Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.193
Filter
1.
Front Endocrinol (Lausanne) ; 15: 1449344, 2024.
Article in English | MEDLINE | ID: mdl-39224121

ABSTRACT

Introduction: Degenerin proteins, such as ßENaC and ASIC2, have been implicated in cardiovascular function. However, their role in metabolic syndrome have not been studied. To begin to assess this interaction, we evaluated the impact of a high fat diet (HFD) on mice lacking normal levels of ASIC2 (ASIC2-/-) and ßENaC (ßENaCm/m). Methods: Twenty-week-old male and female mice were placed on a 60% HFD for 12 weeks. Body weight was measured weekly, and body composition by non-invasive ECHO MRI and fasting blood glucose were measured at 0, 4, 8 and 12 weeks. A glucose tolerance test was administered after 12 weeks. Differences between ASIC2-/-/ßENaCm/m and WT groups were compared using independent t-tests or ANOVA where appropriate within each sex. Data are presented as mean ± SEM and ASIC2-/-/ßENaCm/m vs. WT. Results: At 20 weeks of age, ASIC2-/-/ßENaCm/m mice (n=9F/10M) weighed less and gained less weight than WT (n=12F/16M). Total body fat and lean body masses were reduced in female and male ASIC2-/-/ßENaCm/m mice. Total body fat and lean body masses as % control were identical at the end of 12 weeks. Fasting blood glucoses were lower in female and male ASIC2-/-/ßENaCm/m vs. WT mice after 12 weeks HFD. The area under the curve for the glucose tolerance test was reduced in female and tended (p=.079) to decrease in male ASIC2-/-/ßENaCm/m. Plasma leptin and insulin were reduced in female and male ASIC2-/-/ßENaCm/m vs. WT mice. Plasma insulin in female ASIC2-/-/ßENaCm/m mice remained unchanged throughout the HFD period. Liver and liver fat masses, as well as percent liver fat, were reduced in both female and male ASIC2-/-/ßENaCm/m mice after HFD. Plasma triglycerides, cholesterol, LDL- and HDL-cholesterols were markedly improved in male and/or female ASIC2-/-/ßENaCm/m following the HFD. Discussion: These novel findings suggest that loss of ASIC2 and ßENaC offer a significant protection against HFD-induced metabolic syndrome.


Subject(s)
Acid Sensing Ion Channels , Diet, High-Fat , Metabolic Syndrome , Mice, Knockout , Animals , Diet, High-Fat/adverse effects , Metabolic Syndrome/metabolism , Metabolic Syndrome/etiology , Male , Mice , Female , Acid Sensing Ion Channels/metabolism , Acid Sensing Ion Channels/genetics , Body Composition , Mice, Inbred C57BL , Epithelial Sodium Channels/metabolism , Epithelial Sodium Channels/genetics , Blood Glucose/metabolism , Body Weight , Glucose Tolerance Test
2.
Sci Rep ; 14(1): 20665, 2024 09 05.
Article in English | MEDLINE | ID: mdl-39237601

ABSTRACT

Cardiovascular-kidney-metabolic health reflects the interactions between metabolic risk factors, chronic kidney disease, and the cardiovascular system. A growing body of literature suggests that metabolic syndrome (MetS) in individuals of normal weight is associated with a high prevalence of cardiovascular diseases and an increased mortality. The aim of this study was to establish a non-invasive preclinical model of MetS in support of future research focusing on the effects of novel antidiabetic therapies beyond glucose reduction, independent of obesity. Eighteen healthy adult Beagle dogs were fed an isocaloric Western diet (WD) for ten weeks. Biospecimens were collected at baseline (BAS1) and after ten weeks of WD feeding (BAS2) for measurement of blood pressure (BP), serum chemistry, lipoprotein profiling, blood glucose, glucagon, insulin secretion, NT-proBNP, angiotensins, oxidative stress biomarkers, serum, urine, and fecal metabolomics. Differences between BAS1 and BAS2 were analyzed using non-parametric Wilcoxon signed-rank testing. The isocaloric WD model induced significant variations in several markers of MetS, including elevated BP, increased glucose concentrations, and reduced HDL-cholesterol. It also caused an increase in circulating NT-proBNP levels, a decrease in serum bicarbonate, and significant changes in general metabolism, lipids, and biogenic amines. Short-term, isocaloric feeding with a WD in dogs replicated key biological features of MetS while also causing low-grade metabolic acidosis and elevating natriuretic peptides. These findings support the use of the WD canine model for studying the metabolic effects of new antidiabetic therapies independent of obesity.


