Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.177
Filter
1.
Physiol Rep ; 12(13): e16097, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38955666

ABSTRACT

Latent associations between low serum amylase and reduced plasma insulin levels and increased adiposity have been described previously in a small study of asymptomatic middle-aged humans. In the present study, we sought to determine the nature of such changes during the longitudinal progression from metabolically normal to overt type 2 diabetes mellitus (T2DM) in nonhuman primates (NHPs), a disease that appears to be the same in both pathophysiology and underlying mechanisms as that which most commonly develops in middle-aged adult humans. Amylase and lipase levels were characterized in 157 unrelated adult rhesus monkeys (Macaca mulatta); 38% developed T2DM while under study. In all monkeys, multivariable linear regression analysis revealed that amylase could be negatively predicted by % body fat (ß -0.29; p = 0.002), age (ß -0.27; p = 0.005), and HbA1c (ß -0.18; p = 0.037). Amylase levels were positively predicted by lipase levels (ß = 0.19; p = -0.024) in all NHPs included in the study. Amylase was significantly lower in NHPs with metabolic syndrome (p < 0.001), prediabetes (PreDM) (p < 0.001), and T2DM (p < 0.001) compared to metabolically normal adult NHPs. Lipase increased in NHPs with PreDM (p = 0.005) and T2DM (p = 0.04) compared to normal NHPs. This is the first longitudinal study of any species, including humans, to show the dynamics of amylase and lipase during the metabolic progression from normal to metabolic syndrome, to PreDM and then to overt T2DM. The extraordinary similarity between humans and monkeys in T2DM, in pancreatic pathophysiology and in metabolic functions give these findings high translational value.


Subject(s)
Amylases , Diabetes Mellitus, Type 2 , Lipase , Macaca mulatta , Metabolic Syndrome , Animals , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Lipase/blood , Lipase/metabolism , Male , Metabolic Syndrome/blood , Metabolic Syndrome/metabolism , Longitudinal Studies , Amylases/blood , Amylases/metabolism , Female
2.
Front Endocrinol (Lausanne) ; 15: 1272886, 2024.
Article in English | MEDLINE | ID: mdl-38989003

ABSTRACT

Background: Obesity is associated with impaired glucose metabolism and hepatic insulin resistance. The aim was to investigate the associations of hepatic glucose uptake (HGU) and endogenous glucose production (EGP) to sedentary behavior (SB), physical activity (PA), cardiorespiratory fitness, dietary factors, and metabolic risk markers. Methods: Forty-four adults with metabolic syndrome (mean age 58 [SD 7] years, BMI ranging from 25-40kg/; 25 females) were included. HGU was measured by positron emission tomography during the hyperinsulinemic-euglycemic clamp. EGP was calculated by subtracting the glucose infusion rate during clamp from the glucose rate of disappearance. SB and PA were measured with hip-worn accelerometers (26 [SD3] days). Fitness was assessed by maximal bicycle ergometry with respiratory gas measurements and dietary intake of nutrients by 4-day food diaries. Results: HGU was not associated with fitness or any of the SB or PA measures. When adjusted for sex, age, and body fat-%, HGU was associated with whole-body insulin sensitivity (ß=0.58), water-insoluble dietary fiber (ß=0.29), energy percent (E%) of carbohydrates (ß=-0.32), saccharose (ß=-0.32), mono- and polyunsaturated fatty acids (ß=0.35, ß=0.41, respectively). EGP was associated with whole-body insulin sensitivity (ß=-0.53), and low-density lipoprotein cholesterol [ß=-0.31], and when further adjusted for accelerometry wear time, EGP was associated with standing [ß=-0.43]. (p-value for all< 0.05). Conclusions: Standing more, consuming a diet rich in fiber and unsaturated fatty acids, and a lower intake of carbohydrates, especially sugar, associate beneficially with hepatic insulin sensitivity. Habitual SB, PA, or fitness may not be the primary modulators of HGU and EGP. However, these associations need to be confirmed with intervention studies.


