Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.782
Filter
1.
BMC Med ; 22(1): 433, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39379948

ABSTRACT

BACKGROUND: Postpartum depression (PPD) constitutes a significant mental health disorder affecting almost one fifth of pregnancies globally. Despite extensive research, the precise etiological mechanisms underlying PPD remain elusive. However, several risk factors like genetic predisposition, hormonal fluctuations, and stress-related environmental and psychosocial triggers have been found to be implicated in its development. MAIN: Recently, an increased risk of PPD has been reported to be associated with gestational diabetes mellitus (GDM), which is characterized by the disruption of glucose metabolism, primarily attributed to the emergence of insulin resistance (IR). While IR during pregnancy seems to be an evolutionary adaptative mechanism to handle the profound metabolic alterations during pregnancy, its subsequent resolution following delivery necessitates a reconfiguration of the metabolic landscape in both peripheral tissues and the central nervous system (CNS). Considering the pivotal roles of energy metabolism, particularly glucose metabolism, in CNS functions, we propose a novel model that such pronounced changes in IR and the associated glucose metabolism seen postpartum might account for PPD development. This concept is based on the profound influences from insulin and glucose metabolism on brain functions, potentially via modulating neurotransmitter actions of dopamine and serotonin. Their sudden postpartum disruption is likely to be linked to mood changes, as observed in PPD. CONCLUSIONS: The detailed pathogenesis of PPD might be multifactorial and still remains to be fully elucidated. Nevertheless, our hypothesis might account in part for an additional etiological factor to PPD development. If our concept is validated, it can provide guidance for future PPD prevention, diagnosis, and intervention.


Subject(s)
Depression, Postpartum , Diabetes, Gestational , Insulin Resistance , Humans , Female , Depression, Postpartum/metabolism , Pregnancy , Insulin Resistance/physiology , Diabetes, Gestational/metabolism , Diabetes, Gestational/physiopathology
2.
Front Endocrinol (Lausanne) ; 15: 1456629, 2024.
Article in English | MEDLINE | ID: mdl-39377073

ABSTRACT

Fetal and neonatal development is a critical period for the establishment of the future metabolic health and disease risk of an individual. Both maternal undernutrition and overnutrition can result in abnormal fetal organ development resulting in inappropriate birth size, child and adult obesity, and increased risk of Type 2 diabetes and cardiovascular diseases. Inappropriate adaptive changes to the maternal pancreas, placental function, and the development of the fetal pancreas in response to nutritional stress during pregnancy are major contributors to a risk trajectory in the offspring. This interconnected maternal-placental-fetal metabolic axis is driven by endocrine signals in response to the availability of nutritional metabolites and can result in cellular stress and premature aging in fetal tissues and the inappropriate expression of key genes involved in metabolic control as a result of long-lasting epigenetic changes. Such changes result is insufficient pancreatic beta-cell mass and function, reduced insulin sensitivity in target tissues such as liver and white adipose and altered development of hypothalamic satiety centres and in basal glucocorticoid levels. Whilst interventions in the obese mother such as dieting and increased exercise, or treatment with insulin or metformin in mothers who develop gestational diabetes, can improve metabolic control and reduce the risk of a large-for-gestational age infant, their effectiveness in changing the adverse metabolic trajectory in the child is as yet unclear.


Subject(s)
Fetal Development , Pancreas , Humans , Pregnancy , Female , Pancreas/metabolism , Pancreas/embryology , Fetal Development/physiology , Maternal Nutritional Physiological Phenomena , Prenatal Exposure Delayed Effects/metabolism , Diet , Animals , Placenta/metabolism , Diabetes, Gestational/metabolism
3.
Sci Rep ; 14(1): 21398, 2024 09 13.
Article in English | MEDLINE | ID: mdl-39271793

ABSTRACT

Gestational diabetes mellitus (GDM) adversely affects offspring glucose homeostasis and risk of developing obesity. Here, we examined the association between glycemia in pregnant women with overweight or obesity without GDM and offspring metabolic health. Maternal fasting glucose concentrations and glucose 2-h after an oral glucose tolerance test (OGTT) were measured in 208 women with a pre-pregnancy body mass index (BMI) of 28-45 kg/m2 without GDM. Offspring outcomes were collected at birth, 3, and 5 years of age. Linear mixed models with time as fixed factor and subject ID as random effects were used for analysis. No associations were found between maternal fasting or 2-h glucose concentrations with offspring glucose and insulin concentrations from birth to 5 years of age. However, maternal fasting glucose in GW 28 and 36, and 2-h OGTT glucose in GW 28 were positively associated with C-peptide concentration at birth. Maternal fasting glucose concentrations in GW 28 and 36 were positively associated with weight-for-length, and maternal fasting glucose in GW 36 was associated with BMI z-score at birth. In summary, blood glucose in pregnant women with overweight or obesity is positively associated with offspring C-peptide concentration, weight-for-length, and BMI z-score at birth, even in the absence of GDM.