Subject(s)
Disease Models, Animal , Hypoglycemic Agents , Metabolic Syndrome , Obesity , Animals , Dogs , Metabolic Syndrome/drug therapy , Metabolic Syndrome/metabolism , Obesity/metabolism , Obesity/drug therapy , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Male , Blood Glucose/metabolism , Biomarkers/blood , Blood Pressure/drug effects , Natriuretic Peptide, Brain/blood , Natriuretic Peptide, Brain/metabolism , Oxidative Stress/drug effects , Female
3.
J Exp Med ; 221(9)2024 09 02.
Article in English | MEDLINE | ID: mdl-39133222

ABSTRACT

The retinoid nuclear receptor pathway, activated by the vitamin A metabolite retinoic acid, has been extensively investigated for over a century. This study has resulted in conflicting hypotheses about how the pathway regulates health and how it should be pharmaceutically manipulated. These disagreements arise from a fundamental contradiction: retinoid agonists offer clear benefits to select patients with rare bone growth disorders, acute promyelocytic leukemia, and some dermatologic diseases, yet therapeutic retinoid pathway activation frequently causes more harm than good, both through acute metabolic dysregulation and a delayed cancer-promoting effect. In this review, we discuss controlled clinical, mechanistic, and genetic data to suggest several disease settings where inhibition of the retinoid pathway may be a compelling therapeutic strategy, such as solid cancers or metabolic syndromes, and also caution against continued testing of retinoid agonists in cancer patients. Considerable evidence suggests a central role for retinoid regulation of immunity and metabolism, with therapeutic opportunities to antagonize retinoid signaling proposed in cancer, diabetes, and obesity.


Subject(s)
Metabolic Syndrome , Neoplasms , Signal Transduction , Humans , Neoplasms/metabolism , Animals , Metabolic Syndrome/metabolism , Receptors, Retinoic Acid/metabolism , Retinoids/metabolism
5.
Gut Microbes ; 16(1): 2390136, 2024.
Article in English | MEDLINE | ID: mdl-39163273

ABSTRACT

Abdominal obesity-related metabolic syndrome (MetS) has emerged as a significant global public health issue that affects human health. Flavonoids, such as quercetin, have been reported to exert obvious anti-obesity and lipid-lowering effects in both humans and animal models. However, the precise underlying mechanism remains elusive. In this study, we investigated the potential roles of gut microbiota-bile acids (BAs) interactions in quercetin-induced anti-obesity effects and metabolic benefits. Oral administration of quercetin significantly enhanced energy metabolism through activating thermogenesis of brown adipose tissues (BAT) and browning of white adipose tissues (WAT), thus mitigating metabolic dysfunctions in an abdominal obesity-related MetS mouse model. Further mechanistic studies demonstrated that quercetin treatment substantially promoted the generation of non-12α-hydroxylated BAs (non-12OH BAs), particularly ursodeoxycholic acid (UDCA) and lithocholic acid (LCA), in serum via regulating the overall structure of gut microbiota and enriching Lactobacillus. High level of non-12OH BAs bind to Takeda G protein-coupled receptor 5 (TGR5) on adipocytes to stimulate thermogenesis. Remarkably, fecal microbiota transplantation (FMT) from quercetin-treated mice replicated the effects of quercetin on non-12OH BAs generation and energy expenditure, which suggested gut microbiota reshape and concomitant BAs regulation were responsible for the benefits on energy metabolism of quercetin in the MetS mouse model. Our findings not only highlighted the critical role of gut microbiota-BAs crosstalk in mediating quercetin-induced energy expenditure, but also enriched the pharmacological mechanisms of quercetin in ameliorating MetS-related diseases.


Subject(s)
Adipose Tissue, Brown , Bile Acids and Salts , Energy Metabolism , Gastrointestinal Microbiome , Metabolic Syndrome , Mice, Inbred C57BL , Quercetin , Thermogenesis , Quercetin/pharmacology , Animals , Gastrointestinal Microbiome/drug effects , Mice , Energy Metabolism/drug effects , Metabolic Syndrome/metabolism , Metabolic Syndrome/microbiology , Metabolic Syndrome/drug therapy , Male , Bile Acids and Salts/metabolism , Thermogenesis/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/drug effects , Adipose Tissue, White/metabolism , Adipose Tissue, White/drug effects , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Disease Models, Animal , Fecal Microbiota Transplantation
6.
BMC Endocr Disord ; 24(1): 141, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39103858