Subject(s)
Dietary Fiber , Fatty Acids, Unsaturated , Insulin Resistance , Liver , Metabolic Syndrome , Sedentary Behavior , Humans , Female , Male , Middle Aged , Metabolic Syndrome/metabolism , Dietary Fiber/administration & dosage , Liver/metabolism , Fatty Acids, Unsaturated/metabolism , Fatty Acids, Unsaturated/administration & dosage , Standing Position , Exercise , Aged , Adult , Glucose Clamp Technique , Cardiorespiratory Fitness/physiology
3.
World J Gastroenterol ; 30(23): 2964-2980, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38946874

ABSTRACT

Metabolic dysfunction-associated fatty liver disease (MAFLD) is a hepatic manifestation of the metabolic syndrome. It is one of the most common liver diseases worldwide and shows increasing prevalence rates in most countries. MAFLD is a progressive disease with the most severe cases presenting as advanced fibrosis or cirrhosis with an increased risk of hepatocellular carcinoma. Gut microbiota play a significant role in the pathogenesis and progression of MAFLD by disrupting the gut-liver axis. The mechanisms involved in maintaining gut-liver axis homeostasis are complex. One critical aspect involves preserving an appropriate intestinal barrier permeability and levels of intestinal lumen metabolites to ensure gut-liver axis functionality. An increase in intestinal barrier permeability induces metabolic endotoxemia that leads to steatohepatitis. Moreover, alterations in the absorption of various metabolites can affect liver metabolism and induce liver steatosis and fibrosis. Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) are a class of drugs developed for the treatment of type 2 diabetes mellitus. They are also commonly used to combat obesity and have been proven to be effective in reversing hepatic steatosis. The mechanisms reported to be involved in this effect include an improved regulation of glycemia, reduced lipid synthesis, ß-oxidation of free fatty acids, and induction of autophagy in hepatic cells. Recently, multiple peptide receptor agonists have been introduced and are expected to increase the effectiveness of the treatment. A modulation of gut microbiota has also been observed with the use of these drugs that may contribute to the amelioration of MAFLD. This review presents the current understanding of the role of the gut-liver axis in the development of MAFLD and use of members of the GLP-1 RA family as pleiotropic agents in the treatment of MAFLD.


Subject(s)
Gastrointestinal Microbiome , Glucagon-Like Peptide-1 Receptor , Liver , Humans , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Gastrointestinal Microbiome/drug effects , Liver/metabolism , Liver/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/microbiology , Animals , Metabolic Syndrome/drug therapy , Metabolic Syndrome/metabolism , Metabolic Syndrome/microbiology , Hypoglycemic Agents/therapeutic use , Hypoglycemic Agents/pharmacology , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/microbiology , Incretins/therapeutic use , Incretins/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Glucagon-Like Peptide-1 Receptor Agonists
4.
Int J Mol Sci ; 25(13)2024 Jun 24.
Article in English | MEDLINE | ID: mdl-39000013

