterine growth restriction and gestational diabetes mellitus. The early identification of at-risk pregnancies could significantly improve the management, therapy and prognosis of these pregnancies, especially if these at-risk pregnancies are identified in the first trimester. The aim of this review was to summarize the possible biomarkers that can be used to diagnose early placental dysfunction and, consequently, at-risk pregnancies. We divided the biomarkers into proteins and non-proteins. Among the protein biomarkers, some are already used in clinical practice, such as the sFLT1/PLGF ratio or PAPP-A; others are not yet validated, such as HTRA1, Gal-3 and CD93. In the literature, many studies analyzed the role of several protein biomarkers, but their results are contrasting. On the other hand, some non-protein biomarkers, such as miR-125b, miR-518b and miR-628-3p, seem to be linked to an increased risk of complicated pregnancy. Thus, a first trimester heterogeneous biomarkers panel containing protein and non-protein biomarkers may be more appropriate to identify and discriminate several complications that can affect pregnancies.


Asunto(s)
Biomarcadores , Placenta , Resultado del Embarazo , Primer Trimestre del Embarazo , Humanos , Embarazo , Femenino , Primer Trimestre del Embarazo/metabolismo , Placenta/metabolismo , Preeclampsia/diagnóstico , Preeclampsia/metabolismo , MicroARNs/genética , Proteína Plasmática A Asociada al Embarazo/metabolismo , Diabetes Gestacional/diagnóstico , Diabetes Gestacional/metabolismo
8.
BMC Microbiol ; 24(1): 161, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38730357

RESUMEN

Gestational diabetes mellitus (GDM) is characterized by insulin resistance and low-grade inflammation, and most studies have demonstrated gut dysbiosis in GDM pregnancies. Overall, they were manifested as a reduction in microbiome diversity and richness, depleted short chain fatty acid (SCFA)-producing genera and a dominant of Gram-negative pathogens releasing lipopolysaccharide (LPS). The SCFAs functioned as energy substance or signaling molecules to interact with host locally and beyond the gut. LPS contributed to pathophysiology of diseases through activating Toll-like receptor 4 (TLR4) and involved in inflammatory responses. The gut microbiome dysbiosis was not only closely related with GDM, it was also vital to fetal health through vertical transmission. In this review, we summarized gut microbiota signature in GDM pregnancies of each trimester, and presented a brief introduction of microbiome derived SCFAs. We then discussed mechanisms of microbiome-host interactions in the physiopathology of GDM and associated metabolic disorders. Finally, we compared offspring microbiota composition from GDM with that from normal pregnancies, and described the possible mechanism.


Asunto(s)
Diabetes Gestacional , Disbiosis , Ácidos Grasos Volátiles , Microbioma Gastrointestinal , Diabetes Gestacional/microbiología , Diabetes Gestacional/metabolismo , Humanos , Embarazo , Femenino , Disbiosis/microbiología , Ácidos Grasos Volátiles/metabolismo , Bacterias/clasificación , Bacterias/genética , Bacterias/metabolismo , Bacterias/aislamiento & purificación , Interacciones Microbiota-Huesped , Lipopolisacáridos/metabolismo
9.
Environ Sci Technol ; 58(20): 8675-8684, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38728584

RESUMEN

Pregnant women are physiologically prone to glucose intolerance, while the puerperium represents a critical phase for recovery. However, how air pollution disrupts glucose homeostasis during the gestational and early postpartum periods remains unclear. This prospective cohort study conducted an oral glucose tolerance test and measured the insulin levels of 834 pregnant women in Guangzhou, with a follow-up for 443 puerperae at 6-8 weeks postpartum. Residential PM2.5 and five chemical components were estimated by an established spatiotemporal model. The adjusted linear model showed that an IQR increase in gestational PM2.5 exposure was associated with an increase of 0.17 mmol/L (95% CI: 0.06, 0.28) in fasting plasma glucose (FPG) and 0.24 (95% CI: 0.05, 0.42) in the insulin resistance index. Postpartum PM2.5 exposure was linked to a 0.17 mmol/L (95% CI: 0.05, 0.28) elevation in FPG per IQR, with a strengthened association found in women with gestational diabetes (Pinteraction = 0.003). In the quantile-based g-computation model, NO3- consistently contributed to the combined effect of PM2.5 components on gestational and postpartum FPG. This study was the first to suggest that PM2.5 components were associated with exacerbated gestational insulin resistance and elevated postpartum FPG. Targeted interventions reducing the emissions of toxic PM2.5 components are essential to improving maternal glucose metabolism.