Subject(s)
Blood Glucose , Body Mass Index , Glucose Tolerance Test , Homeostasis , Obesity , Overweight , Humans , Female , Pregnancy , Adult , Blood Glucose/metabolism , Obesity/metabolism , Obesity/blood , Overweight/metabolism , Overweight/blood , Diabetes, Gestational/metabolism , Diabetes, Gestational/blood , Infant, Newborn , Child, Preschool , Insulin/blood , Insulin/metabolism , C-Peptide/blood , Fasting/blood , Pregnancy Complications/metabolism , Pregnancy Complications/blood
4.
Int J Mol Sci ; 25(17)2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39273309

ABSTRACT

Gestational diabetes mellitus (GDM) represents a prevalent complication during pregnancy, exerting both short-term and long-term impacts on maternal and offspring health. This review offers a comprehensive outline of DNA methylation modifications observed in various maternal and offspring tissues affected by GDM, emphasizing the intricate interplay between DNA methylation dynamics, gene expression, and the pathogenesis of GDM. Furthermore, it explores the influence of environmental pollutants, maternal nutritional supplementation, and prenatal gut microbiota on GDM development through alterations in DNA methylation profiles. Additionally, this review summarizes recent advancements in DNA methylation-based diagnostics and predictive models in early GDM detection and risk assessment for subsequent type 2 diabetes. These insights contribute significantly to our understanding of the epigenetic mechanisms underlying GDM development, thereby enhancing maternal and fetal health outcomes and advocating further efforts in this field.


Subject(s)
DNA Methylation , Diabetes, Gestational , Epigenesis, Genetic , Diabetes, Gestational/genetics , Diabetes, Gestational/metabolism , Humans , Pregnancy , Female , Gastrointestinal Microbiome/genetics , Animals , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism
5.
Clin Sci (Lond) ; 138(18): 1179-1207, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39289953

ABSTRACT

With the global prevalence of diabetes increasing, more people of reproductive age are experiencing hyperglycaemic pregnancies. Maternal Type 1 (T1DM) or Type 2 (T2DM) diabetes mellitus, and gestational diabetes mellitus (GDM) are associated with maternal cardiovascular and metabolic complications. Pregnancies complicated by maternal diabetes also increase the risk of short- and long-term health complications for the offspring, including altered fetal growth and the onset of T2DM and cardiometabolic diseases throughout life. Despite advanced methods for improving maternal glucose control, the prevalence of adverse maternal and offspring outcomes associated with maternal diabetes remains high. The placenta is a key organ at the maternal-fetal interface that regulates fetal growth and development. In pregnancies complicated by maternal diabetes, altered placental development and function has been linked to adverse outcomes in both mother and fetus. Emerging evidence suggests that microRNAs (miRNAs) are key molecules involved in mediating these changes. In this review, we describe the role of miRNAs in normal pregnancy and discuss how miRNA dysregulation in the placenta and maternal circulation is associated with suboptimal placental development and pregnancy outcomes in individuals with maternal diabetes. We also discuss evidence demonstrating that miRNA dysregulation may affect the long-term health of mothers and their offspring. As such, miRNAs are potential candidates as biomarkers and therapeutic targets in diabetic pregnancies at risk of adverse outcomes.


Subject(s)
Diabetes, Gestational , MicroRNAs , Placenta , Pregnancy in Diabetics , Humans , Pregnancy , Female , MicroRNAs/genetics , MicroRNAs/metabolism , Diabetes, Gestational/genetics , Diabetes, Gestational/metabolism , Pregnancy in Diabetics/genetics , Pregnancy in Diabetics/metabolism , Placenta/metabolism , Pregnancy Outcome
6.
Physiol Res ; 73(4): 609-619, 2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39264081

ABSTRACT

Gestational diabetes mellitus (GDM) is a common disease during pregnancy that has adverse effects on both the mother and fetus. There are currently rare researches on the effect of vitamin supplementation on GDM pregnant mother and their offspring on animal and cell levels systematically. This work supplemented the GDM pregnant mouse model with vitamin D and found that vitamin D can effectively alleviate the hyperglycemia in GDM pregnant mice, increase blood insulin and adiponectin concentrations, and improve GTT and ITT in pregnant mice. In addition, vitamin D can reduce the incidence of death and high birth weight of offspring caused by GDM. The offspring of GDM pregnant mice had higher blood glucose levels in the first 5 weeks after birth compared to the normal group, and then returned to normal levels. Vitamin D can alleviate abnormal glucose metabolism in newborn mice. The therapeutic effect exhibited by vitamin D may be due to their anti-inflammatory effects, as vitamin D supplementation significantly reduces the levels of TFN-?, MCP-1, IL-1? and IL-8 in the blood. Vitamin D also regulates liver lipid metabolism, resulting in a decrease in liver lipid accumulation and a decrease in blood triglycerides (TG) and cholesterol (CHO). The results of this study demonstrate that vitamin D supplementation can serve as an effective treatment strategy for alleviating GDM symptoms. Keywords: Gestational diabetes mellitus, Vitamin D, Glucose metabolism, Anti-inflammatory.