ABSTRACT

Obesity is a global health concern implicated in numerous chronic degenerative diseases, including type 2 diabetes, dyslipidemia, and neurodegenerative disorders. It is characterized by chronic low-grade inflammation, gut microbiota dysbiosis, insulin resistance, glucose intolerance, and lipid metabolism disturbances. Here, we investigated the therapeutic potential of environmental enrichment (EE) to prevent the progression of gut dysbiosis in mice with high-fat diet (HFD)-induced metabolic syndrome. C57BL/6 male mice with obesity and metabolic syndrome, continuously fed with an HFD, were exposed to EE. We analyzed the gut microbiota of the mice by sequencing the 16s rRNA gene at different intervals, including on day 0 and 12 and 24 weeks after EE exposure. Fasting glucose levels, glucose tolerance, insulin resistance, food intake, weight gain, lipid profile, hepatic steatosis, and inflammatory mediators were evaluated in serum, adipose tissue, and the colon. We demonstrate that EE intervention prevents the progression of HFD-induced dysbiosis, reducing taxa associated with metabolic syndrome (Tepidimicrobium, Acidaminobacteraceae, and Fusibacter) while promoting those linked to healthy physiology (Syntrophococcus sucrumutans, Dehalobacterium, Prevotella, and Butyricimonas). Furthermore, EE enhances intestinal barrier integrity, increases mucin-producing goblet cell population, and upregulates Muc2 expression in the colon. These alterations correlate with reduced systemic lipopolysaccharide levels and attenuated colon inflammation, resulting in normalized glucose metabolism, diminished adipose tissue inflammation, reduced liver steatosis, improved lipid profiles, and a significant reduction in body weight gain despite mice's continued HFD consumption. Our findings highlight EE as a promising anti-inflammatory strategy for managing obesity-related metabolic dysregulation and suggest its potential in developing probiotics targeting EE-modulated microbial taxa.


Subject(s)
Diet, High-Fat , Dysbiosis , Gastrointestinal Microbiome , Mice, Inbred C57BL , Obesity , Animals , Diet, High-Fat/adverse effects , Dysbiosis/microbiology , Mice , Obesity/metabolism , Obesity/microbiology , Male , Glucose/metabolism , Mice, Obese , Insulin Resistance , Metabolic Syndrome/metabolism , Metabolic Syndrome/etiology , Metabolic Syndrome/microbiology
5.
Physiol Rep ; 12(13): e16126, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39001594

ABSTRACT

Molecular mechanisms associated to improvement of metabolic syndrome (MetS) during exercise are not fully elucidated. MetS was induced in 250 g male Wistar rats by 30% sucrose in drinking water. Control rats receiving tap water were controls, both groups received solid standard diet. After 14 weeks, an endurance exercised group, and a sedentary were formed for 8 weeks. The soleus and extensor digitorum longus (EDL) muscles were dissected to determine contractile performance, expression of myosin heavy chain isoforms, PGC1α, AMPKα2, NFATC1, MEF2a, SIX1, EYA1, FOXO1, key metabolic enzymes activities. Exercise mildly improved MetS features. MetS didn't alter the contractile performance of the muscles. Exercise didn't altered expression of PGC1α, NFATC1, SIX1 and EYA1 on MetS EDL whereas NFATC1 increased in soleus. Only citrate synthase was affected by MetS on the EDL and this was partially reverted by exercise. Soleus α-ketoglutarate dehydrogenase activity was increased by exercise but MetS rendered the muscle resistant to this effect. MetS affects mostly the EDL muscle, and endurance exercise only partially reverts this. Soleus muscle seems more resilient to MetS. We highlight the importance of studying both muscles during MetS, and their metabolic remodeling on the development and treatment of MetS by exercise.


Subject(s)
Energy Metabolism , Metabolic Syndrome , Physical Conditioning, Animal , Rats, Wistar , Animals , Male , Metabolic Syndrome/metabolism , Metabolic Syndrome/physiopathology , Rats , Muscle, Skeletal/metabolism , Sucrose/metabolism , Sucrose/administration & dosage , Muscle Fibers, Skeletal/metabolism , Muscle Contraction , Phenotype
6.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(3): 783-792, 2024 May 20.
Article in Chinese | MEDLINE | ID: mdl-38948289