ABSTRACT

BACKGROUND: Numerous studies have revealed the role of dietary fatty acids in human health. However, few studies have evaluated dietary fatty acid patterns and their association with metabolic parameters. The current study aimed to explore the association between dietary fatty acid patterns and risk factors for metabolic syndrome (MetS) among overweight and obese adults. METHODS: This cross-sectional study involved 340 participants who were overweight or obese. The study included assessments of body composition and anthropometric measurements. Dietary fatty acid consumption was evaluated using a validated Food Frequency Questionnaire (FFQ) containing 168 items. Additionally, biochemical parameters, including serum total cholesterol (TC), triglyceride (TG), high-density lipoprotein cholesterol (HDL-C), fasting serum glucose (FSG), and insulin levels, were measured using enzymatic methods. Fatty acid patterns were determined by principal component analysis (PCA), and the association between these dietary FA patterns and risk factors related to MetS components was assessed using logistic regression. RESULTS: Factor analysis conducted in this study explored three dietary fatty acid patterns: saturated fatty acids (SFA), polyunsaturated fatty acids (PUFA), and long-chain combined fatty acids (LC-CFA). Those at the highest tertile of the SFA pattern had lower diastolic blood pressure (DBP) (P = 0.03). Low-density lipoprotein cholesterol (LDL) was lower in the second and third tertiles (P ≤ 0.05). Also, higher fasting blood glucose (FBS) was observed in the second and third tertiles (P < 0.05), and the homeostatic model assessment of insulin resistance (HOMA-IR) was higher in the third tertile (P = 0.049). In the PUFA pattern, FBS was lower in the third tertile (P = 0.03). In the LC-CFA pattern, lower TC was achieved in higher tertiles (P = 0.04). CONCLUSION: Our findings demonstrated that consuming high and moderate SFA patterns is associated with higher FBS and HOMA-IR. Also, increased consumption of SCFAs is related to lower DPB and LDL. Individuals who consumed more PUFA, especially linoleic acid, had lower FBS. These outcomes might be beneficial in managing MetS and leading to a new field of research.


Subject(s)
Dietary Fats , Fatty Acids , Metabolic Syndrome , Obesity , Overweight , Humans , Male , Female , Cross-Sectional Studies , Adult , Obesity/metabolism , Overweight/metabolism , Middle Aged , Fatty Acids/metabolism , Metabolic Syndrome/metabolism , Metabolic Syndrome/epidemiology , Metabolic Syndrome/etiology , Dietary Fats/metabolism , Risk Factors , Metabolome , Biomarkers/blood , Biomarkers/analysis
7.
Front Endocrinol (Lausanne) ; 15: 1370578, 2024.
Article in English | MEDLINE | ID: mdl-39109080

ABSTRACT

Objective: Polycystic ovary syndrome (PCOS) is an endocrine metabolic disorder in reproductive-aged women. The study was designed to investigate the metabolic characteristics of different phenotypes in women with PCOS of reproductive age. Methods: A total of 442 women with PCOS were recruited in this cross-sectional study. According to different phenotypes, all women were divided into three groups: the chronic ovulatory dysfunction and hyperandrogenism group (OD-HA group, n = 138), the chronic ovulatory dysfunction and polycystic ovarian morphology group (OD-PCOM group, n = 161), and the hyperandrogenism and polycystic ovarian morphology group (HA-PCOM group, n = 143). The metabolic risk factors and prevalence rates of metabolic disorders among the three groups were compared. Results: The body mass index (BMI), waist circumference, and waist-to-hip ratio (WHR) of women from the OD-HA group and HA-PCOM group were significantly higher than those of women from the OD-PCOM group (p < 0.05). The serum insulin concentration and homeostasis model assessment of insulin resistance (HOMA IR) at 2 h and 3 h after oral glucose powder in women from the OD-HA group and HA-PCOM group were significantly higher than those from the OD-PCOM group (p < 0.05). The serum total cholesterol (TC), triglyceride (TG), and low-density lipoprotein cholesterol (LDL-C) in women from the OD-HA group and HA-PCOM group were significantly higher than those in women from the OD-PCOM group (p < 0.05). The prevalence rates of impaired glucose tolerance (IGT), type 2 diabetes mellitus (T2DM), insulin resistance (IR), metabolic syndrome (MS), nonalcoholic fatty liver disease (NAFLD), and dyslipidemia of women with PCOS were 17.9%, 3.6%, 58.4%, 29.4%, 46.6%, and 43.4%, respectively. The prevalence rates of IGT, IR, MS, NAFLD, and dyslipidemia of women in the OD-HA group and HA-PCOM group were significantly higher than those of women in the OD-PCOM group (p < 0.05). T concentration (>1.67 nmol/L) and Ferriman-Gallwey (F-G) score (>3) significantly increased the risk of metabolic disorders in women with PCOS (p < 0.05). Conclusion: The phenotypes of OD-HA and HA-PCOM in women with PCOS were vulnerable to metabolic disorders compared to OD-PCOM. Thus, the metabolic disorders in women with PCOS especially those with the HA phenotype should be paid more attention in order to reduce long-term complications.