Subject(s)
Blood Glucose , Diabetes, Gestational , Disease Models, Animal , Vitamin D , Animals , Diabetes, Gestational/metabolism , Diabetes, Gestational/prevention & control , Diabetes, Gestational/blood , Diabetes, Gestational/drug therapy , Female , Pregnancy , Vitamin D/pharmacology , Vitamin D/therapeutic use , Mice , Blood Glucose/metabolism , Blood Glucose/drug effects , Glucose/metabolism , Dietary Supplements , Mice, Inbred C57BL
7.
Int J Mol Sci ; 25(18)2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39337670

ABSTRACT

Gestational diabetes mellitus (GDM) is a common condition during pregnancy. The prevalence of GDM is continuously increasing worldwide. Due to accessible diagnostic methods and a clear understanding of risk factors, GDM can be effectively diagnosed and managed. Galectins may influence immunomodulatory and inflammatory processes. This study examines the expression of galectin-7 in the placentas of women with gestational diabetes (GDM), compares it to its expression in healthy pregnancies, and evaluates the associated clinical outcomes. The placentas of 40 healthy women and 40 GDM placentas were included in the cohort. The expression level of galecin-7 was measured in the syncytiotrophoblast (SCT) and in the decidua of the placenta by immunohistochemistry and double immunofluorescence staining. The evaluation was performed by an immunoreactivity score (IRS). The study results show an increased expression of galectin-7 in the SCT and the decidua of GDM placentas as compared to the placentas of the control group. Elevated levels of galectin-7 were observed in both the nucleus and the cytoplasm. This study investigated the hypothesis that galectins are involved in pathophysiological processes of gestational diabetes. Statistical analysis of gene expression patterns confirmed that galectin-7 is indeed upregulated in GDM placentas. Further studies are needed to show the correlation of galectin-7 and the development and maintenance of gestational diabetes mellitus.


Subject(s)
Diabetes, Gestational , Galectins , Placenta , Humans , Diabetes, Gestational/metabolism , Diabetes, Gestational/genetics , Female , Pregnancy , Galectins/metabolism , Galectins/genetics , Placenta/metabolism , Adult , Trophoblasts/metabolism , Decidua/metabolism , Decidua/pathology , Case-Control Studies
8.
Kaohsiung J Med Sci ; 40(10): 916-925, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39230472

ABSTRACT

This study aims to investigate the effects of the Galectin-3 (Gal-3) inhibitor TD139 on inflammation and the extracellular signal-regulated kinase (ERK)/c-Jun N-terminal kinase (JNK)/p38 pathway in gestational diabetes mellitus (GDM). Human placental tissues were treated with TD139 and TNF-α, assessing Gal-3, ERK/JNK/p38 activation, and inflammatory cytokines. GDM was induced in mice via subcutaneous injections of streptozotocin (STZ). After confirming GDM, mice were treated with 15 mg/kg TD139 on GD 10.5 12.5, 14.5, 16.5, and 18.5. Serum inflammatory cytokines were measured on GD 20.5, and post-delivery placental tissues were analyzed. Data were analyzed using one-way or two-way repeated measures ANOVA with post hoc tests. TD139 suppressed TNF-α-induced increases in Gal-3, IL-1ß, IL-6, MCP-1, and ERK/JNK/p38 activation in placental tissues. In STZ-induced GDM mice, TD139 reduced glucose levels, weight loss, and food and water intake. TD139 significantly lowered TNF-α, IL-1ß, IL-6, and MCP-1 in serum and placental tissues and inhibited the ERK/JNK/p38 pathway. TD139 improved pup numbers in GDM mice compared to untreated ones. TD139 reduces inflammation and inhibits the ERK/JNK/p38 pathway in TNF-α stimulated placental tissues and STZ-induced GDM mice, suggesting its therapeutic potential for managing GDM-related placental inflammation and improving pregnancy outcomes. The study used TNF-α to mimic GDM in placental tissues and an STZ-induced GDM mouse model, which may not fully represent human GDM complexity. Future research should explore alternative models, and broader signaling pathways, and thoroughly evaluate TD139's safety in pregnancy.