ABSTRACT

Alpha-ketoglutarate (α-KG), an endogenous intermediate of the tricarboxylic acid cycle, is involved in a variety of cellular metabolic pathways. It serves as an energy donor, a precursor of amino acid biosynthesis, and an epigenetic regulator. α-KG plays physiological functions in immune regulation, oxidative stress, and anti-aging as well. In recent years, it has been reported that the level of α-KG in the body is closely associated with metabolic syndrome, including obesity, hyperglycemia, and other pathological factors. Exogenous supplementation of α-KG improves obesity, blood glucose levels, and cardiovascular disease risks associated with metabolic syndrome. Furthermore, α-KG regulates the common pathological mechanisms of metabolic syndrome, suggesting the potential application prospect of α-KG in metabolic syndrome. In order to provide a theoretical basis for further exploration of the application of α-KG in metabolic syndrome, we focused on α-KG and metabolic syndrome in this article and summarized the latest research progress in the role of α-KG in improving the pathological condition and disease progression of metabolic syndrome. For the next step, researchers may focus on the co-pathogenesis of metabolic syndrome and investigate whether α-KG can be used to achieve the therapeutic goal of "homotherapy for heteropathy" in the treatment of metabolic syndrome.


Subject(s)
Ketoglutaric Acids , Metabolic Syndrome , Metabolic Syndrome/metabolism , Ketoglutaric Acids/metabolism , Humans , Obesity/metabolism , Obesity/complications , Animals , Oxidative Stress
7.
J Clin Invest ; 134(14)2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39007272

ABSTRACT

A growing body of research has identified circadian-rhythm disruption as a risk factor for metabolic health. However, the underlying biological basis remains complex, and complete molecular mechanisms are unknown. There is emerging evidence from animal and human research to suggest that the expression of core circadian genes, such as circadian locomotor output cycles kaput gene (CLOCK), brain and muscle ARNT-Like 1 gene (BMAL1), period (PER), and cyptochrome (CRY), and the consequent expression of hundreds of circadian output genes are integral to the regulation of cellular metabolism. These circadian mechanisms represent potential pathophysiological pathways linking circadian disruption to adverse metabolic health outcomes, including obesity, metabolic syndrome, and type 2 diabetes. Here, we aim to summarize select evidence from in vivo animal models and compare these results with epidemiologic research findings to advance understanding of existing foundational evidence and potential mechanistic links between circadian disruption and altered clock gene expression contributions to metabolic health-related pathologies. Findings have important implications for the treatment, prevention, and control of metabolic pathologies underlying leading causes of death and disability, including diabetes, cardiovascular disease, and cancer.


Subject(s)
CLOCK Proteins , Circadian Rhythm , Diabetes Mellitus, Type 2 , Humans , Animals , Circadian Rhythm/genetics , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Obesity/genetics , Obesity/metabolism , Metabolic Syndrome/genetics , Metabolic Syndrome/metabolism , Circadian Clocks/genetics
8.
Nutrients ; 16(12)2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38931177

ABSTRACT

CONTEXT/OBJECTIVE: In order to better understand which metabolic differences are related to insulin resistance in metabolic syndrome (MetSyn), we used hyperinsulinemic-euglycemic (HE) clamps in individuals with MetSyn and related peripheral insulin resistance to circulating biomarkers. DESIGN/METHODS: In this cross-sectional study, HE-clamps were performed in treatment-naive men (n = 97) with MetSyn. Subjects were defined as insulin-resistant based on the rate of disappearance (Rd). Machine learning models and conventional statistics were used to identify biomarkers of insulin resistance. Findings were replicated in a cohort with n = 282 obese men and women with (n = 156) and without (n = 126) MetSyn. In addition to this, the relation between biomarkers and adipose tissue was assessed by nuclear magnetic resonance imaging. RESULTS: Peripheral insulin resistance is marked by changes in proteins related to inflammatory processes such as IL-1 and TNF-receptor and superfamily members. These proteins can distinguish between insulin-resistant and insulin-sensitive individuals (AUC = 0.72 ± 0.10) with MetSyn. These proteins were also associated with IFG, liver fat (rho 0.36, p = 1.79 × 10-9) and visceral adipose tissue (rho = 0.35, p = 6.80 × 10-9). Interestingly, these proteins had the strongest association in the MetSyn subgroup compared to individuals without MetSyn. CONCLUSIONS: MetSyn associated with insulin resistance is characterized by protein changes related to body fat content, insulin signaling and pro-inflammatory processes. These findings provide novel targets for intervention studies and should be the focus of future in vitro and in vivo studies.


