Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.937
Filtrer
1.
J Clin Psychiatry ; 85(3)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38959498

RÉSUMÉ

Objectives: Bipolar disorder (BD) is highly heritable and associated with increased rates of metabolic syndrome (MetS). However, little is known about MetS in offspring of parents with BD. We therefore examined this topic in the Pittsburgh Bipolar Offspring Study cohort.Methods: Participants included 199 parents (n = 116 BD, diagnosed using DSM-IV; n = 83 non-BD) and 330 offspring (mean age 19.9 ± 5.3 years), including 198 high-risk offspring of parents with BD (n = 80 affected with a mood disorder) and 132 control offspring. We defined MetS and its components using International Diabetes Federation (IDF) guidelines (primary) and National Cholesterol Education Program (NCEP) guidelines (secondary). Multivariable analyses controlled for age and socioeconomic status in offspring. Sensitivity analyses controlled for psychotropic medications.Results: There was higher prevalence of MetS in parents with BD as compared to controls. NCEP-defined MetS was significantly more prevalent among affected high-risk offspring (16.3%) and controls (15.2%) vs unaffected high-risk offspring (6.0%; χ2 = 6.54, P = .04). There was greater mean number of MetS components (IDF: 1.7 ± 1.1; NCEP: 1.4 ± 1.0) among affected high-risk offspring vs unaffected high-risk offspring (IDF: 1.2 ± 1.0; NCEP: 1.0 ± 1.0) and controls (IDF: 1.3 ± 1.2; NCEP: 1.1 ± 1.1; IDF: H[2] = 10.26, P = .006; NCEP: H[2] = 9.18, P = .01). Most findings became nonsignificant in multivariable analyses. Some between-group results became nonsignificant after controlling for second-generation antipsychotics.Conclusions: This preliminary study found increased risk of MetS among affected high-risk offspring, which may be attributable to socioeconomic status. Prospective studies may determine timing of MetS onset in relation to mood disorder onset, and the role of socioeconomic status in moderating this association.


Sujet(s)
Trouble bipolaire , Enfant de personnes handicapées , Syndrome métabolique X , Humains , Trouble bipolaire/épidémiologie , Trouble bipolaire/génétique , Syndrome métabolique X/épidémiologie , Syndrome métabolique X/génétique , Mâle , Femelle , Adulte , Enfant de personnes handicapées/statistiques et données numériques , Jeune adulte , Adolescent , Prévalence , Parents , Facteurs de risque , Études cas-témoins , Enfant
2.
PLoS One ; 19(6): e0303085, 2024.
Article de Anglais | MEDLINE | ID: mdl-38941315

RÉSUMÉ

BACKGROUND: The study of non-communicable diseases (NCDs) in a developing country like Thailand has rarely been conducted in long-term cohorts, especially among the working-age population. We aim to assess the prevalence and incidence of risk factors and their associations underlying NCDs, especially type-2 diabetes mellitus (T2DM) among healthcare workers enrolled in the Siriraj Health (SIH) study cohort. METHODS: The SIH study was designed as a longitudinal cohort and conducted at Siriraj hospital, Thailand. A total of 5,011 participants (77% women) were recruited and follow-up. Physical examinations, blood biochemical analyses, family history assessments, behavior evaluations, and genetics factors were assessed. RESULTS: The average age was 35.44±8.24 years and 51% of participants were overweight and obese. We observed that men were more likely to have a prevalence of T2DM and dyslipidemia (DLP) compared to women. Aging was significantly associated with pre-diabetes and T2DM (P<0.001). Additionally, aging, metabolic syndrome, and elevated triglycerides were associated with the development of pre-diabetes and T2DM. The minor T allele of the rs7903146(C/T) and rs4506565 (A/T) were associated with a high risk of developing pre-diabetes with odds ratios of 2.74 (95% confidence interval [CI]: 0.32-23.3) and 2.71 (95% CI: 0.32-23.07), respectively; however, these associations were statistically insignificant (P>0.05). CONCLUSION: The findings of the SIH study provide a comprehensive understanding of the health status, risk factors, and genetic factors related to T2DM in a specific working population and highlight areas for further research and intervention to address the growing burden of T2DM and NCDs.


Sujet(s)
Diabète de type 2 , Personnel de santé , État prédiabétique , Humains , Diabète de type 2/génétique , Diabète de type 2/épidémiologie , Mâle , Femelle , Thaïlande/épidémiologie , Adulte , Facteurs de risque , Adulte d'âge moyen , État prédiabétique/épidémiologie , État prédiabétique/génétique , Études longitudinales , Prévalence , Prédisposition génétique à une maladie , Études de cohortes , Syndrome métabolique X/épidémiologie , Syndrome métabolique X/génétique , Polymorphisme de nucléotide simple , Peuples d'Asie du Sud-Est
3.
Front Endocrinol (Lausanne) ; 15: 1392466, 2024.
Article de Anglais | MEDLINE | ID: mdl-38911042