Subject(s)
Insulin Resistance , Phenotype , Polycystic Ovary Syndrome , Humans , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/epidemiology , Female , Adult , Cross-Sectional Studies , Young Adult , Body Mass Index , Hyperandrogenism/complications , Hyperandrogenism/epidemiology , Hyperandrogenism/metabolism , Risk Factors , Waist-Hip Ratio , Metabolic Syndrome/epidemiology , Metabolic Syndrome/metabolism , Metabolic Syndrome/complications , Waist Circumference , Reproduction/physiology
8.
Life Sci ; 355: 123009, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39197574

ABSTRACT

Adipose tissue dysfunction is a key feature of metabolic syndrome, which increases the risk of periodontitis, an inflammatory disease induced by bacteria that affects the gingiva and other components of periodontal tissue. Recent studies indicate that molecules from inflamed periodontal tissue contribute to adipose tissue dysfunction. However, the cellular mechanisms and interactions between adipose tissue and gingiva driving the progression of metabolic and periodontal conditions remain unclear. To address this, we developed a chimeric (mouse/human) co-culture tissue model (which identifies the origins of species-specific cytokines) to investigate these interactions. Using tissue-specific functional cells and immunocytes, we constructed equivalents of adipose tissue (ATE) and gingiva (GTE), co-cultivating them under inflammatory conditions induced by bacterial endotoxin, lipopolysaccharide (LPS). Our findings showed that exposure to LPS resulted in a notable reduction in lipid accumulation, GLUT4 expression, and adiponectin secretion in ATE, along with increased macrophage colonies forming around lipid droplets, as well as elevated levels of triglyceride, leptin, and IL-6. In GTE, LPS triggered significant inflammatory responses, characterized by increased macrophage accumulation, elevated COX-2 expression, and heightened secretion of inflammatory cytokines. LPS also reduced epithelial thickness and the expression of keratin 19 and collagen IV, indicating impaired barrier function and gingival integrity. Co-culturing ATE with GTE exacerbated these LPS-induced harmful effects in both tissues. In conclusion, our findings suggest that interplay between gingiva and adipose tissue can intensify the inflammatory and dysfunctional changes caused by LPS. This co-culture tissue model offers a valuable tool for future studies on periodontitis and metabolic syndrome.


Subject(s)
Adipose Tissue , Coculture Techniques , Gingiva , Inflammation , Lipopolysaccharides , Gingiva/metabolism , Gingiva/pathology , Animals , Adipose Tissue/metabolism , Humans , Mice , Inflammation/metabolism , Inflammation/pathology , Periodontitis/metabolism , Periodontitis/pathology , Cytokines/metabolism , Mice, Inbred C57BL , Macrophages/metabolism , Male , Metabolic Syndrome/metabolism
9.
Adv Clin Chem ; 123: 129-156, 2024.
Article in English | MEDLINE | ID: mdl-39181620

ABSTRACT

Cortisol, a stress hormone, plays a crucial role in regulating metabolic, hemodynamic, inflammatory, and behavioral processes. Its secretion is governed by the hypothalamic-pituitary-adrenal axis. However, prolonged activation of this axis and increased cortisol bioavailability in tissues can result in detrimental metabolic effects. Chronic exposure to excessive cortisol is associated with insulin resistance and visceral obesity, both significant contributors to metabolic syndrome. This review delves into the regulation of the hypothalamic-pituitary-adrenal axis, the molecular mechanisms underlying cortisol synthesis and its actions, as well as the key factors influencing cortisol bioavailability. Furthermore, it provides a summary of available clinical research data on the involvement of cortisol in metabolic syndrome, alongside a discussion on the various biomatrices used for cortisol measurement in clinical settings.


Subject(s)
Hydrocortisone , Hypothalamo-Hypophyseal System , Metabolic Syndrome , Humans , Metabolic Syndrome/metabolism , Hydrocortisone/metabolism , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism
10.
Sci Rep ; 14(1): 18970, 2024 08 16.
Article in English | MEDLINE | ID: mdl-39152139

ABSTRACT

Mitochondrial dysfunction, characterized by elevated oxidative stress, impaired energy balance, and dysregulated mitochondrial dynamics, is a hallmark of metabolic syndrome (MetS) and its comorbidities. Ferulic acid (FA), a principal phenolic compound found in whole grains, has demonstrated potential in ameliorating oxidative stress and preserving energy homeostasis. However, the influence of FA on mitochondrial health within the context of MetS remains unexplored. Moreover, the impact of FA on autophagy, which is essential for maintaining energy homeostasis and mitochondrial integrity, is not fully understood. Here, we aimed to study the mechanisms of action of FA in regulating mitochondrial health and autophagy using palmitate-treated HepG2 hepatocytes as a MetS cell model. We found that FA improved mitochondrial health by restoring redox balance and optimizing mitochondrial dynamics, including biogenesis and the fusion/fission ratio. Additionally, FA was shown to recover autophagy and activate AMPK-related cell signaling. Our results provide new insights into the therapeutic potential of FA as a mitochondria-targeting agent for the prevention and treatment of MetS.