Subject(s)
Biomarkers , Insulin Resistance , Metabolic Syndrome , Proteome , Humans , Metabolic Syndrome/metabolism , Male , Female , Cross-Sectional Studies , Middle Aged , Adult , Biomarkers/blood , Glucose Clamp Technique , Obesity/metabolism , Adipose Tissue/metabolism , Insulin/blood , Insulin/metabolism , Intra-Abdominal Fat/metabolism
10.
Int J Mol Sci ; 25(12)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38928349

ABSTRACT

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.


Subject(s)
Adipose Tissue , Mesenchymal Stem Cells , Metabolic Syndrome , MicroRNAs , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Female , Male , Metabolic Syndrome/metabolism , Metabolic Syndrome/genetics , Mesenchymal Stem Cells/metabolism , Adult , Middle Aged , Adipose Tissue/metabolism , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/genetics , Obesity/metabolism , Obesity/genetics , Interleukin-10/metabolism , Interleukin-10/genetics , Gene Expression Regulation , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics
11.
Stem Cell Res Ther ; 15(1): 160, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38835014

ABSTRACT

BACKGROUND: Metabolic syndrome (MetS) is a significant epidemiological problem worldwide. It is a pre-morbid, chronic and low-grade inflammatory disorder that precedes many chronic diseases. Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs) could be used to treat MetS because they express high regenerative capacity, strong immunomodulatory properties and allogeneic biocompatibility. This study aims to investigate WJ-MSCs as a therapy against MetS in a rat model. METHODS: Twenty-four animals were fed with high-fat high-fructose (HFHF) diet ad libitum. After 16 weeks, the animals were randomised into treatment groups (n = 8/group) and received a single intravenous administration of vehicle, that is, 3 × 106 cells/kg or 10 × 106 cells/kg of WJ-MSCs. A healthy animal group (n = 6) fed with a normal diet received the same vehicle as the control (CTRL). All animals were periodically assessed (every 4 weeks) for physical measurements, serum biochemistry, glucose tolerance test, cardiovascular function test and whole-body composition. Post-euthanasia, organs were weighed and processed for histopathology. Serum was collected for C-reactive protein and inflammatory cytokine assay. RESULTS: The results between HFHF-treated groups and healthy or HFHF-CTRL did not achieve statistical significance (α = 0.05). The effects of WJ-MSCs were masked by the manifestation of different disease subclusters and continuous supplementation of HFHF diet. Based on secondary analysis, WJ-MSCs had major implications in improving cardiopulmonary morbidities. The lungs, liver and heart show significantly better histopathology in the WJ-MSC-treated groups than in the untreated CTRL group. The cells produced a dose-dependent effect (high dose lasted until week 8) in preventing further metabolic decay in MetS animals. CONCLUSIONS: The establishment of safety and therapeutic proof-of-concept encourages further studies by improving the current therapeutic model.


Subject(s)
Disease Models, Animal , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Metabolic Syndrome , Wharton Jelly , Animals , Metabolic Syndrome/therapy , Metabolic Syndrome/pathology , Metabolic Syndrome/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Rats , Wharton Jelly/cytology , Mesenchymal Stem Cell Transplantation/methods , Male , Injections, Intravenous , Humans , Diet, High-Fat/adverse effects
12.
Lipids Health Dis ; 23(1): 186, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38872138