Subject(s)
Diabetes, Gestational , Galectin 3 , MAP Kinase Signaling System , Placenta , Pregnancy , Animals , Diabetes, Gestational/drug therapy , Diabetes, Gestational/metabolism , Female , Mice , Humans , Placenta/metabolism , Placenta/drug effects , Galectin 3/metabolism , Galectin 3/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Inflammation/drug therapy , p38 Mitogen-Activated Protein Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , Cytokines/metabolism , Cytokines/blood , Streptozocin , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism
9.
Nutr Diabetes ; 14(1): 77, 2024 09 27.
Article in English | MEDLINE | ID: mdl-39333472

ABSTRACT

BACKGROUND: Ferritin, a key indicator of body iron levels, has been reported to associate with type 2 diabetes (T2DM) and the onset of Gestational diabetes mellitus (GDM). However, limited research explores the association between mid-pregnancy ferritin levels and the risk of postpartum abnormal glucose metabolism (AGM) in patients with GDM. METHODS: A retrospective cohort study was conducted in 1514 women with GDM recruited from January 2016 to January 2021, and 916 women were included. Demographic characteristics, medical history and family history, pregnancy complications were recorded. Multiple logistic regression models were performed to assess the association between mid-pregnancy ferritin levels and the risk of postpartum AGM. RESULTS: Following the postpartum oral glucose tolerance test, 307 (33.5%) exhibited AGM. The AGM group had higher mid-pregnancy serum ferritin levels [AGM vs NGT: 23 (11.7, 69) µg/L vs 17.80 (9.85, 40.7) µg/L, P < 0.001] and had a larger proportion of women with ferritin levels ≥30 µg/L (AGM vs NGT: 43.6% vs 31.4%, P < 0.001). Logistic regression analysis demonstrated that women with ferritin levels≥ 30 µg/L had a 1.566 times higher risk of developing postpartum AGM. CONCLUSIONS: These findings indicate that elevated mid-pregnancy ferritin levels are significantly and independently associated with increased postpartum AGM risk in women with previous GDM. Consequently, cautious consideration is necessary for prescribing iron supplements in prenatal care, particularly for non-anemic women with GDM at high risk of developing diabetes after delivery.


Subject(s)
Blood Glucose , Diabetes, Gestational , Ferritins , Glucose Tolerance Test , Postpartum Period , Humans , Female , Diabetes, Gestational/blood , Diabetes, Gestational/metabolism , Pregnancy , Ferritins/blood , Adult , Retrospective Studies , Postpartum Period/blood , Blood Glucose/metabolism , Blood Glucose/analysis , Diabetes Mellitus, Type 2/blood , Risk Factors
10.
J Obstet Gynaecol Res ; 50(10): 1813-1829, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39288911

ABSTRACT

INTRODUCTION: Hyperglycemia is closely related to trophoblast dysfunction during pregnancy and results in suppressed invasion, migration, and pro-inflammatory cell death of trophoblasts. Hyperglycemia is a dependent risk factor for gestational hypertension accompanied by decreased placental growth factor (PLGF), which is important for maternal and fetal development. However, there is currently a lack of evidence to support whether PLGF can alleviate trophoblast cell dysfunction caused by high blood sugar. Here, we aim to clarify the effect of hyperglycemia on trophoblast dysfunction and determine how PLGF affects this process. METHODS: The changes in placental tissue histomorphology from gestational diabetes mellitus (GDM) patients were compared with those of normal placentas. HTR8/SVneo cells were cultured in different amounts of glucose to examine cellular pyroptosis, migration, and invasion as well as PLGF levels. Furthermore, the levels of pyroptosis-related proteins (NLRP3, pro-caspase1, caspase1, IL-1ß, and Gasdermin D [GSDMD]) as well as autophagy-related proteins (LC3-II, Beclin1, and p62) were examined by Western blotting. The GFP-mRFP-LC3-II system and transmission electron microscopy were used to detect mitophagy levels, and small interfering RNAs targeting BCL2 Interacting Protein 3 (siBNIP3) and PTEN-induced kinase 1 (siPINK1) were used to determine the role of mitophagy in pyroptotic death of HTR-8/SVneo cells. RESULTS: Our results show that hyperglycemia upregulates NLRP3, pro-caspase1, caspase1, IL-1ß at the protein level in GDM patients. High glucose (HG, 25 mM) inhibits viability, invasion, and migration of trophoblast cells while suppressing superoxide dismutase levels and promoting malondialdehyde production, thus leading to a senescence associated beta-gal-positive cell burst. PLGF levels in nucleus and the cytosol are also inhibited by HG, whereas PLGF treatment inhibited pyroptosis-related protein levels of NLRP3, pro-caspase1, caspase1, IL-1ß, and GSDMD, Gasdermin D N-terminal domain (GSDMD-N). HG-induced mitochondrial dysfunction and BNIP3 and PINK1/Parkin expression. Knocking down BINP3 and PINK1 abolished the protective role of PLGF by preventing mitophagy. CONCLUSION: PLGF inhibited hyperglycemia, while PLGF reversed hyperglycemic injury by promoting mitophagy via the BNIP3/PINK1/Parkin pathway. Altogether, these results suggest that PLGF may protect against trophoblast dysfunction in diabetes.