Subject(s)
AMP-Activated Protein Kinases , Autophagy , Coumaric Acids , Hepatocytes , Metabolic Syndrome , Mitochondrial Dynamics , Signal Transduction , Coumaric Acids/pharmacology , Autophagy/drug effects , Humans , Metabolic Syndrome/metabolism , Metabolic Syndrome/drug therapy , Metabolic Syndrome/pathology , Mitochondrial Dynamics/drug effects , Signal Transduction/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , AMP-Activated Protein Kinases/metabolism , Hep G2 Cells , Palmitates/pharmacology , Palmitates/toxicity , Mitochondria/metabolism , Mitochondria/drug effects , Oxidative Stress/drug effects
11.
Environ Int ; 190: 108929, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39098089

ABSTRACT

The global prevalence of Metabolic Syndrome (MetS) is continuously rising and exposure to environmental toxicants such as arsenic could be contributing to this rapid surge. In this study, we have assessed the effects of prenatal arsenic exposure on insulin resistance and MetS parameters in a mouse model, and an underlying mechanism was identified. We found that prenatal arsenic exposure promotes insulin resistance and adipocyte dysfunction which leads to the early onset of MetS in male offspring. Primary adipocytes isolated from 20-week-old arsenic-exposed offspring showed hypertrophy, elevated basal lipolysis, and impaired insulin response along with enhanced expression of Tumor necrosis factor-alpha (TNF-α). TNF-α levels were consistently high at gestational day 15.5 (GD15.5) as well as primary adipocytes of 6-week-old arsenic-exposed male offspring. Along with TNF-α, downstream p-JNK1/2 levels were also increased, which led to inhibitory phosphorylation of IRS1and reduced GLUT4 translocation upon insulin stimulation in adipocytes. Insulin response and downstream signaling were restored upon TNF-α inhibition, confirming its central role. The persistent overexpression of TNF-α in adipocytes of arsenic-exposed mice resulted from diminished EZH2 occupancy and reduced H3K27me3 (gene silencing histone marks) at the TNF-α promoter. This further led to chromatin relaxation, recruitment of c-Jun and CBP/p300, formation of an enhanceosome complex, and TNF-α expression. Our findings show how prenatal arsenic exposure can epigenetically modulate TNF-α expression to promote adipocyte dysfunction and insulin resistance which contributes to the early onset of MetS in offspring.


Subject(s)
Arsenic , Enhancer of Zeste Homolog 2 Protein , Insulin Resistance , Metabolic Syndrome , Prenatal Exposure Delayed Effects , Tumor Necrosis Factor-alpha , Animals , Metabolic Syndrome/chemically induced , Metabolic Syndrome/metabolism , Tumor Necrosis Factor-alpha/metabolism , Mice , Female , Arsenic/toxicity , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Male , Enhancer of Zeste Homolog 2 Protein/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Disease Models, Animal , Histones/metabolism , Promoter Regions, Genetic , Adipocytes/drug effects , Adipocytes/metabolism
13.
Lipids Health Dis ; 23(1): 273, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39198792