Asunto(s)
Material Particulado , Periodo Posparto , Humanos , Femenino , Estudios Prospectivos , Embarazo , Adulto , China , Glucemia , Glucosa/metabolismo , Diabetes Gestacional/metabolismo , Contaminación del Aire , Resistencia a la Insulina , Contaminantes Atmosféricos , Estudios de Cohortes , Pueblos del Este de Asia
10.
Reprod Toxicol ; 1232024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38706688

RESUMEN

Exposure to gestational diabetes mellitus (GDM) during pregnancy has significant consequences for the unborn baby and newborn infant. However, whether and how GDM exposure induces the development of neonatal brain hypoxia/ischemia-sensitive phenotype and the underlying molecular mechanisms remain unclear. In this study, we used a late GDM rat model induced by administration of streptozotocin (STZ) on gestational day 12 and investigated its effects of GDM on neonatal brain development. The pregnant rats exhibited increased blood glucose levels in a dose-dependent manner after STZ administration. STZ-induced maternal hyperglycemia led to reduced blood glucose levels in neonatal offspring, resulting in growth restriction and an increased brain to body weight ratio. Importantly, GDM exposure increased susceptibility to hypoxia/ischemia (HI)-induced brain infarct sizes compared to the controls in both male and female neonatal offspring. Further molecular analysis revealed alterations in the PTEN/AKT/mTOR/autophagy signaling pathway in neonatal male offspring brains, along with increased ROS production and autophagy-related proteins (Atg5 and LC3-II). Treatment with the PTEN inhibitor bisperoxovanadate (BPV) eliminated the differences in HI-induced brain infarct sizes between the GDM-exposed and the control groups. These findings provide novel evidence of the development of a brain hypoxia/ischemia-sensitive phenotype in response to GDM exposure and highlight the role of the PTEN/AKT/mTOR/autophagy signaling pathway in this process.


Asunto(s)
Autofagia , Encéfalo , Diabetes Gestacional , Hipoxia-Isquemia Encefálica , Transducción de Señal , Estreptozocina , Animales , Femenino , Masculino , Embarazo , Ratas , Animales Recién Nacidos , Autofagia/efectos de los fármacos , Glucemia , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/patología , Diabetes Gestacional/inducido químicamente , Diabetes Gestacional/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Efectos Tardíos de la Exposición Prenatal , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfohidrolasa PTEN/metabolismo , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
11.
Sci Rep ; 14(1): 12051, 2024 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-38802412

RESUMEN

GDM, as a metabolic disease during pregnancy, regulates GLUT3 translocation by AMPK, thereby affecting glucose uptake in trophoblasts. It provides a new research idea and therapeutic target for alleviating intrauterine hyperglycemia in GDM. STZ was used to construct GDM mice, inject AICAR into pregnant mice, and observe fetal and placental weight; flow cytometry was employed for the detection of glucose uptake by primary trophoblast cells; immunofluorescence was applied to detect the localization of GLUT3 and AMPK in placental tissue; Cocofal microscope was used to detect the localization of GLUT3 in trophoblast cells;qRT-PCR and Western blot experiments were carried out to detect the expression levels of GLUT3 and AMPK in placental tissue; CO-IP was utilized to detect the interaction of GLUT3 and AMPK. Compared with the normal pregnancy group, the weight of the fetus and placenta of GDM mice increased (P < 0.001), and the ability of trophoblasts to take up glucose decreased (P < 0.001). In addition, AMPK activity in trophoblasts and membrane localization of GLUT3 in GDM mice were down-regulated compared with normal pregnant mice (P < 0.05). There is an interaction between GLUT3 and AMPK. Activating AMPK in trophoblasts can up-regulate the expression of GLUT3 membrane protein in trophoblasts of mice (P < 0.05) and increase the glucose uptake of trophoblasts (P < 0.05). We speculate that inhibition of AMPK activity in GDM mice results in aberrant localization of GLUT3, which in turn attenuates glucose uptake by placental trophoblast cells. AICAR activates AMPK to increase the membrane localization of GLUT3 and improve the glucose uptake capacity of trophoblasts.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Diabetes Gestacional , Transportador de Glucosa de Tipo 3 , Glucosa , Transducción de Señal , Trofoblastos , Animales , Trofoblastos/metabolismo , Femenino , Embarazo , Glucosa/metabolismo , Ratones , Proteínas Quinasas Activadas por AMP/metabolismo , Transportador de Glucosa de Tipo 3/metabolismo , Transportador de Glucosa de Tipo 3/genética , Diabetes Gestacional/metabolismo , Placenta/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Ribonucleótidos/farmacología
12.
J Proteome Res ; 23(6): 1937-1947, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38776154