Subject(s)
Diabetes, Gestational , Hyperglycemia , Mitophagy , Placenta Growth Factor , Pyroptosis , Trophoblasts , Humans , Pyroptosis/drug effects , Pyroptosis/physiology , Trophoblasts/metabolism , Female , Pregnancy , Placenta Growth Factor/metabolism , Diabetes, Gestational/metabolism , Hyperglycemia/metabolism , Mitophagy/drug effects , Adult , Cell Line
11.
J Diabetes Res ; 2024: 1386469, 2024.
Article in English | MEDLINE | ID: mdl-39109165

ABSTRACT

Background: Gestational diabetes mellitus (GDM) is a pregnancy-related diabetic condition that may cause serious complications. However, its pathogenesis remains unclear. Placental damage due to GDM may lead to several health issues that cannot be ignored. Thus, we aimed to identify the mechanisms underlying GDM by screening differentially expressed genes (DEGs) related to vascular endothelial cells in the GDM databases and verify the expression of these DEGs in the placentas of women afflicted by GDM. Methods: We used GDM microarray datasets integrated from the Gene Expression Omnibus (GEO) database. Functional annotation and protein-protein interaction (PPI) analyses were used to screen DEGs. Placental tissues from 20 pregnant women with GDM and 20 healthy pregnant women were collected, and differential gene expression in the placental tissues was verified via qRT-PCR, western blotting, and immunofluorescence. Results: Bioinformatics analysis revealed three significant DEGs: SNAIL2, PAPP-A, and TGFß1. These genes were all predicted to be underexpressed in patients with GDM. The results of qRT-PCR, western blot, and immunofluorescence analyses indicated that SNAIL2 and PAPP-A in the placenta tissue of patients with GDM were significantly underexpressed. However, TGFß1 in the placenta tissues of GDM was significantly overexpressed. Conclusion: SNAIL2, TGFß1, and PAPP-A may affect the placentas of pregnant women with GDM, warranting further investigation.


Subject(s)
Diabetes, Gestational , Placenta , Pregnancy-Associated Plasma Protein-A , Snail Family Transcription Factors , Transforming Growth Factor beta1 , Humans , Diabetes, Gestational/metabolism , Diabetes, Gestational/genetics , Pregnancy , Female , Placenta/metabolism , Snail Family Transcription Factors/metabolism , Snail Family Transcription Factors/genetics , Adult , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Pregnancy-Associated Plasma Protein-A/genetics , Pregnancy-Associated Plasma Protein-A/metabolism , Gene Expression Profiling , Computational Biology , Case-Control Studies , Protein Interaction Maps
12.
J Nanobiotechnology ; 22(1): 509, 2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39182087

ABSTRACT

BACKGROUND: Extracellular vesicles (EVs) are membrane-enclosed structures containing lipids, proteins, and RNAs that play a crucial role in cell-to-cell communication. However, the precise mechanism through which circulating EVs disrupt hepatic glucose homeostasis in gestational diabetes mellitus (GDM) remains unclear. RESULTS: Circulating EVs isolated from human plasma were co-cultured with mammalian liver cells to investigate the potential induction of hepatic insulin resistance by GDM-EVs using glucose output assays, Seahorse assays, metabolomics, fluxomics, qRT-PCR, bioinformatics analyses, and luciferase assays. Our findings demonstrated that hepatocytes exposed to GDM-EVs exhibited increased gluconeogenesis, attenuated energy metabolism, and upregulated oxidative stress. Particularly noteworthy was the discovery of miR-1299 as the predominant miRNA in GDM-EVs, which directly targeting the 3'-untranslated regions (UTR) of STAT3. Our experiments involving loss- and gain-of-function revealed that miR-1299 inhibits the insulin signaling pathway by regulating the STAT3/FAM3A axis, resulting in increased insulin resistance through the modulation of mitochondrial function and oxidative stress in hepatocytes. Moreover, experiments conducted in vivo on mice inoculated with GDM-EVs confirmed the development of glucose intolerance, insulin resistance, and downregulation of STAT3 and FAM3A. CONCLUSIONS: These results provide insights into the role of miR-1299 derived from circulating GDM-EVs in the progression of insulin resistance in hepatic cells via the STAT3/FAM3A axis and downstream metabolic reprogramming.