ABSTRACT

INTRODUCTION: We previously documented the beneficial effects of rice bran oil (RBO) on cardiac function and atherogenic cardiometabolic factors in men with coronary artery disease. Therefore, the existing evidence in this area aims to be expanded by investigating the impact of adding RBO to a daily standard diet on emerging insulin resistance surrogate markers, lipid peroxidation, antioxidant status, and metabolic disturbances in individuals with metabolic syndrome (MetSyn) through an open-label controlled trial. METHODS: A total of 50 overweight/obese adults (mean body mass index (BMI) = 31.08 kg/m2) with at least 3 MetSyn components were randomly allocated to either the control group, which received a standard diet plan, or the intervention group, which was supplemented with 30 g/d RBO for 8 weeks. BMI, MetSyn components, metabolic score for insulin resistance (METS-IR), triglyceride‒glucose‒BMI (TyG‒BMI), malondialdehyde (MDA), total antioxidant capacity (TAC), and plasma polyphenol levels were measured before and after this open-label trial. RESULTS: Analysis of covariance (ANCOVA) adjusted for baseline values revealed that, compared with patients who received only a standard diet, those who were supplemented with 30 g/d RBO presented significantly lower total cholesterol (P value = 0.005; effect size (ES):-0.92), LDL-cholesterol (P value = 0.048; ES:-0.62), fasting blood glucose (P value = 0.014; ES:-0.77), MDA (P value = 0.002; ES: -1.01), METS-IR (P value < 0.001; ES: -1.24), and TyG-BMI (P value = 0.007; ES:-0.85) after 8 weeks. Additionally, RBO consumption resulted in significantly higher levels of HDL-C (P value = 0.004; ES:0.94) and TAC (P value < 0.0001; ES:2.05). However, no significant changes were noted in BMI, waist circumference, serum triglycerides, plasma polyphenols, or blood pressure. CONCLUSION: Although the current findings suggest that the hypocholesterolemic, antihyperglycemic, and antioxidative effects of 30 g/d RBO seem to be promising for MetSyn patients, they should be considered preliminary. Therefore, further well-designed clinical trials with larger sample sizes and longer durations are needed to confirm these findings.


Subject(s)
Antioxidants , Cardiometabolic Risk Factors , Lipid Peroxidation , Metabolic Syndrome , Obesity , Overweight , Rice Bran Oil , Humans , Metabolic Syndrome/diet therapy , Metabolic Syndrome/blood , Metabolic Syndrome/drug therapy , Metabolic Syndrome/metabolism , Male , Middle Aged , Antioxidants/metabolism , Obesity/diet therapy , Obesity/blood , Obesity/drug therapy , Obesity/metabolism , Lipid Peroxidation/drug effects , Adult , Overweight/diet therapy , Overweight/blood , Female , Body Mass Index , Insulin Resistance , Malondialdehyde/blood , Triglycerides/blood , Blood Glucose/metabolism
14.
Signal Transduct Target Ther ; 9(1): 212, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39191722

ABSTRACT

Hyperuricemia, characterized by elevated levels of serum uric acid (SUA), is linked to a spectrum of commodities such as gout, cardiovascular diseases, renal disorders, metabolic syndrome, and diabetes, etc. Significantly impairing the quality of life for those affected, the prevalence of hyperuricemia is an upward trend globally, especially in most developed countries. UA possesses a multifaceted role, such as antioxidant, pro-oxidative, pro-inflammatory, nitric oxide modulating, anti-aging, and immune effects, which are significant in both physiological and pathological contexts. The equilibrium of circulating urate levels hinges on the interplay between production and excretion, a delicate balance orchestrated by urate transporter functions across various epithelial tissues and cell types. While existing research has identified hyperuricemia involvement in numerous biological processes and signaling pathways, the precise mechanisms connecting elevated UA levels to disease etiology remain to be fully elucidated. In addition, the influence of genetic susceptibilities and environmental determinants on hyperuricemia calls for a detailed and nuanced examination. This review compiles data from global epidemiological studies and clinical practices, exploring the physiological processes and the genetic foundations of urate transporters in depth. Furthermore, we uncover the complex mechanisms by which the UA induced inflammation influences metabolic processes in individuals with hyperuricemia and the association with its relative disease, offering a foundation for innovative therapeutic approaches and advanced pharmacological strategies.


Subject(s)
Hyperuricemia , Uric Acid , Hyperuricemia/genetics , Humans , Uric Acid/metabolism , Uric Acid/blood , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Gout/genetics , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Metabolic Syndrome/genetics , Metabolic Syndrome/metabolism
15.
Front Endocrinol (Lausanne) ; 15: 1442740, 2024.
Article in English | MEDLINE | ID: mdl-39165513