RÉSUMÉ

Background: The onset and progression of chronic kidney disease (CKD) has been linked to metabolic syndrome (MetS), with the results of recent observational studies supporting a potential link between renal failure and MetS. The causal nature of this relationship, however, remains uncertain. This study thus leveraged a Mendelian Randomization (MR) approach to probe the causal link of MetS with renal failure. Methods: A genetic database was initially used to identify SNPs associated with MetS and components thereof, after which causality was evaluated through the inverse variance weighted (IVW), MR-Egger regression, and weighted media techniques. Results were subsequently validated through sensitivity analyses. Results: IVW (OR = 1.48, 95% CI = 1.21-1.82, P =1.60E-04) and weighted median (OR = 1.58, 95% CI =1.15-2.17, P = 4.64E-03) analyses revealed that MetS was linked to an elevated risk of renal failure. When evaluating the specific components of MetS, waist circumference was found to be causally related to renal failure using the IVW (OR= 1.58, 95% CI = 1.39-1.81, P = 1.74e-11), MR-Egger (OR= 1.54, 95% CI = 1.03-2.29, P = 0.036), and weighted median (OR= 1.82, 95% CI = 1.48-2.24, P = 1.17e-8). The IVW method also revealed a causal association of hypertension with renal failure (OR= 1.95, 95% CI = 1.34-2.86, P = 5.42e-04), while renal failure was not causally related to fasting blood glucose, triglyceride levels, or HDL-C levels. Conclusion: These data offer further support for the existence of a causal association of MetS with kidney failure. It is thus vital that MetS be effectively managed in patients with CKD in clinical settings, particularly for patients with hypertension or a high waist circumference who are obese. Adequate interventions in these patient populations have the potential to prevent or delay the development of renal failure.


Sujet(s)
Analyse de randomisation mendélienne , Syndrome métabolique X , Polymorphisme de nucléotide simple , Humains , Syndrome métabolique X/génétique , Syndrome métabolique X/complications , Mâle , Femelle , Insuffisance rénale/génétique , Adulte d'âge moyen , Tour de taille , Facteurs de risque
4.
Sci Rep ; 14(1): 14338, 2024 06 21.
Article de Anglais | MEDLINE | ID: mdl-38906920

RÉSUMÉ

Previous research has produced inconsistent findings concerning the connection between metabolic syndrome and prostate cancer. It is challenging for observational studies to establish a conclusive causal relationship between the two. However, Mendelian randomization can provide stronger evidence of causality in this context. To examine the causal link between a metabolic composite and its components with prostate cancer, we performed a two-sample Mendelian randomization (MR) study utilizing aggregated data from genome-wide association studies, followed by meta-analyses. In our study, we employed inverse variance weighting as the primary method for MR analysis. Additionally, we assessed potential sources of heterogeneity and horizontal pleiotropy through the Cochran's Q test and MR-Egger regression. Moreover, we used multivariate MR to determine whether smoking versus alcohol consumption had an effect on the outcomes. We found no causal relationship between metabolic syndrome and its components and prostate cancer(MetS, odds ratio [OR] = 0.95, 95% confidence interval [CI] = 0.738-1.223, p = 0.691; TG, [OR] = 1.02, 95%[CI] = 0.96-1.08, p = 0.59); HDL, [OR] = 1.02, 95% [CI] = 0.97-1.07, p = 0.47; DBP, [OR] = 1.00, 95%[CI] = 0.99-1.01, p = 0.87; SBP, [OR] = 1.00, 95%[CI] = 0.99-1.00, p = 0.26; FBG [OR] = 0.92, 95%[CI] = 0.81-1.05, p = 0.23; WC, [OR] = 0.93, 95%[CI] = 0.84-1.03, p = 0.16). Finally, the MVMR confirms that the metabolic syndrome and its components are independent of smoking and alcohol consumption in prostate cancer. We didn't find significant evidence to determine a causal relationship between the metabolic syndrome and its components and prostate cancer through MR analysis. Further research is necessary to explore the potential pathogenesis between the two diseases.


Sujet(s)
Étude d'association pangénomique , Analyse de randomisation mendélienne , Syndrome métabolique X , Tumeurs de la prostate , Humains , Mâle , Consommation d'alcool/effets indésirables , Syndrome métabolique X/génétique , Odds ratio , Polymorphisme de nucléotide simple , Tumeurs de la prostate/génétique , Facteurs de risque , Fumer/effets indésirables
5.
BMC Genomics ; 25(1): 590, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38867151

RÉSUMÉ

BACKGROUND: The association between Apolipoprotein A5 (APOA5) genetic polymorphisms and susceptibility to metabolic syndrome (MetS) has been established by many studies, but there have been conflicting results from the literature. We performed a meta-analysis of observational studies to evaluate the association between APOA5 gene polymorphisms and the prevalence of MetS. METHODS: PubMed, Web of Science, Embase, and Scopus were searched up to April 2024. The random effects model was used to estimate the odds ratios (ORs) and 95% confidence intervals (CI) of the association between APOA5 gene polymorphisms and the prevalence of MetS development. The potential sources of heterogeneity were evaluated by subgroup analyses and sensitivity analyses. RESULTS: A total of 30 studies with 54,986 subjects (25,341 MetS cases and 29,645 healthy controls) were included. The presence of rs662799 and rs651821 polymorphisms is associated with an approximately 1.5-fold higher likelihood of MetS prevalence (OR = 1.42, 95% CI: 1.32, 1.53, p < 0.001; I2 = 67.1%; P-heterogeneity < 0.001; and OR = 1.50, 95% CI: 1.36-1.65, p < 0.001), respectively. MetS is also more prevalent in individuals with the genetic variants rs3135506 and rs2075291. There was no evidence of a connection with rs126317. CONCLUSION: The present findings suggest that polymorphisms located in the promoter and coding regions of the APOA5 gene are associated with an increased prevalence of MetS in the adult population. Identifying individuals with these genetic variations could lead to early disease detection and the implementation of preventive strategies to reduce the risk of MetS and its related health issues. However, because the sample size was small and there was evidence of significant heterogeneity for some APOA5 gene polymorphisms, these results need to be confirmed by more large-scale and well-designed studies.