Subject(s)
Diabetes, Gestational , Extracellular Vesicles , Glucose , Hepatocytes , Homeostasis , Insulin Resistance , Liver , MicroRNAs , STAT3 Transcription Factor , Animals , Female , Humans , Mice , Pregnancy , 3' Untranslated Regions , Diabetes, Gestational/metabolism , Diabetes, Gestational/genetics , Extracellular Vesicles/metabolism , Glucose/metabolism , Hep G2 Cells , Hepatocytes/metabolism , Liver/metabolism , Mice, Inbred C57BL , MicroRNAs/metabolism , MicroRNAs/genetics , Oxidative Stress , Signal Transduction , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics
13.
Adipocyte ; 13(1): 2390833, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39162358

ABSTRACT

BACKGROUND: Neuregulin 4 (Nrg4) is a brown adipose tissue-derived adipokine that greatly affects systemic metabolism and improves metabolic derangements. Although abnormal circulating levels of Nrg4 are common in obesity, it remains elusive whether low or elevated levels of this batokine are associated with the onset of metabolic diseases. AIM: To assess Nrg4 levels and its role as a feasible biomarker to predict the severity of obesity, gestational diabetes mellitus (GDM), type 2 diabetes mellitus (T2DM), non-alcoholic fatty liver disease (NAFLD), and cardiovascular diseases (CVD). METHODS: A search for relevant studies was performed systematically using prominent search engines, including PubMed, Google Scholar, and Embase, by following PRISMA guidelines. RESULTS: Ample clinical evidence reported low serum/plasma levels of Nrg4 in obesity and these were inversely proportional to the indices of metabolic syndrome, including body mass index, waist circumference, triglycerides, fasting plasma glucose, and homoeostatic model assessment for insulin resistance as well as high-sensitivity C-reactive protein. Low circulating Nrg4 levels may aid in the prediction of morbid obesity, and subsequent GDM, T2DM, NAFLD, and CVD. CONCLUSION: Current clinical evidence emphasizes that the circulating levels of Nrg4 are decreased in morbid obesity, and it also highlights that Nrg4 May serve as a potential prognostic biomarker for obesity-related metabolic diseases.


Subject(s)
Biomarkers , Neuregulins , Obesity , Humans , Neuregulins/blood , Neuregulins/metabolism , Biomarkers/blood , Obesity/blood , Obesity/metabolism , Prognosis , Metabolic Diseases/blood , Metabolic Diseases/metabolism , Metabolic Diseases/diagnosis , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/metabolism , Female , Pregnancy , Severity of Illness Index , Diabetes, Gestational/blood , Diabetes, Gestational/metabolism
14.
Sci Rep ; 14(1): 20136, 2024 08 29.
Article in English | MEDLINE | ID: mdl-39209899

ABSTRACT

Pregestational diabetes mellitus (PGDM) has an impact on fetal bone formation, but the underlying mechanism is still obscure. Although miRNAs have been extensively investigated throughout bone formation, their effects on fetal bone development caused by PGDM still need clarification. This study intends to examine the mechanism by which hyperglycemia impairs the bone formation of offspring via miR-322-5p (miR-322). In this study, miR-322 was selected by systemically screening utilizing bioinformatics and subsequent validation experiments. Using streptozotocin (STZ)-induced diabetic mice and ATDC5 cell lines, we found that miR-322 was abundantly expressed in the proliferative and hypertrophic zones of the growth plate, and its expression pattern was disturbed in the presence of hyperglycemia, suggesting that miR-322 is involved in the chondrocyte proliferation and differentiation in absence/presence of hyperglycemia. This observation was proved by manipulating miR-322 expression in ATDC5 cells by transfecting mimic and inhibitor of miR-322. Furthermore, Adamts5, Col12a1, and Cbx6 were identified as the potential target genes of miR-322, verified by the co-transfection of miR-322 inhibitor and the siRNAs, respectively. The evaluation criteria are the chondrocyte proliferation and differentiation and their relevant key gene expressions (proliferation: Sox9 and PthIh; differentiation: Runx2 and Col10a1) after manipulating the gene expressions in ATDC5 cells. This study revealed the regulative role miR-322 on chondrocyte proliferation and differentiation of growth plate by targeting Adamts5, Col12a1, and Cbx6 in hyperglycemia during pregnancy. This translational potential represents a promising avenue for advancing our understanding of bone-related complications in diabetic pregnancy and mitigating bone deficiencies in diabetic pregnant individuals, improving maternal and fetal outcomes.


Subject(s)
Cell Differentiation , Cell Proliferation , Chondrocytes , Diabetes Mellitus, Experimental , Diabetes, Gestational , Growth Plate , MicroRNAs , Animals , Female , Mice , Pregnancy , Cell Differentiation/genetics , Cell Line , Cell Proliferation/genetics , Chondrocytes/metabolism , Chondrocytes/pathology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes, Gestational/metabolism , Diabetes, Gestational/genetics , Diabetes, Gestational/pathology , Growth Plate/metabolism , Growth Plate/pathology , MicroRNAs/genetics , MicroRNAs/metabolism
15.
Gene ; 931: 148894, 2024 Dec 30.
Article in English | MEDLINE | ID: mdl-39191355