ABSTRACT

Background: Obesity-induced metabolic dysfunction increases the risk of developing tumors, however, the relationship between metabolic obesity phenotypes and prostate cancer (PCa) remains unclear. Methods: The term metabolic obesity phenotypes was introduced based on metabolic status and BMI categories. Participants were categorized into four groups: metabolically healthy nonobesity (MHNO), metabolically healthy obesity (MHO), metabolically unhealthy nonobesity (MUNO), and metabolically unhealthy obesity (MUO). Propensity score matching was conducted based on age, ethnicity, marriage, etc. Univariate and multivariate conditional logistic regression analyses were used to assess the relationship between metabolic obesity phenotypes, metabolic risk factors, and PCa. Sensitivity analysis was performed to verify the robustness of the results. Results: After propensity score matching among 564 PCa patients and 1418 healthy individuals, 209 were selected for each of the case and control groups. There were no statistically significant differences in the basic characteristics between the two groups. Univariate and multivariate conditional logistic regression suggested that the risk of developing PCa in both MHO and MUO individuals was higher than in MHNO individuals. Specifically, the risk of developing PCa in MHO individuals was 2.166 times higher than in MHNO individuals (OR=2.166, 95%CI: 1.133-4.139), and the risk in MUO individuals was is 2.398 times higher than in MHNO individuals(OR=2.398, 95%CI:1.271-4.523). Individuals with hyperglycemia and elevated triglycerides also had a higher risk of developing PCa (hyperglycemia:OR=1.488, 95%CI: 1.001-2.210; elevated triglycerides: OR=2.292, 95%CI: 1.419-3.702). Those with more than or equal to three metabolic risk factors had an increased risk of PCa (OR=1.990, 95%CI: 1.166-3.396). Sensitivity analysis indicated an increased risk of PCa in MUO individuals compared to MHNO individuals. Conclusion: In this retrospective study, individuals with MHO and MUO had a higher risk of developing PCa.


Subject(s)
Obesity , Phenotype , Propensity Score , Prostatic Neoplasms , Humans , Male , Prostatic Neoplasms/epidemiology , Prostatic Neoplasms/metabolism , Middle Aged , Obesity/complications , Obesity/metabolism , China/epidemiology , Risk Factors , Aged , Case-Control Studies , Body Mass Index , Metabolic Syndrome/complications , Metabolic Syndrome/epidemiology , Metabolic Syndrome/metabolism
16.
Life Sci ; 355: 122967, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39142504

ABSTRACT

Olfactory dysfunction, influenced by factors such as aging and environmental stress, is linked to various neurological disorders. The olfactory bulb's connections to brain areas like the hypothalamus, piriform cortex, entorhinal cortex, and limbic system make olfactory dysfunction a contributor to a range of neuropathological conditions. Recent research has underscored that olfactory deficits are prevalent in individuals with both metabolic syndrome and dementia. These systemic metabolic alterations correlate with olfactory impairments, potentially affecting brain regions associated with the olfactory bulb. In cases of metabolic syndrome, phenomena such as insulin resistance and disrupted glucose metabolism may result in compromised olfactory function, leading to multiple neurological issues. This review synthesizes key findings on the interplay between metabolic-induced olfactory dysfunction and neuropathology. It emphasizes the critical role of olfactory assessment in diagnosing and managing neurological diseases related to metabolic syndrome.


Subject(s)
Metabolic Syndrome , Olfactory Bulb , Humans , Metabolic Syndrome/metabolism , Olfactory Bulb/metabolism , Olfactory Bulb/pathology , Olfaction Disorders/metabolism , Olfaction Disorders/etiology , Olfaction Disorders/physiopathology , Animals , Nervous System Diseases/metabolism , Nervous System Diseases/pathology
17.
Eur Rev Med Pharmacol Sci ; 28(15): 4046-4059, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39194200

ABSTRACT

OBJECTIVE: A metabolism score for visceral fat (METS-VF) is an innovative method to access abdominal fat and visceral fat. So far, the relationship between the METS-VF index and chronic obstructive pulmonary disease (COPD) has remained unclear. We investigated the relationship between the METS-VF index and COPD prevalence utilizing data from the National Health and Nutrition Examination Survey (NHANES) 2007-2018. PATIENTS AND METHODS: A binary logistic regression analysis was performed using NHANES 2007-2018 data to assess the relationship between the METS-VF index and COPD prevalence. The relationship was verified by fitted smooth curves, generalized additive models, threshold effect analyses, subgroup analyses, and sensitivity analyses. RESULTS: In total, 7,680 subjects were recruited for the study, including 772 self-reported having COPD. The METS-VF index was positively related to COPD prevalence when adjusted for all covariates. The METS-VF index was classified by quartiles, and participants who scored highest on METS-VF were at a greater risk of COPD than those who scored lowest. According to a threshold effect analysis, the METS-VF index was negatively correlated with COPD prevalence with a METS-VF index <7.00, without statistical significance. Once the METS-VF index exceeded 7.00, there was a robust positive correlation between the METS-VF index and COPD prevalence. In the analysis of subgroups, the METS-VF index was positively correlated with COPD prevalence among subjects who were male, aged 40-59, and without asthma or hypertension. The results were robust in sensitivity analyses. METS-VF showed a significantly better diagnostic value for COPD than Body Mass Index (BMI). CONCLUSIONS: The METS-VF index has a non-linear and positive correlation with COPD prevalence in the middle-aged and elderly American population.