ABSTRACT

BACKGROUND: Evidence on the association between visceral lipid accumulation and infertility remains limited and controversial. Therefore, the current investigation is the first investigation to unveil this correlation by utilizing novel indicators of visceral lipid accumulation. METHODS: The present study utilized the NHANES 2013-2020 dataset. Researchers utilized multiple logistic regression, smoothed curve fitting, and subgroup analysis to investigate the associations of waist circumference (WC), metabolic score for visceral fat (METS-VF), lipid accumulation product (LAP), visceral adiposity index (VAI) with infertility. Additionally, the eXtreme Gradient Boosting (XGBoost) algorithm model was utilized to evaluate the relative importance of the factors. RESULTS: After adjusting for potential factors that could influence the results, researchers discovered that all these four indicators of visceral lipid accumulation exhibited strong positive correlations with the probability of infertility. The subgroup analysis demonstrated that the correlations remained consistent in the majority of subgroups (P for interaction > 0.05). The results of XGBoost algorithm model indicate that METS-VF is the most meaningful factor in infertility. The ROC curve research revealed that while METS-VF had the greatest AUC values, there was no variation in the AUC value of different markers of visceral fat accumulation (P > 0.05). CONCLUSIONS: The present investigation discovered that increased WC, METS-VF, LAP, and VAI were associated with a heightened prevalence of infertility.


Subject(s)
Intra-Abdominal Fat , Waist Circumference , Humans , Female , Intra-Abdominal Fat/metabolism , Adult , Cross-Sectional Studies , United States/epidemiology , Middle Aged , Infertility, Female/metabolism , ROC Curve , Infertility/metabolism , Lipid Metabolism , Metabolic Syndrome/metabolism , Nutrition Surveys , Adiposity
13.
Cell Syst ; 15(6): 497-509.e3, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38866010

ABSTRACT

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.


Subject(s)
Metabolic Syndrome , Quantitative Trait Loci , Animals , Mice , Quantitative Trait Loci/genetics , Metabolic Syndrome/genetics , Metabolic Syndrome/metabolism , Humans , Male , Genetic Predisposition to Disease/genetics , Female , Mice, Inbred C57BL , Genome-Wide Association Study/methods , Systems Biology/methods
14.
Clin Chim Acta ; 561: 119819, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38901629

ABSTRACT

Metabolic syndrome (MetS) represents a significant public health concern due to its association with an increased risk of cardiovascular disease, type 2 diabetes, and other serious health conditions. Despite extensive research, the underlying molecular mechanisms contributing to MetS pathogenesis remain elusive. This review aims to provide a comprehensive overview of the molecular mechanisms linking MetS and cluster of differentiation (CD) markers, which play critical roles in immune regulation and cellular signaling. Through an extensive literature review with a systematic approach, we examine the involvement of various CD markers in MetS development and progression, including their roles in adipose tissue inflammation, insulin resistance, dyslipidemia, and hypertension. Additionally, we discuss potential therapeutic strategies targeting CD markers for the management of MetS. By synthesizing current evidence, this review contributes to a deeper understanding of the complex interplay between immune dysregulation and metabolic dysfunction in MetS, paving the way for the development of novel therapeutic interventions.


Subject(s)
Metabolic Syndrome , Metabolic Syndrome/metabolism , Humans , Biomarkers/metabolism
15.
Adv Gerontol ; 37(1-2): 138-143, 2024.
Article in Russian | MEDLINE | ID: mdl-38944784

ABSTRACT

Metabolic syndrome is a group of disorders that are closely related to both the risk of developing type 2 diabetes mellitus and cardiovascular diseases, and generally leading to the phenomenon of premature aging of the body. Excessive accumulation of adipose tissue contributes to the development of chronic immune inflammation and oxidative stress, which are both precursors to various disorders, such as insulin resistance, arterial hypertension and dyslipidemia, but also trigger inflammatory processes in patients. An increasing number of studies support the importance of chronic immune inflammation in the pathogenesis of metabolic syndrome, as pro-inflammatory markers such as TNF-α, IL-1ß, IL-6, monocyte chemotactic protein-1 and growth of vascular endothelium. Among a wide range of cytokines, monocyte chemotactic protein-1 is considered one of the most important chemokines, which activates monocytes and other immune cells actively involved in inflammation. Another important point of chronic immune inflammation is its impact on the mental health of patients with metabolic syndrome. Increased levels of anxiety and depression are associated with levels of pro-inflammatory cytokines produced by adipose tissue, which ultimately has an adverse effect on the cognitive status of patients.