Sujet(s)
Apolipoprotéine A-V , Prédisposition génétique à une maladie , Syndrome métabolique X , Polymorphisme de nucléotide simple , Syndrome métabolique X/génétique , Syndrome métabolique X/épidémiologie , Apolipoprotéine A-V/génétique , Humains , Odds ratio
6.
Alcohol Alcohol ; 59(4)2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38832907

RÉSUMÉ

AIMS: Alcohol drinking is associated with central obesity, hypertension, and hyperlipidemia, which further causes metabolic syndrome (MetS). However, prior epidemiological studies on such associations lack experimental evidence for a causal relationship. This study aims to explore the causal relationship between drinking behavior and MetS in Taiwan population by using Mendelian randomization (MR) analysis. METHODS: A cross-sectional study was conducted using the Taiwan Biobank database, which comprised 50 640 Han Chinese who were 30-70 years old without cancer from 2008 to 2020. In MR analysis, we constructed weighted and unweighted genetic risk scores by calculating SNP alleles significantly associated with alcohol drinking. We calculated odds ratios and 95% confidence interval (CI) by using a two-stage regression model. RESULTS: A total of 50 640 participants were included with a mean age of 49.5 years (SD: 1.67 years), 36.6% were men. The adjusted odds ratio (aOR) of MetS per 5% increase in the likelihood of genetic predisposition to drink based on weighted genetic risk score with adjustment was 1.11 (95% CI: 1.10, 1.12, P < .001). Analysis was also conducted by grouping the likelihood of genetic predisposition to drink based on quartiles with multivariate adjustment. Using Q1 as the reference group, the aORs of MetS for Q2, Q3, and Q4 were 1.19 (1.12, 1.27, p < .001), 1.31 (1.23, 1.40, p < .001), and 1.87 (1.75, 2.00, p < .001), respectively, for the weighted genetic risk score. CONCLUSIONS: This study shows a modest relationship between drinking behavior and MetS by using MR analysis.


Sujet(s)
Consommation d'alcool , Analyse de randomisation mendélienne , Syndrome métabolique X , Humains , Syndrome métabolique X/génétique , Syndrome métabolique X/épidémiologie , Mâle , Adulte d'âge moyen , Femelle , Études transversales , Adulte , Consommation d'alcool/génétique , Consommation d'alcool/épidémiologie , Consommation d'alcool/psychologie , Taïwan/épidémiologie , Sujet âgé , Prédisposition génétique à une maladie/génétique , Polymorphisme de nucléotide simple/génétique
7.
Nutrients ; 16(11)2024 May 25.
Article de Anglais | MEDLINE | ID: mdl-38892547

RÉSUMÉ

BACKGROUND: Variants in fat mass and the obesity-associated protein (FTO) gene have long been recognized as the most significant genetic predictors of body fat mass and obesity. Nevertheless, despite the overall evidence, there are conflicting reports regarding the correlation between different polymorphisms of the FTO gene and body mass index (BMI). Additionally, it is unclear whether FTO influences metabolic syndrome (MetS) through mechanisms other than BMI's impact. In this work, we aimed to analyze the impact of the following FTO polymorphisms on the BMI as well as MetS components in a population of young adult men. METHODS: The patient group consisted of 279 Polish young adult men aged 28.92 (4.28) recruited for the MAGNETIC trial. The single-nucleotide polymorphisms (SNPs), located in the first intron of the FTO gene, were genotyped, and the results were used to identify "protective" and "risk" haplotypes and diplotypes based on the literature data. Laboratory, as well as anthropometric measurements regarding MetS, were performed. Measured MetS components included those used in the definition in accordance with the current guidelines. Data regarding dietary patterns were also collected, and principal components of the dietary patterns were identified. RESULTS: No statistically significant correlations were identified between the analyzed FTO diplotypes and BMI (p = 0.53) or other MetS components (waist circumference p = 0.55; triglycerides p = 0.72; HDL cholesterol p = 0.33; blood glucose p = 0.20; systolic blood pressure p = 0.06; diastolic blood pressure p = 0.21). Stratification by the level of physical activity or adherence to the dietary patterns also did not result in any statistically significant result. CONCLUSIONS: Some studies have shown that FTO SNPs such as rs1421085, rs1121980, rs8050136, rs9939609, and rs9930506 have an impact on the BMI or other MetS components; nevertheless, this was not replicated in this study of Polish young adult males.


Sujet(s)
Alpha-ketoglutarate-dependent dioxygenase FTO , Indice de masse corporelle , Haplotypes , Mode de vie , Syndrome métabolique X , Polymorphisme de nucléotide simple , Humains , Alpha-ketoglutarate-dependent dioxygenase FTO/génétique , Mâle , Syndrome métabolique X/génétique , Syndrome métabolique X/épidémiologie , Adulte , Pologne , Jeune adulte , Régime alimentaire , Prédisposition génétique à une maladie , Comportement alimentaire ,
8.
Cell Syst ; 15(6): 497-509.e3, 2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38866010

RÉSUMÉ

Susceptibility to metabolic syndrome (MetS) is dependent on genetics, environment, and gene-by-environment interactions, rendering the study of underlying mechanisms challenging. The majority of experiments in model organisms do not incorporate genetic variation and lack specific evaluation criteria for MetS. Here, we derived a continuous metric, the metabolic health score (MHS), based on standard clinical parameters and defined its molecular signatures in the liver and circulation. In human UK Biobank, the MHS associated with MetS status and was predictive of future disease incidence, even in individuals without MetS. Using quantitative trait locus analyses in mice, we found two MHS-associated genetic loci and replicated them in unrelated mouse populations. Through a prioritization scheme in mice and human genetic data, we identified TNKS and MCPH1 as candidates mediating differences in the MHS. Our findings provide insights into the molecular mechanisms sustaining metabolic health across species and uncover likely regulators. A record of this paper's transparent peer review process is included in the supplemental information.