Subject(s)
Intra-Abdominal Fat , Nutrition Surveys , Pulmonary Disease, Chronic Obstructive , Humans , Pulmonary Disease, Chronic Obstructive/epidemiology , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/diagnosis , Middle Aged , Male , Intra-Abdominal Fat/metabolism , Female , Aged , United States/epidemiology , Prevalence , Metabolic Syndrome/epidemiology , Metabolic Syndrome/metabolism , Metabolic Syndrome/diagnosis
18.
Bull Exp Biol Med ; 177(3): 395-400, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39134815

ABSTRACT

We compared 2 models of metabolic syndrome in rats: high-fat diet (58% calories) with single streptozotocin injection at a dose of 25 mg/kg and replacement of water with 20% fructose solution. The model with fructose solution did not cause the main signs of metabolic syndrome over 24 weeks: concentrations of glucose, triglycerides, cholesterol, weight, and BP did not significantly differ from the control group (standard diet). At the same time, single streptozotocin administration was followed by the development of persistent hyperglycemia, hypertriglyceridemia, hypercholesterolemia, and signs of visceral obesity. High-fat diet combined with injection of streptozotocin in a low dose can be considered a more representative model of metabolic syndrome in humans.


Subject(s)
Blood Glucose , Diet, High-Fat , Metabolic Syndrome , Streptozocin , Triglycerides , Animals , Diet, High-Fat/adverse effects , Rats , Male , Metabolic Syndrome/metabolism , Triglycerides/blood , Triglycerides/metabolism , Blood Glucose/metabolism , Rats, Wistar , Hyperglycemia/metabolism , Hyperglycemia/chemically induced , Cholesterol/blood , Cholesterol/metabolism , Body Weight/drug effects , Fructose/administration & dosage , Hypertriglyceridemia/metabolism , Hypertriglyceridemia/chemically induced , Hypertriglyceridemia/blood , Hypertriglyceridemia/etiology , Hypercholesterolemia/metabolism , Hypercholesterolemia/etiology , Dietary Carbohydrates/administration & dosage , Blood Pressure/drug effects
19.
Physiol Res ; 73(S1): S165-S183, 2024 08 30.
Article in English | MEDLINE | ID: mdl-39212142

ABSTRACT

Metabolomics and lipidomics have emerged as tools in understanding the connections of metabolic syndrome (MetS) with cardiovascular diseases (CVD), type 1 and type 2 diabetes (T1D, T2D), and metabolic dysfunction-associated steatotic liver disease (MASLD). This review highlights the applications of these omics approaches in large-scale cohort studies, emphasizing their role in biomarker discovery and disease prediction. Integrating metabolomics and lipidomics has significantly advanced our understanding of MetS pathology by identifying unique metabolic signatures associated with disease progression. However, challenges such as standardizing analytical workflows, data interpretation, and biomarker validation remain critical for translating research findings into clinical practice. Future research should focus on optimizing these methodologies to enhance their clinical utility and address the global burden of MetS-related diseases.


Subject(s)
Cardiovascular Diseases , Diabetes Mellitus, Type 2 , Lipidomics , Metabolic Syndrome , Metabolomics , Humans , Metabolic Syndrome/metabolism , Metabolomics/methods , Lipidomics/methods , Diabetes Mellitus, Type 2/metabolism , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/diagnosis , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/complications , Animals , Biomarkers/metabolism , Fatty Liver/metabolism
20.
Int J Mol Sci ; 25(16)2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39201418

ABSTRACT

Type 2 diabetes mellitus (T2DM), often featuring hyperglycemia or insulin resistance, is a global health concern that is increasing in prevalence in the United States and worldwide. A common complication is metabolic dysfunction-associated steatotic liver disease (MASLD), the hepatic manifestation of metabolic syndrome that is also rapidly increasing in prevalence. The majority of patients with T2DM will experience MASLD, and likewise, individuals with MASLD are at an increased risk for developing T2DM. These two disorders may act synergistically, in part due to increased lipotoxicity and inflammation within the liver, among other causes. However, the pathophysiological mechanisms by which this occurs are unclear, as is how the improvement of one disorder can ameliorate the other. This review aims to discuss the pathogenic interactions between T2D and MASLD, and will highlight novel therapeutic targets and ongoing clinical trials for the treatment of these diseases.


Subject(s)
Diabetes Mellitus, Type 2 , Humans , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/complications , Animals , Metabolic Syndrome/metabolism , Metabolic Syndrome/complications , Insulin Resistance , Fatty Liver/metabolism , Fatty Liver/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/complications
SELECTION OF CITATIONS
SEARCH DETAIL