Subject(s)
Biomarkers , Inflammation , Metabolic Syndrome , Humans , Metabolic Syndrome/metabolism , Metabolic Syndrome/psychology , Metabolic Syndrome/immunology , Metabolic Syndrome/physiopathology , Inflammation/metabolism , Inflammation/immunology , Inflammation/psychology , Biomarkers/metabolism , Biomarkers/blood , Aged , Cytokines/metabolism , Cytokines/blood , Middle Aged , Aging/psychology , Aging/immunology , Oxidative Stress/physiology
16.
Int J Mol Sci ; 25(11)2024 May 21.
Article in English | MEDLINE | ID: mdl-38891759

ABSTRACT

Metabolic dysfunction-associated steatotic liver disease (MASLD), previously known as nonalcoholic fatty liver disease, is a steatotic liver disease associated with metabolic syndrome (MetS), especially obesity, hypertension, diabetes, hyperlipidemia, and hypertriglyceridemia. MASLD in 43-44% of patients can progress to metabolic dysfunction-associated steatohepatitis (MASH), and 7-30% of these cases will progress to liver scarring (cirrhosis). To date, the mechanism of MASLD and its progression is not completely understood and there were no therapeutic strategies specifically tailored for MASLD/MASH until March 2024. The conventional antiobesity and antidiabetic pharmacological approaches used to reduce the progression of MASLD demonstrated favorable peripheral outcomes but insignificant effects on liver histology. Alternatively, phyto-synthesized metal-based nanoparticles (MNPs) are now being explored in the treatment of various liver diseases due to their unique bioactivities and reduced bystander effects. Although phytonanotherapy has not been explored in the clinical treatment of MASLD/MASH, MNPs such as gold NPs (AuNPs) and silver NPs (AgNPs) have been reported to improve metabolic processes by reducing blood glucose levels, body fat, and inflammation. Therefore, these actions suggest that MNPs can potentially be used in the treatment of MASLD/MASH and related metabolic diseases. Further studies are warranted to investigate the feasibility and efficacy of phytonanomedicine before clinical application.


Subject(s)
Non-alcoholic Fatty Liver Disease , Phytotherapy , Humans , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Phytotherapy/methods , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Animals , Metabolic Syndrome/drug therapy , Metabolic Syndrome/metabolism , Metabolic Syndrome/complications , Metabolic Diseases/drug therapy , Metabolic Diseases/etiology , Metabolic Diseases/metabolism
17.
Biomolecules ; 14(6)2024 May 29.
Article in English | MEDLINE | ID: mdl-38927040

ABSTRACT

Metabolic syndrome (MetS) is a cluster of metabolic abnormalities affecting ~25% of adults and is linked to chronic diseases such as cardiovascular disease, cancer, and neurodegenerative diseases. Oxidative stress and inflammation are key drivers of MetS. Hesperidin, a citrus bioflavonoid, has demonstrated antioxidant and anti-inflammatory properties; however, its effects on MetS are not fully established. We aimed to determine the optimal dose of hesperidin required to improve oxidative stress, systemic inflammation, and glycemic control in a novel mouse model of MetS. Male 5-week-old C57BL/6 mice were fed a high-fat, high-salt, high-sugar diet (HFSS; 42% kcal fat content in food and drinking water with 0.9% saline and 10% high fructose corn syrup) for 16 weeks. After 6 weeks of HFSS, mice were randomly allocated to either the placebo group or low- (70 mg/kg/day), mid- (140 mg/kg/day), or high-dose (280 mg/kg/day) hesperidin supplementation for 12 weeks. The HFSS diet induced significant metabolic disturbances. HFSS + placebo mice gained almost twice the weight of control mice (p < 0.0001). Fasting blood glucose (FBG) increased by 40% (p < 0.0001), plasma insulin by 100% (p < 0.05), and HOMA-IR by 150% (p < 0.0004), indicating insulin resistance. Hesperidin supplementation reduced plasma insulin by 40% at 140 mg/kg/day (p < 0.0001) and 50% at 280 mg/kg/day (p < 0.005). HOMA-IR decreased by 45% at both doses (p < 0.0001). Plasma hesperidin levels significantly increased in all hesperidin groups (p < 0.0001). Oxidative stress, measured by 8-OHdG, was increased by 40% in HFSS diet mice (p < 0.001) and reduced by 20% with all hesperidin doses (p < 0.005). In conclusion, hesperidin supplementation reduced insulin resistance and oxidative stress in HFSS-fed mice, demonstrating its dose-dependent therapeutic potential in MetS.