Sujet(s)
Syndrome métabolique X , Locus de caractère quantitatif , Animaux , Souris , Locus de caractère quantitatif/génétique , Syndrome métabolique X/génétique , Syndrome métabolique X/métabolisme , Humains , Mâle , Prédisposition génétique à une maladie/génétique , Femelle , Souris de lignée C57BL , Étude d'association pangénomique/méthodes , Biologie des systèmes/méthodes
9.
Nutr J ; 23(1): 61, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38862960

RÉSUMÉ

BACKGROUND: The Mediterranean diet (MedDiet) has demonstrated efficacy in preventing age-related cognitive decline and modulating plasma concentrations of endocannabinoids (eCBs) and N-acylethanolamines (NAEs, or eCB-like compounds), which are lipid mediators involved in multiple neurological disorders and metabolic processes. Hypothesizing that eCBs and NAEs will be biomarkers of a MedDiet intervention and will be related to the cognitive response, we investigated this relationship according to sex and apolipoprotein E (APOE) genotype, which may affect eCBs and cognitive performance. METHODS: This was a prospective cohort study of 102 participants (53.9% women, 18.8% APOE-ɛ4 carriers, aged 65.6 ± 4.5 years) from the PREDIMED-Plus-Cognition substudy, who were recruited at the Hospital del Mar Research Institute (Barcelona). All of them presented metabolic syndrome plus overweight/obesity (inclusion criteria of the PREDIMED-Plus) and normal cognitive performance at baseline (inclusion criteria of this substudy). A comprehensive battery of neuropsychological tests was administered at baseline and after 1 and 3 years. Plasma concentrations of eCBs and NAEs, including 2-arachidonoylglycerol (2-AG), anandamide (AEA), oleoylethanolamide (OEA), palmitoylethanolamide (PEA), and N-docosahexaenoylethanolamine (DHEA), were also monitored. Baseline cognition, cognitive changes, and the association between eCBs/NAEs and cognition were evaluated according to gender (crude models), sex (adjusted models), and APOE genotype. RESULTS: At baseline, men had better executive function and global cognition than women (the effect size of gender differences was - 0.49, p = 0.015; and - 0.42, p = 0.036); however, these differences became nonsignificant in models of sex differences. After 3 years of MedDiet intervention, participants exhibited modest improvements in memory and global cognition. However, greater memory changes were observed in men than in women (Cohen's d of 0.40 vs. 0.25; p = 0.017). In men and APOE-ε4 carriers, 2-AG concentrations were inversely associated with baseline cognition and cognitive changes, while in women, cognitive changes were positively linked to changes in DHEA and the DHEA/AEA ratio. In men, changes in the OEA/AEA and OEA/PEA ratios were positively associated with cognitive changes. CONCLUSIONS: The MedDiet improved participants' cognitive performance but the effect size was small and negatively influenced by female sex. Changes in 2-AG, DHEA, the OEA/AEA, the OEA/PEA and the DHEA/AEA ratios were associated with cognitive changes in a sex- and APOE-dependent fashion. These results support the modulation of the endocannabinoid system as a potential therapeutic approach to prevent cognitive decline in at-risk populations. TRIAL REGISTRATION: ISRCTN89898870.


Sujet(s)
Cognition , Régime méditerranéen , Endocannabinoïdes , Génotype , Syndrome métabolique X , Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen , Amides , Apolipoprotéines E/génétique , Acides arachidoniques/sang , Marqueurs biologiques/sang , Cognition/physiologie , Régime méditerranéen/statistiques et données numériques , Endocannabinoïdes/sang , Éthanolamines/sang , Glycérides/sang , Syndrome métabolique X/génétique , Acides oléiques/sang , Acides palmitiques/sang , Amides gras polyinsaturés N-alkylés/sang , Études prospectives , Facteurs sexuels
10.
J ASEAN Fed Endocr Soc ; 39(1): 45-52, 2024.
Article de Anglais | MEDLINE | ID: mdl-38863909

RÉSUMÉ

Introduction: The purpose of this study was to determine the possible differences in genetic polymorphisms and serum levels of chromogranin A (CgA), according to age and sex, in subjects with and without metabolic syndrome (MetS). Methodology: The genotyping and serum level of CgA and biochemical parameters were measured by the T-ARMS-PCR and PCR-RFLP and ELISA and spectrophotometer methods, respectively. Results: A comparison of males with and without MetS showed significantly lower high-density lipoprotein-cholesterol (HDL-C) levels than those of females.At ages 30-70 years, both sexes showed significant differences in triglycerides (TG), fasting blood sugar (FBS), CgA levels and waist circumference (WC) when compared to the two groups. Both sexes with MetS indicated significant differences in systolic blood pressure (SBP) at ages 40-70 years, while at ages 40-59 years, there was a significant difference in HDL-C level in males.There was a significant correlation between serum levels of FBS, TG, SBP and WC (in both sexes), and CgA in subjects with MetS. Significant correlation was found between HDL-C level and diastolic blood pressure (DBP), and CgA level in males and females, respectively. CgA genotype frequency (T-415C and C+87T polymorphisms) showed no significant differences between males and females with and without MetS, while there was only a significant difference in frequency of the genotypes T-415C when compared to males with and without MetS. Conclusion: The CgA appears to be strongly associated with MetS components in both sexes. Variation in CgA gene expression may affect the T-415C polymorphism in males. This may mean that the structure of CgA genetics differs in different ethnic groups. Differences in the serum level and expression of CgA gene may show valuable study results that it may be expected a relationship between these variables and the MetS.