RESUMEN

Lactylation is a novel post-translational modification of proteins. Although the histone lactylation modification has been reported to be involved in glucose metabolism, its role and molecular pathways in gestational diabetes mellitus (GDM) are still unclear. This study aims to elucidate the histone lactylation modification landscapes of GDM patients and explore lactylation-modification-related genes involved in GDM. We employed a combination of RNA-seq analysis and chromatin immunoprecipitation sequencing (ChIP-seq) analysis to identify upregulated differentially expressed genes (DEGs) with hyperhistone lactylation modification in GDM. We demonstrated that the levels of lactate and histone lactylation were significantly elevated in GDM patients. DEGs were involved in diabetes-related pathways, such as the PI3K-Akt signaling pathway, Jak-STAT signaling pathway, and mTOR signaling pathway. ChIP-seq analysis indicated that histone lactylation modification in the promoter regions of the GDM group was significantly changed. By integrating the results of RNA-seq and ChIP-seq analysis, we found that CACNA2D1 is a key gene for histone lactylation modification and is involved in the progression of GDM by promoting cell vitality and proliferation. In conclusion, we identified the key gene CACNA2D1, which upregulated and exhibited hypermodification of histone lactylation in GDM. These findings establish a theoretical groundwork for the targeted therapy of GDM.


Asunto(s)
Secuenciación de Inmunoprecipitación de Cromatina , Diabetes Gestacional , Histonas , Procesamiento Proteico-Postraduccional , Diabetes Gestacional/genética , Diabetes Gestacional/metabolismo , Humanos , Femenino , Embarazo , Histonas/metabolismo , Histonas/genética , Transducción de Señal/genética , RNA-Seq , Adulto
13.
Biomed Pharmacother ; 175: 116775, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38776680

RESUMEN

Gestational diabetes mellitus (GDM) is a pregnancy-specific disease characterized by impaired glucose tolerance during pregnancy. Although diagnosis and clinical management have improved significantly, there are still areas where therapeutic approaches need further improvement. Recent evidence suggests that CCL2, a chemokine involved in immunoregulatory and inflammatory processes, is closely related to GDM. However, the potential value for clinical therapeutic applications and the mechanism of CCL2 in adipose tissue macrophages (ATMs) of GDM remain to be elucidated. Here, we found that CCL2 was enriched in macrophages of the visceral adipose tissue from GDM women and HFD-induced GDM mice. The combination of in vitro and in vivo experiments showed that Ccl2 silencing inhibited the inflammatory response of macrophage by blocking calcium transport between ER and mitochondria and reducing excessive ROS generation. Additionally, the ATS-9R/siCcl2 oligopeptide complex targeting adipose tissue was created. Under the delivery of ATS-9R peptide, Ccl2 siRNA is expressed in ATMs, which reduces inflammation in adipose tissue and, as a result, mitigates insulin resistance. All of these findings point to the possibility that the ATS-9R/siCcl2 complex, which targets adipose tissue, is able to reduce insulin resistance in GDM and the inflammatory response in macrophages. The ATS-9R/siCcl2 oligopeptide complex targeting adipose tissue seems to be a viable treatment for GDM pregnancies.