ABSTRACT

BACKGROUND: m6A modification plays a vital role in gestational diabetes mellitus (GDM) progression. However, the role of METTL3 and differential m6A-modified circRNAs in GDMremainsto be investigated. METHODS: Placental tissue samples from GDM patients and normal controls (NC) were collected to measure changes in m6A modification levels. MeRIP-seq on placental tissue was performed to detect differential m6A-modified circRNAs.High glucose (HG)-treated JEG3 cells were used to establish the GDM cell model. Differentially expressed circRNAs levels in GDM and NC groups were measured by qRT-PCR. We knocked down METTL3 to study its function. Additionally, we conducted functional recovery experiments. Dot blot assay was utilized to assess changes in m6A levels. MeRIP-qPCR was performed to evaluate the effect of knocking down METTL3 on m6A modification of hsa_circ_0072380 in JEG3 cells. RESULTS: Compared with the NC group, the GDM group exhibited increased levels of m6A modification and METTL3 expression. Differences in m6A modification of circRNAs exist between the GDM and NC groups. Hsa_circ_0000994, hsa_circ_0058733, and hsa_circ_0072380 were significantly down-regulated in the GDM group while hsa_circ_0036376, hsa_circ_0000471, and hsa_circ_0001173 showed no significant differences between two groups. HG treatment promoted METTL3 expression and m6A level of JEG3 cells, and inhibited cell proliferation, migration, and invasion abilities. Knocking down METTL3 reversed these effects. After HG treatment, hsa_circ_0072380 was significantly down-regulated. Knocking down METTL3 led to up-regulation of hsa_circ_0072380, while knocking down hsa_circ_0072380 restored the function of SiMETTL3. Additionally, knocking down METTL3 significantly reduced the m6A modification of hsa_circ_0072380. CONCLUSION: METTL3 mediated m6A modification of hsa_circ_0072380 to regulate GDM progression.


Subject(s)
Diabetes, Gestational , Methyltransferases , RNA, Circular , Humans , Methyltransferases/genetics , Methyltransferases/metabolism , Female , Diabetes, Gestational/metabolism , Diabetes, Gestational/genetics , Pregnancy , RNA, Circular/genetics , RNA, Circular/metabolism , Adult , Cell Proliferation , Disease Progression , Adenosine/analogs & derivatives , Adenosine/metabolism , Placenta/metabolism , Cell Line, Tumor
16.
Food Chem ; 460(Pt 3): 140663, 2024 Dec 01.
Article in English | MEDLINE | ID: mdl-39142199

ABSTRACT

Gestational diabetes mellitus (GDM) is a prevalent metabolic disorder during pregnancy that alters the metabolites in human milk. Integrated Gas Chromatography-Mass Spectrometry (GC-MS) and Liquid Chromatography-Mass Spectrometry (LC-MS) were employed for comprehensive identification and comparison of metabolites in mature human milk (MHM) from women with and without GDM. A total of 268 differentially expressed metabolites (DEMs) were identified. Among these, linoleic acid, arachidonic acid, 9R-HODE and L-glutamic acid were significantly elevated and 12,13-DHOME was significantly decreased in MHM of women with GDM. These metabolites are significantly enriched in linoleic acid metabolism, fatty acid biosynthesis, galactose metabolism and ABC transporters pathways. Disorders in these metabolic pathways are associated with insulin resistance and poor glucose metabolism indicating these conditions may persist postpartum.


Subject(s)
Diabetes, Gestational , Metabolomics , Milk, Human , Humans , Female , Milk, Human/chemistry , Milk, Human/metabolism , Pregnancy , Diabetes, Gestational/metabolism , Adult , Gas Chromatography-Mass Spectrometry , Chromatography, Liquid
18.
Diabetes Res Clin Pract ; 216: 111830, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39159865

ABSTRACT

AIMS: Metabolic characteristics and outcomes were compared among pregnant individuals with varying levels of glucose intolerance. METHODS: 827 participants from a randomized clinical trial comparing the IADPSG and Carpenter Coustan Criteria were grouped as follows: normal glucose tolerance, mild glucose intolerance (100 g OGTT with one abnormal value) and treated GDM (diagnosed by Carpenter Coustan or IADPSG criteria). Differences in metabolic characteristics and perinatal outcomes were assessed using inverse probability of treatment weighting. RESULTS: Mild glucose intolerance had lower insulin sensitivity and beta cell response than normal glucose tolerance, and similar findings to treated GDM. Small for gestational age (SGA) (OR 0.13, 95% CI 0.08-0.24) and neonatal composite morbidity were lower (OR 0.53, 95% CI 0.38-0.74), and maternal composite morbidity higher (OR 2.03, 95% CI 1.57-2.62) when comparing mild intolerance to normal glucose tolerance. Large for gestational age (OR 3.42 95% CI 1.39-8.41) was higher while SGA (OR 0.21, 95% CI 0.05-0.81) and neonatal composite morbidity (OR 0.31, 95% CI 0.17-0.57) were lower with mild glucose intolerance compared to treated GDM. CONCLUSIONS: Mild glucose intolerance has a similar metabolic profile to treated GDM, and outcome differences are likely related to knowledge of diagnosis and treatment. CLINICAL TRIALS REGISTRY: NCT02309138.