Sujet(s)
Chromogranine A , Syndrome métabolique X , Humains , Mâle , Femelle , Adulte d'âge moyen , Syndrome métabolique X/génétique , Syndrome métabolique X/sang , Adulte , Sujet âgé , Chromogranine A/sang , Chromogranine A/génétique , Facteurs sexuels , Facteurs âges , Polymorphisme génétique/génétique , Pression sanguine/génétique , Génotype , Triglycéride/sang , Cholestérol HDL/sang , Tour de taille/génétique
11.
Int J Mol Sci ; 25(12)2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38928349

RÉSUMÉ

The role of adipose mesenchymal stem cells (Ad-MSCs) in metabolic syndrome remains unclear. We aimed to assess the expression of selected microRNAs in Ad-MSCs of non-diabetic adults in relation to Ad-MSC secretion of protein regulators and basic metabolic parameters. Ten obese, eight overweight, and five normal weight subjects were enrolled: 19 females and 4 males; aged 43.0 ± 8.9 years. Ad-MSCs were harvested from abdominal subcutaneous fat. Ad-MSC cellular expressions of four microRNAs (2-ΔCt values) and concentrations of IL-6, IL-10, VEGF, and IGF-1 in the Ad-MSC-conditioned medium were assessed. The expressions of miR-21, miR-122, or miR-192 did not correlate with clinical parameters (age, sex, BMI, visceral fat, HOMA-IR, fasting glycemia, HbA1c, serum lipids, CRP, and eGFR). Conversely, the expression of miR-155 was lowest in obese subjects (3.69 ± 2.67 × 10-3 vs. 7.07 ± 4.42 × 10-3 in overweight and 10.25 ± 7.05 × 10-3 in normal weight ones, p = 0.04). The expression of miR-155 correlated inversely with BMI (sex-adjusted r = -0.64; p < 0.01), visceral adiposity (r = -0.49; p = 0.03), and serum CRP (r = -0.63; p < 0.01), whereas it correlated positively with serum HDL cholesterol (r = 0.51; p = 0.02). Moreover, miR-155 synthesis was associated marginally negatively with Ad-MSC secretion of IGF-1 (r = -0.42; p = 0.05), and positively with that of IL-10 (r = 0.40; p = 0.06). Ad-MSC expression of miR-155 appears blunted in visceral obesity, which correlates with Ad-MSC IGF-1 hypersecretion and IL-10 hyposecretion, systemic microinflammation, and HDL dyslipidemia. Ad-MSC studies in metabolic syndrome should focus on miR-155.


Sujet(s)
Tissu adipeux , Cellules souches mésenchymateuses , Syndrome métabolique X , microARN , Humains , microARN/génétique , microARN/métabolisme , Femelle , Mâle , Syndrome métabolique X/métabolisme , Syndrome métabolique X/génétique , Cellules souches mésenchymateuses/métabolisme , Adulte , Adulte d'âge moyen , Tissu adipeux/métabolisme , Facteur de croissance IGF-I/métabolisme , Facteur de croissance IGF-I/génétique , Obésité/métabolisme , Obésité/génétique , Interleukine-10/métabolisme , Interleukine-10/génétique , Régulation de l'expression des gènes , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique
12.
Nutrients ; 16(9)2024 Apr 25.
Article de Anglais | MEDLINE | ID: mdl-38732523

RÉSUMÉ

The presence of vitamin D3 deficiency associated with the presence of metabolic syndrome (MS) has important public health effects. This study aims to investigate the relationship between vitamin D3 deficiency, MS and vitamin D3 receptor (VDR), GC Vitamin D binding protein (GC), and cytochrome P450 family 2 subfamily R member 1 (CYP2R1) gene polymorphisms, and genes whose encoded proteins are responsible for vitamin D3 metabolism and transport. A total of 58 participants were included in this study (age 39 ± 12 years) and were selected over a 12-month period. They were divided into four groups, depending on the presence of polymorphisms in VDR, GC, and CYP2R1 genes and their weight status. At baseline, in months 3, 6, and 12, biochemical parameters including 25(OH)D3, total cholesterol, LDL cholesterol, HDL cholesterol, triglycerides, and homeostatic model assessment (HOMA index), the insulin resistance indicator were measured. Our results show that all subjects in the polymorphism group supplemented with vitamin D3 reached an optimal level of vitamin D3 associated with high concentrations of 25(OH)D3. Weight loss was most significant in patients in the POW group (overweight patients).


Sujet(s)
Cholécalciférol , Cholestanetriol 26-monooxygenase , Syndrome métabolique X , Récepteur calcitriol , Carence en vitamine D , Protéine de liaison à la vitamine D , Adulte , Femelle , Humains , Mâle , Adulte d'âge moyen , Cholécalciférol/sang , Cholestanetriol 26-monooxygenase/génétique , Famille-2 de cytochromes P450/génétique , Insulinorésistance/génétique , Syndrome métabolique X/génétique , Polymorphisme génétique , Récepteur calcitriol/génétique , Carence en vitamine D/génétique , Carence en vitamine D/sang , Protéine de liaison à la vitamine D/génétique
13.
Redox Biol ; 73: 103139, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38696898