Asunto(s)
Tejido Adiposo , Quimiocina CCL2 , Diabetes Gestacional , Resistencia a la Insulina , Macrófagos , Ratones Endogámicos C57BL , Oligopéptidos , Animales , Diabetes Gestacional/metabolismo , Diabetes Gestacional/tratamiento farmacológico , Femenino , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Embarazo , Quimiocina CCL2/metabolismo , Ratones , Humanos , Oligopéptidos/farmacología , Tejido Adiposo/metabolismo , Adulto , Dieta Alta en Grasa , Grasa Intraabdominal/metabolismo
14.
J Obstet Gynaecol ; 44(1): 2350761, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38785148

RESUMEN

BACKGROUND: Asiaticoside (AS) has been reported to improve the changes induced by high glucose stimulation, and it may have potential therapeutic effects on gestational diabetes mellitus (GDM). This study aims to explore the effect of AS on the cell model of GDM and the action mechanism of the PI3K/AKT pathway. METHODS: The GDM model was established in HTR-8/Svneo cells with a high glucose (HG) medium. After the cytotoxicity assay of AS, cells were divided into the control group, HG group and HG + AS group to conduct control experiment in cells. The cell proliferation and migration were detected by CCK-8 assay and scratch test, respectively. The mRNA levels of PI3K, AKT2, mTORC1, and GLUT4 in PI3K/AKT signalling pathway were measured by RT-PCR, and the protein expressions of these signalling molecules were monitored by western blot. RESULTS: AS showed a promotion effect on the cell proliferation rate of HTR-8/Svneo cells, and 80 µmol/L AS with a treatment time of 48 h had no cytotoxicity. The cell proliferation rate, migration rate, mRNA levels and protein expressions of PI3K, AKT2, mTORC1, and GLUT4 in the HG group were significantly lower than those in the control group, which were significantly increased in the HG + AS group (p < 0.05). CONCLUSIONS: AS can facilitate the cell proliferation and migration in the cell model of GDM, and might play a role in GDM treatment via PI3K/AKT pathway.


Asiaticoside possesses various pharmacological effects and has been reported to show a beneficial effect on the treatment of diabetes mellitus. This research firstly investigated the effect and mechanism of asiaticoside on gestational diabetes mellitus, and found that asiaticoside could facilitate the cell proliferation and migration of HTR-8/Svneo cells treated with high glucose, and affect the signalling molecules of PI3K/AKT pathway. Therefore, asiaticoside may be a novel useful therapeutic drug in the treatment of gestational diabetes mellitus.


Asunto(s)
Movimiento Celular , Proliferación Celular , Diabetes Gestacional , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Triterpenos , Humanos , Diabetes Gestacional/metabolismo , Femenino , Embarazo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proliferación Celular/efectos de los fármacos , Triterpenos/farmacología , Transducción de Señal/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Movimiento Celular/efectos de los fármacos , Línea Celular , Trofoblastos/efectos de los fármacos , Trofoblastos/metabolismo , Glucosa/farmacología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo
15.
Placenta ; 151: 27-36, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38701658