Subject(s)
Diabetes, Gestational , Glucose Intolerance , Pregnancy Outcome , Humans , Pregnancy , Female , Glucose Intolerance/epidemiology , Adult , Diabetes, Gestational/metabolism , Infant, Newborn , Glucose Tolerance Test , Infant, Small for Gestational Age , Blood Glucose/metabolism , Blood Glucose/analysis , Insulin Resistance/physiology
19.
J Matern Fetal Neonatal Med ; 37(1): 2228450, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39115013

ABSTRACT

INTRODUCTION: The efficacy of myo-inositol supplementation to treat gestational diabetes remains controversial, and this meta-analysis aims to study the efficacy of myo-inositol supplementation on metabolic status for gestational diabetes. METHODS: Several databases including PubMed, EMbase, Web of science, EBSCO, and Cochrane library databases were systemically searched from inception to October 2021, and we included the randomized controlled trials (RCTs) assessing the effect of myo-inositol supplementation on the outcomes of women with gestational diabetes. Gestational diabetes was diagnosed if at least one threshold of glucose concentration was exceeded and the three thresholds included 92, 180, and 153 mg/dl for 0, 1 and 2 h, respectively, after a 75-g, 2-h glucose tolerance test. RESULTS: Four RCTs and 317 patients were included in this meta-analysis. Compared with routine treatment in pregnant women with gestational diabetes, myo-inositol supplementation could lead to remarkably decreased treatment requirement with insulin (odd ratio [OR] = 0.24; 95% confidence interval [CI] = 0.11-0.52; p = .0003) and homeostasis model assessment of insulin resistance (HOMA-IR, standard mean difference [SMD]= -1.18; 95% CI= -1.50 to -0.87; p < .00001), but demonstrated no obvious impact on birth weight (SMD= -0.11; 95% CI= -0.83 to 0.61 g; p = .76), cesarean section (OR = 0.82; 95% CI = 0.46-1.47; p = .51) or the need of NICU (OR = 0.88; 95% CI = 0.03-26.57; p = .94). CONCLUSIONS: Myo-inositol supplementation is effective to decrease the need of insulin treatment and HOMA-IR for gestational diabetes.


Subject(s)
Diabetes, Gestational , Inositol , Randomized Controlled Trials as Topic , Humans , Diabetes, Gestational/drug therapy , Diabetes, Gestational/metabolism , Diabetes, Gestational/diet therapy , Inositol/therapeutic use , Pregnancy , Female , Insulin Resistance , Dietary Supplements , Insulin/therapeutic use , Blood Glucose/metabolism , Blood Glucose/analysis
20.
PLoS One ; 19(8): e0298506, 2024.
Article in English | MEDLINE | ID: mdl-39088422

ABSTRACT

BACKGROUND: In recent years, numerous methods have been introduced to predict glucose levels using machine-learning techniques on patients' daily behavioral and continuous glucose data. Nevertheless, a definitive consensus remains elusive regarding modeling the combined effects of diet and exercise for optimal glucose prediction. A notable challenge is the propensity for observational patient datasets from uncontrolled environments to overfit due to skewed feature distributions of target behaviors; for instance, diabetic patients seldom engage in high-intensity exercise post-meal. METHODS: In this study, we introduce a unique application of Bayesian transfer learning for postprandial glucose prediction using randomized controlled trial (RCT) data. The data comprises a time series of three key variables: continuous glucose levels, exercise expenditure, and carbohydrate intake. For building the optimal model to predict postprandial glucose levels we initially gathered balanced training data from RCTs on healthy participants by randomizing behavioral conditions. Subsequently, we pretrained the model's parameter distribution using RCT data from the healthy cohort. This pretrained distribution was then adjusted, transferred, and utilized to determine the model parameters for each patient. RESULTS: The efficacy of the proposed method was appraised using data from 68 gestational diabetes mellitus (GDM) patients in uncontrolled settings. The evaluation underscored the enhanced performance attained through our method. Furthermore, when modeling the joint impact of diet and exercise, the synergetic model proved more precise than its additive counterpart. CONCLUSION: An innovative application of the transfer-learning utilizing randomized controlled trial data can improve the challenging modeling task of postprandial glucose prediction for GDM patients, integrating both dietary and exercise behaviors. For more accurate prediction, future research should focus on incorporating the long-term effects of exercise and other glycemic-related factors such as stress, sleep.


Subject(s)
Bayes Theorem , Blood Glucose , Exercise , Machine Learning , Postprandial Period , Humans , Blood Glucose/metabolism , Blood Glucose/analysis , Female , Postprandial Period/physiology , Exercise/physiology , Diet , Pregnancy , Diabetes, Gestational/blood , Diabetes, Gestational/metabolism , Adult
SELECTION OF CITATIONS
SEARCH DETAIL