RÉSUMÉ

In this study, we observed worsening metabolic crosstalk in mouse models with concomitant metabolic disorders such as hyperhomocysteinemia (HHcy), hyperlipidemia, and hyperglycemia and in human coronary artery disease by analyzing metabolic profiles. We found that HHcy worsening is most sensitive to other metabolic disorders. To identify metabolic genes and metabolites responsible for the worsening metabolic crosstalk, we examined mRNA levels of 324 metabolic genes in Hcy, glucose-related and lipid metabolic systems. We examined Hcy-metabolites (Hcy, SAH and SAM) by LS-ESI-MS/MS in 6 organs (heart, liver, brain, lung, spleen, and kidney) from C57BL/6J mice. Through linear regression analysis of Hcy-metabolites and metabolic gene mRNA levels, we discovered that SAH-responsive genes were responsible for most metabolic changes and all metabolic crosstalk mediated by Serine, Taurine, and G3P. SAH-responsive genes worsen glucose metabolism and cause upper glycolysis activation and lower glycolysis suppression, indicative of the accumulation of glucose/glycogen and G3P, Serine synthesis inhibition, and ATP depletion. Insufficient Serine due to negative correlation of PHGDH with SAH concentration may inhibit the folate cycle and transsulfurarion pathway and consequential reduced antioxidant power, including glutathione, taurine, NADPH, and NAD+. Additionally, we identified SAH-activated pathological TG loop as the consequence of increased fatty acid (FA) uptake, FA ß-oxidation and Ac-CoA production along with lysosomal damage. We concluded that HHcy is most responsive to other metabolic changes in concomitant metabolic disorders and mediates worsening metabolic crosstalk mainly via SAH-responsive genes, that organ-specific Hcy metabolism determines organ-specific worsening metabolic reprogramming, and that SAH, acetyl-CoA, Serine and Taurine are critical metabolites mediating worsening metabolic crosstalk, redox disturbance, hypomethylation and hyperacetylation linking worsening metabolic reprogramming in metabolic syndrome.


Sujet(s)
Syndrome métabolique X , Animaux , Souris , Humains , Syndrome métabolique X/métabolisme , Syndrome métabolique X/génétique , Mâle , Modèles animaux de maladie humaine , Hyperhomocystéinémie/métabolisme , Hyperhomocystéinémie/génétique , Souris de lignée C57BL , Glucose/métabolisme , Métabolome , Métabolomique/méthodes , Voies et réseaux métaboliques
14.
PLoS One ; 19(5): e0298161, 2024.
Article de Anglais | MEDLINE | ID: mdl-38691570

RÉSUMÉ

INTRODUCTION: The RISKMet project aims to: (1) identify risk factors for metabolic syndrome (MetS) by comparing patients with and without MetS; (2) characterise patients treated with second-generation antipsychotics (SGAs) about MetS diagnosis; (3) study behavioural patterns, including physical activity (PA) and dietary habits, in patients and healthy individuals using a prospective cohort design. METHOD: The RISKMet project investigates MetS in individuals treated with SGAs, focusing on both adult and paediatric populations. The study utilizes a case-control design to examine potential risk factors for MetS, categorizing participants as MetS+ considered as "Cases" and MetS- considered as "Controls" matched by sex and age. The evaluation of factors such as MetS, lifestyle habits, and environmental influences is conducted at two time points, T0 and T3, after 3 months. Subsequently, the project aims to assess body parameters, including physical examinations, and blood, and stool sample collection, to evaluate metabolic markers and the impact of SGAs. The analysis includes pharmacological treatment data and genetic variability. Behavioural markers related to lifestyle, eating behaviour, PA, and mood are assessed at both T0 and T3 using interviews, accelerometers, and a mobile app. The study aims to improve mental and physical well-being in SGA-treated individuals, establish a biobank for MetS research, build an evidence base for physical health programs, and develop preventive strategies for SGA-related comorbidities. CONCLUSIONS: This project innovates MetS monitoring in psychiatry by using intensive digital phenotyping, identifying biochemical markers, assessing familial risks, and including genetically similar healthy controls. STUDY REGISTRATION NUMBER: ISRCTN18419418 at www.isrctn.com.


Sujet(s)
Neuroleptiques , Syndrome métabolique X , Humains , Syndrome métabolique X/induit chimiquement , Syndrome métabolique X/génétique , Neuroleptiques/effets indésirables , Neuroleptiques/usage thérapeutique , Facteurs de risque , Mâle , Femelle , Adulte , Études cas-témoins , Études prospectives , Mode de vie , Exercice physique , Adulte d'âge moyen , Enfant
15.
PLoS One ; 19(5): e0304410, 2024.
Article de Anglais | MEDLINE | ID: mdl-38809924

RÉSUMÉ

The association between Alzheimer's disease and metabolic disorders as significant risk factors is widely acknowledged. However, the intricate molecular mechanism intertwining these conditions remains elusive. To address this knowledge gap, we conducted a thorough investigation using a bioinformatics method to illuminate the molecular connections and pathways that provide novel perspectives on these disorders' pathological and clinical features. Microarray datasets (GSE5281, GSE122063) from the Gene Expression Omnibus (GEO) database facilitated the way to identify genes with differential expression in Alzheimer's disease (141 genes). Leveraging CoreMine, CTD, and Gene Card databases, we extracted genes associated with metabolic conditions, including hypertension, non-alcoholic fatty liver disease, and diabetes. Subsequent analysis uncovered overlapping genes implicated in metabolic conditions and Alzheimer's disease, revealing shared molecular links. We utilized String and HIPPIE databases to visualize these shared genes' protein-protein interactions (PPI) and constructed a PPI network using Cytoscape and MCODE plugin. SPP1, CD44, IGF1, and FLT1 were identified as crucial molecules in the main cluster of Alzheimer's disease and metabolic syndrome. Enrichment analysis by the DAVID dataset was employed and highlighted the SPP1 as a novel target, with its receptor CD44 playing a significant role in the inflammatory cascade and disruption of insulin signaling, contributing to the neurodegenerative aspects of Alzheimer's disease. ECM-receptor interactions, focal adhesion, and the PI3K/Akt pathways may all mediate these effects. Additionally, we investigated potential medications by repurposing the molecular links using the DGIdb database, revealing Tacrolimus and Calcitonin as promising candidates, particularly since they possess binding sites on the SPP1 molecule. In conclusion, our study unveils crucial molecular bridges between metabolic syndrome and AD, providing insights into their pathophysiology for therapeutic interventions.