RESUMEN

INTRODUCTION: This study aimed to screen circRNAs involved in gestational diabetes mellitus (GDM)-related macrosomia. One differentially expressed circRNA (DEC), hsa_circ_0024838, was further tested for its potential role and mechanism in trophoblasts. METHODS: DECs in GDM were selected through GSE182737 and GSE194119. The targets were predicted for DECs and microRNAs (miRNAs), to complete the construction of the circRNA-miRNA-gene network. Functional annotation and related biological pathway enrichment analysis were performed on the target genes of miRNAs in the network. Subsequently, the expression levels of hsa_circ_0024838, miR-543, and HIF1A mRNA were identified by real-time quantitative real-time PCR (RT-qPCR) in GDM patients. Trophoblast activity was assessed via CCK-8 assay, apoptosis assay, and Matrigel invasion assay. Finally, interactions between miR-543 and either hsa_circ_0024838 or HIF1A were confirmed using dual-luciferase reporter assays. RESULTS: A GDM-related circRNA-miRNA-genes interaction network was constructed, consisting of 35 circRNAs, 46 miRNAs, and 122 target genes. Functional enrichment revealed that the enriched pathways were involved in GDM. Hsa_circ_0024838 and HIF1A mRNA expression levels were upregulated in GDM, while miR-543 expression levels were downregulated. A significant positive correlation between hsa_circ_0024838 and newborn weight was observed. Both hsa_circ_0024838 and HIF1A possessed binding sites for miR-543. Overexpressing hsa_circ_0024838 in high-glucose (HG)-cultured trophoblasts can partially reverse HG-induced reduction in trophoblast cell proliferation/migration and increase apoptosis. But this reversal can be negated by co-transfection with miR-543 mimics. The effects of miR-543 can be counteracted by HIF1A. DISCUSSION: Hsa_circ_0024838 can regulate the expression of HIF1A by interacting with miR-543. This regulates the HIF1A signaling pathway and enhance vitality in trophoblast cells.


Asunto(s)
Diabetes Gestacional , Subunidad alfa del Factor 1 Inducible por Hipoxia , MicroARNs , ARN Circular , Trofoblastos , Humanos , Diabetes Gestacional/genética , Diabetes Gestacional/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Femenino , ARN Circular/genética , ARN Circular/metabolismo , Embarazo , Trofoblastos/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Adulto
16.
Adv Clin Chem ; 121: 172-269, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38797542

RESUMEN

Reproductive success consists of a sequential events chronology, starting with the ovum fertilization, implantation of the embryo, placentation, and cellular processes like proliferation, apoptosis, angiogenesis, endocrinology, or metabolic changes, which taken together finally conduct the birth of healthy offspring. Currently, many factors are known that affect the regulation and proper maintenance of pregnancy in humans, domestic animals, or rodents. Among the determinants of reproductive success should be distinguished: the maternal microenvironment, genes, and proteins as well as numerous pregnancy hormones that regulate the most important processes and ensure organism homeostasis. It is well known that white adipose tissue, as the largest endocrine gland in our body, participates in the synthesis and secretion of numerous hormones belonging to the adipokine family, which also may regulate the course of pregnancy. Unfortunately, overweight and obesity lead to the expansion of adipose tissue in the body, and its excess in both women and animals contributes to changes in the synthesis and release of adipokines, which in turn translates into dramatic changes during pregnancy, including those taking place in the organ that is crucial for the proper progress of pregnancy, i.e. the placenta. In this chapter, we are summarizing the current knowledge about levels of adipokines and their role in the placenta, taking into account the physiological and pathological conditions of pregnancy, e.g. gestational diabetes mellitus, preeclampsia, or intrauterine growth restriction in humans, domestic animals, and rodents.


Asunto(s)
Adipoquinas , Embarazo , Humanos , Adipoquinas/metabolismo , Femenino , Animales , Placenta/metabolismo , Diabetes Gestacional/metabolismo
17.
Diabetes Metab Res Rev ; 40(4): e3814, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38769695

RESUMEN

AIMS: This study aimed to evaluate the association between gestational diabetes mellitus (GDM) and circulating folate metabolites, folic acid (FA) intake, and the methylenetetrahydrofolate reductase (MTHFR) and methionine synthase reductase (MTRR) genotype. MATERIALS AND METHODS: A prospective pregnancy cohort study was conducted in Beijing, China, from 2022 to 2023. Circulating folate metabolites, including red blood cell (RBC) 5-methyltetrahydrofolate (5-MTHF), 5, 10-methylene-tetrahydrofolate (5,10-CH2-THF), 5- formyltetrahydrofolate (5-CHO-THF), and unmetabolised folic acid (UMFA), and plasma homocysteine (HCY), 5-MTHF, and methylmalonic acid (MMA), were determined at 6-17 weeks and 20-26 weeks of gestation. FA intake and the MTHFR and MTRR genotype were also examined. GDM was diagnosed between 24 and 28 weeks of pregnancy by a 75-g oral glucose tolerance test (OGTT). The association between the folate status and GDM was ascertained using multivariate generalised linear models, logistic regression models, and restricted cubic spline regression, adjusting for potential confounders. RESULTS: The study included 2032 pregnant women, of whom 392 (19.29%) developed GDM. UMFA above the 75th percentile (≥P75) [adjusted OR (aOR) (95% confidence interval [CI]) = 1.36 (1.01-1.84)], UMFA ≥ P90 [aOR (95% CI) = 1.82 (1.23-2.69)], and HCY ≥ P75 [aOR (95% CI) = 1.40 (1.04-1.88)] in early pregnancy, and RBC 5-MTHF [aOR (95% CI) = 1.48 (1.10-2.00)], RBC 5,10-CH2-THF [aOR (95% CI) = 1.55 (1.15-2.10)], and plasma 5-MTHF [aOR (95% CI) = 1.36 (1.00-1.86)] in mid-pregnancy ≥ P75 are associated with GDM. Higher UMFA levels in early pregnancy show positive associations with the 1-h and 2-h glucose levels during the OGTT, and higher HCY levels are associated with increased fasting glucose levels during the OGTT. In comparison, RBC 5- MTHF and 5,10-CH2-THF, and plasma 5- MTHF in mid-pregnancy are positively associated with the 1-h glucose level (p < 0.05). The MTHFR and MTRR genotype and FA intake are not associated with GDM. CONCLUSIONS: Elevated levels of UMFA and HCY during early pregnancy, along with elevated RBC 5-MTHF and 5,10-CH2-THF and plasma 5-MTHF during mid-pregnancy, are associated with GDM. These findings indicate distinct connections between different folate metabolites and the occurrence of GDM.


Asunto(s)
Diabetes Gestacional , Ácido Fólico , Metilenotetrahidrofolato Reductasa (NADPH2) , Humanos , Femenino , Diabetes Gestacional/sangre , Diabetes Gestacional/metabolismo , Embarazo , Ácido Fólico/sangre , Estudios Prospectivos , Adulto , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Biomarcadores/sangre , Estudios de Seguimiento , Ferredoxina-NADP Reductasa/genética , Genotipo , China/epidemiología , Pronóstico , Segundo Trimestre del Embarazo/sangre , Homocisteína/sangre , Homocisteína/metabolismo
18.
Front Endocrinol (Lausanne) ; 15: 1399694, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38694942

RESUMEN

Gestational diabetes mellitus (GDM) poses a significant global health concern, impacting both maternal and fetal well-being. Early detection and treatment are imperative to mitigate adverse outcomes during pregnancy. This review delves into the pivotal role of insulin function and the influence of genetic variants, including SLC30A8, CDKAL1, TCF7L2, IRS1, and GCK, in GDM development. These genetic variations affect beta-cell function and insulin activity in crucial tissues, such as muscle, disrupting glucose regulation during pregnancy. We propose a hypothesis that this variation may disrupt zinc transport, consequently impairing insulin production and secretion, thereby contributing to GDM onset. Furthermore, we discussed the involvement of inflammatory pathways, such as TNF-alpha and IL-6, in predisposing individuals to GDM. Genetic modulation of these pathways may exacerbate glucose metabolism dysregulation observed in GDM patients. We also discussed how GDM affects cardiovascular disease (CVD) through a direct correlation between pregnancy and cardiometabolic function, increasing atherosclerosis, decreased vascular function, dyslipidemia, and hypertension in women with GDM history. However, further research is imperative to unravel the intricate interplay between inflammatory pathways, genetics, and GDM. This understanding is pivotal for devising targeted gene therapies and pharmacological interventions to rectify genetic variations in SLC30A8, CDKAL1, TCF7L2, IRS1, GCK, and other pertinent genes. Ultimately, this review offers insights into the pathophysiological mechanisms of GDM, providing a foundation for developing strategies to mitigate its impact.