Sujet(s)
Maladie d'Alzheimer , Repositionnement des médicaments , Syndrome métabolique X , Cartes d'interactions protéiques , Biologie des systèmes , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/traitement médicamenteux , Humains , Syndrome métabolique X/métabolisme , Syndrome métabolique X/génétique , Biologie des systèmes/méthodes , Réseaux de régulation génique , Biologie informatique/méthodes , Transduction du signal , Bases de données génétiques , Analyse de profil d'expression de gènes
16.
Sci Rep ; 14(1): 10505, 2024 05 07.
Article de Anglais | MEDLINE | ID: mdl-38714718

RÉSUMÉ

Metabolic syndrome (MetS) is closely related to cardiovascular and cerebrovascular diseases, and genetic predisposition is one of the main triggers for its development. To identify the susceptibility genes for MetS, we investigated the relationship between angiotensin-converting enzyme 2 (ACE2) single nucleotide polymorphisms (SNPs) and MetS in southern China. In total, 339 MetS patients and 398 non-MetS hospitalized patients were recruited. Four ACE2 polymorphisms (rs2074192, rs2106809, rs879922, and rs4646155) were genotyped using the polymerase chain reaction-ligase detection method and tested for their potential association with MetS and its related components. ACE2 rs2074192 and rs2106809 minor alleles conferred 2.485-fold and 3.313-fold greater risks of MetS in women. ACE2 rs2074192 and rs2106809 variants were risk factors for obesity, diabetes, and low-high-density lipoprotein cholesterolemia. However, in men, the ACE2 rs2074192 minor allele was associated with an approximately 0.525-fold reduction in MetS prevalence. Further comparing the components of MetS, ACE2 rs2074192 and rs2106809 variants reduced the risk of obesity and high triglyceride levels. In conclusion, ACE2 rs2074192 and rs2106809 SNPs were independently associated with MetS in a southern Chinese population and showed gender heterogeneity, which can be partially explained by obesity. Thus, these SNPs may be utilized as predictive biomarkers and molecular targets for MetS. A limitation of this study is that environmental and lifestyle differences, as well as genetic heterogeneity among different populations, were not considered in the analysis.


Sujet(s)
Angiotensin-converting enzyme 2 , Prédisposition génétique à une maladie , Syndrome métabolique X , Polymorphisme de nucléotide simple , Humains , Syndrome métabolique X/génétique , Syndrome métabolique X/épidémiologie , Angiotensin-converting enzyme 2/génétique , Femelle , Mâle , Adulte d'âge moyen , Chine/épidémiologie , Études cas-témoins , Allèles , Sujet âgé , Adulte , Facteurs de risque , Peptidyl-Dipeptidase A/génétique , Fréquence d'allèle , Génotype
17.
Clin Epigenetics ; 16(1): 60, 2024 Apr 29.
Article de Anglais | MEDLINE | ID: mdl-38685121

RÉSUMÉ

BACKGROUND: While multiple studies have investigated the relationship between metabolic syndrome (MetS) and its related traits (fasting glucose, triglyceride, HDL cholesterol, blood pressure, waist circumference) and DNA methylation, our understanding of the epigenetic mechanisms in MetS remains limited. Therefore, we performed an epigenome-wide meta-analysis of blood DNA methylation to identify differentially methylated probes (DMPs) and differentially methylated regions (DMRs) associated with MetS and its components using two independent cohorts comprising a total of 2,334 participants. We also investigated the specific genetic effects on DNA methylation, identified methylation quantitative trait loci (meQTLs) through genome-wide association studies and further utilized Mendelian randomization (MR) to assess how these meQTLs subsequently influence MetS status. RESULTS: We identified 40 DMPs and 27 DMRs that are significantly associated with MetS. In addition, we identified many novel DMPs and DMRs underlying inflammatory and steroid hormonal processes. The most significant associations were observed in 3 DMPs (cg19693031, cg26974062, cg02988288) and a DMR (chr1:145440444-145441553) at the TXNIP, which are involved in lipid metabolism. These CpG sites were identified as coregulators of DNA methylation in MetS, TG and FAG levels. We identified a total of 144 cis-meQTLs, out of which only 13 were found to be associated with DMPs for MetS. Among these, we confirmed the identified causal mediators of genetic effects at CpG sites cg01881899 at ABCG1 and cg00021659 at the TANK genes for MetS. CONCLUSIONS: This study observed whether specific CpGs and methylated regions act independently or are influenced by genetic effects for MetS and its components in the Korean population. These associations between the identified DNA methylation and MetS, along with its individual components, may serve as promising targets for the development of preventive interventions for MetS.


Sujet(s)
Ilots CpG , Méthylation de l'ADN , Épigenèse génétique , Prédisposition génétique à une maladie , Étude d'association pangénomique , Syndrome métabolique X , Locus de caractère quantitatif , Humains , Syndrome métabolique X/génétique , Méthylation de l'ADN/génétique , Ilots CpG/génétique , Étude d'association pangénomique/méthodes , République de Corée/épidémiologie , Femelle , Mâle , Adulte d'âge moyen , Prédisposition génétique à une maladie/génétique , Épigenèse génétique/génétique , Analyse de randomisation mendélienne/méthodes , Épigénome/génétique , Adulte , Sujet âgé , Protéines de transport/génétique
18.
Front Endocrinol (Lausanne) ; 15: 1325417, 2024.
Article de Anglais | MEDLINE | ID: mdl-38567309