Asunto(s)
Diabetes Gestacional , Humanos , Diabetes Gestacional/genética , Diabetes Gestacional/metabolismo , Embarazo , Femenino , Inflamación/genética , Inflamación/metabolismo , Predisposición Genética a la Enfermedad
19.
Life Sci ; 350: 122744, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38810793

RESUMEN

AIMS: The prevalence of gestational diabetes mellitus (GDM) has spurred investigations into various interconnected factors, among which gut dysbiosis is notably prominent. Although gut dysbiosis is strongly associated with GDM, the specific role of the gut microbiome in the pathogenesis of GDM remains unknown. This study aims to explore the pathogenesis of GDM from gut microbiota. MATERIALS AND METHODS: In our study, we constructed two GDM mice models: one induced by a high-fat diet (HFD) and the other through fecal microbiota transplantation (FMT) from GDM patients. In vitro, we used a co-culture system of RAW264.7 and 3T3-L1 adipocytes. KEY FINDINGS: We induced a GDM-like state in pregnant mice by FMT from GDM patients, which was consistent with the HFD model. A potential mechanism identified involves the diminished abundance of SCFA-producing microbiota, which reduces SCFAs, particularly propionic acid and butyric acid. In vitro, butyric and propionic acids were observed to alleviate LPS-induced TLR4-NF-κB activation, thereby reducing inflammation levels and inhibiting adipose insulin resistance via the PI3K/AKT signaling pathway. This reduction appears to trigger the polarization of adipose tissue macrophages toward M1 and promote insulin resistance in adipose tissue. SIGNIFICANCE: Our study fills this knowledge gap by finding that alterations in gut microbiota have an independent impact on hyperglycemia and insulin resistance in the GDM state. In vivo and in vitro, gut dysbiosis is linked to adipose tissue inflammation and insulin resistance via the bacterial product SCFAs in the GDM state, providing new insights into the pathogenesis of GDM.


Asunto(s)
Tejido Adiposo , Diabetes Gestacional , Disbiosis , Ácidos Grasos Volátiles , Microbioma Gastrointestinal , Macrófagos , Animales , Diabetes Gestacional/metabolismo , Diabetes Gestacional/microbiología , Femenino , Disbiosis/metabolismo , Ratones , Embarazo , Macrófagos/metabolismo , Ácidos Grasos Volátiles/metabolismo , Tejido Adiposo/metabolismo , Humanos , Células RAW 264.7 , Resistencia a la Insulina , Trasplante de Microbiota Fecal , Dieta Alta en Grasa/efectos adversos , Ratones Endogámicos C57BL , Células 3T3-L1 , Modelos Animales de Enfermedad
20.
Nat Med ; 30(6): 1689-1695, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38627562

RESUMEN

Reduced insulin sensitivity (insulin resistance) is a hallmark of normal physiology in late pregnancy and also underlies gestational diabetes mellitus (GDM). We conducted transcriptomic profiling of 434 human placentas and identified a positive association between insulin-like growth factor binding protein 1 gene (IGFBP1) expression in the placenta and insulin sensitivity at ~26 weeks gestation. Circulating IGFBP1 protein levels rose over the course of pregnancy and declined postpartum, which, together with high gene expression levels in our placenta samples, suggests a placental or decidual source. Higher circulating IGFBP1 levels were associated with greater insulin sensitivity (lesser insulin resistance) at ~26 weeks gestation in the same cohort and in two additional pregnancy cohorts. In addition, low circulating IGFBP1 levels in early pregnancy predicted subsequent GDM diagnosis in two cohorts of pregnant women. These results implicate IGFBP1 in the glycemic physiology of pregnancy and suggest a role for placental IGFBP1 deficiency in GDM pathogenesis.


Asunto(s)
Diabetes Gestacional , Resistencia a la Insulina , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina , Placenta , Humanos , Embarazo , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/sangre , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Femenino , Diabetes Gestacional/metabolismo , Diabetes Gestacional/genética , Diabetes Gestacional/sangre , Placenta/metabolismo , Resistencia a la Insulina/genética , Adulto , Perfilación de la Expresión Génica , Estudios de Cohortes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...