RÉSUMÉ

Background: Observational studies have reported a possible association between metabolic syndrome (MetS) and thyroid autoimmunity. Nevertheless, the relationship between thyroid autoimmunity and MetS remains unclear. The objective of this research was to assess the causal impact of MetS on thyroid autoimmunity through the utilization of Mendelian randomization (MR) methodology. Methods: We performed bidirectional MR to elucidate the causal relationship between MetS and their components and thyroid autoimmunity (positivity of TPOAb). Single nucleotide polymorphisms (SNPs) of MetS and its components were obtained from the publicly available genetic variation summary database. The Thyroidomics Consortium conducted a genome-wide association analysis, which provided summary-level data pertaining to thyroid autoimmunity. The study included several statistical methods, including the inverse variance weighting method (IVW), weighted median, simple mode, weight mode, and MR-Egger methods, to assess the causal link. In addition, to ensure the stability of the results, a sensitivity analysis was conducted. Results: IVW showed that MetS reduced the risk of developing thyroid autoimmunity (OR = 0.717, 95% CI = 0.584 - 0.88, P = 1.48E-03). The investigation into the causative association between components of MetS and thyroid autoimmune revealed a statistically significant link between triglycerides levels and the presence of thyroid autoimmunity (IVW analysis, OR = 0.603, 95%CI = 0.45 -0.807, P = 6.82E-04). The reverse analysis did not reveal any causal relationship between thyroid autoimmunity and MetS, including its five components. Conclusions: We have presented new genetic evidence demonstrating that MetS and its triglyceride components may serve as potential protective factors against thyroid autoimmunity.


Sujet(s)
Syndrome métabolique X , Humains , Syndrome métabolique X/génétique , Auto-immunité/génétique , Étude d'association pangénomique , Analyse de randomisation mendélienne , Glande thyroide
19.
Mol Biol Rep ; 51(1): 493, 2024 Apr 05.
Article de Anglais | MEDLINE | ID: mdl-38580818

RÉSUMÉ

Metabolic syndrome (MetS) is a prevalent and intricate health condition affecting a significant global population, characterized by a cluster of metabolic and hormonal disorders disrupting lipid and glucose metabolism pathways. Clinical manifestations encompass obesity, dyslipidemia, insulin resistance, and hypertension, contributing to heightened risks of diabetes and cardiovascular diseases. Existing medications often fall short in addressing the syndrome's multifaceted nature, leading to suboptimal treatment outcomes and potential long-term health risks. This scenario underscores the pressing need for innovative therapeutic approaches in MetS management. RNA-based treatments, employing small interfering RNAs (siRNAs), microRNAs (miRNAs), and antisense oligonucleotides (ASOs), emerge as promising strategies to target underlying biological abnormalities. However, a summary of research available on the role of RNA-based therapeutics in MetS and related co-morbidities is limited. Murine models and human studies have been separately interrogated to determine whether there have been recent advancements in RNA-based therapeutics to offer a comprehensive understanding of treatment available for MetS. In a narrative fashion, we searched for relevant articles pertaining to MetS co-morbidities such as cardiovascular disease, fatty liver disease, dementia, colorectal cancer, and endocrine abnormalities. We emphasize the urgency of exploring novel therapeutic avenues to address the intricate pathophysiology of MetS and underscore the potential of RNA-based treatments, coupled with advanced delivery systems, as a transformative approach for achieving more comprehensive and efficacious outcomes in MetS patients.


Sujet(s)
Maladies cardiovasculaires , Hypertension artérielle , Insulinorésistance , Syndrome métabolique X , microARN , Humains , Animaux , Souris , Syndrome métabolique X/génétique , Syndrome métabolique X/thérapie , Syndrome métabolique X/complications , Hypertension artérielle/complications , Obésité/complications , Maladies cardiovasculaires/complications , microARN/usage thérapeutique , Petit ARN interférent/génétique , Petit ARN interférent/usage thérapeutique
20.
Diabetes Obes Metab ; 26(7): 2839-2849, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38637979

RÉSUMÉ

AIM: To explore the link between the RBP4 rs3758539 genotype and metabolic syndrome risk factors and whether the impact of this genetic variation displays any potential race discrepancy. MATERIALS AND METHODS: This meta-analysis followed the PRISMA guidelines and was registered with PROSPERO (registration no. CRD42023407999). PubMed, Web of Science, Embase, Cochrane Library, Google Scholar, Airiti Library and CINAHL databases were used for the study search until October 2023. We evaluated the methodological quality using the Joanna Briggs Institute checklist and determined the correlation using a random-effects meta-analysis. RESULTS: The results indicated that individuals with the rs3758539 GA/AA genotype had a higher risk profile, including lower high-density lipoprotein levels [correlation: -0.045, 95% confidence interval (CI): -0.080 to -0.009, p = .015, I2 = 46.9%] and higher body mass index (correlation: 0.117, 95% CI: 0.036-0.197, p = .005, I2 = 82.0%), body fat (correlation: 0.098, 95% CI: 0.004-0.191, p = .041, I2 = 64.0%), and low-density lipoprotein levels (correlation: 0.074, 95% CI: 0.010-0.139, p = .024, I2 = 0%), of developing metabolic syndrome than those with the GG genotype. The subgroup analysis maintained a significantly positive correlation between the rs3758539 GA/AA genotype and body mass index (correlation: 0.163, 95% CI: 0.031-0.289, p = .016, I2 = 88.9%) but a negative correlation with high-density lipoprotein levels (correlation: -0.047, 95% CI: -0.087 to -0.006, p = .025, I2 = 65.7%) in the Asian group only. CONCLUSION: The current meta-analysis supports a significant link between the RBP4 rs3758539 GA/AA genotype and the metabolic syndrome.


Sujet(s)
Génotype , Syndrome métabolique X , Protéines plasmatiques de liaison au rétinol , Syndrome métabolique X/génétique , Humains , Protéines plasmatiques de liaison au rétinol/génétique , Polymorphisme de nucléotide simple , Prédisposition génétique à une maladie , Facteurs de risque , Indice de masse corporelle
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...