Subject(s)
Citrus , Dietary Supplements , Disease Models, Animal , Hesperidin , Insulin Resistance , Metabolic Syndrome , Mice, Inbred C57BL , Oxidative Stress , Animals , Hesperidin/pharmacology , Oxidative Stress/drug effects , Metabolic Syndrome/drug therapy , Metabolic Syndrome/metabolism , Male , Mice , Citrus/chemistry , Dose-Response Relationship, Drug , Blood Glucose/metabolism , Blood Glucose/drug effects , Diet, High-Fat/adverse effects , Antioxidants/pharmacology
18.
Front Endocrinol (Lausanne) ; 15: 1382844, 2024.
Article in English | MEDLINE | ID: mdl-38689728

ABSTRACT

Equine metabolic syndrome (EMS) is a critical endocrine condition in horses, characterized by hyperinsulinemia, hyperlipidemia, and insulin resistance, posing a significant threat to their health. This study investigates the efficacy of supplementing EMS-affected horses with Arthrospira platensis enriched with Cr(III), Mg(II), and Mn(II) ions using biosorption process in improving insulin sensitivity and glucose tolerance, reducing inflammation, and mitigating obesity-related fat accumulation. Our results demonstrate that Arthrospira supplementation reduces baseline insulin and glucose levels, contributing to decreased adipose tissue inflammation. Furthermore, Arthrospira supplementation results in a decrease in body weight and improvements in overall body condition scores and cresty neck scores. Additionally, administration of Arthrospira leads to reduced levels of triglycerides and aspartate aminotransferase, indicating a decrease in hepatic adiposity and inflammation. These findings suggest that Arthrospira, enriched with essential micro- and macroelements, can be an advanced feed additive to enhance insulin sensitivity, promote weight reduction, and alleviate inflammatory processes, thereby improving the overall condition of horses affected by EMS. The use of Arthrospira as a feed additive has the potential to complement conventional management strategies for EMS.


Subject(s)
Animal Feed , Chromium , Dietary Supplements , Horse Diseases , Inflammation , Insulin Resistance , Magnesium , Manganese , Metabolic Syndrome , Spirulina , Animals , Horses , Inflammation/metabolism , Metabolic Syndrome/veterinary , Metabolic Syndrome/metabolism , Horse Diseases/metabolism , Horse Diseases/prevention & control , Animal Feed/analysis , Magnesium/metabolism , Male , Female
19.
Front Endocrinol (Lausanne) ; 15: 1369600, 2024.
Article in English | MEDLINE | ID: mdl-38711979

ABSTRACT

Background: The Metabolic Score for Insulin Resistance (METS-IR) offers a promising and reliable non-insulin-based approach to assess insulin resistance and evaluate cardiometabolic risk. However, evidence for the association between METS-IR and hypertension was still limited. Methods: Participants from the National Health and Nutrition Examination Survey (NHANES) database from 2007-2016 were selected for weighted multivariable regression analyses, subgroup analyses and restricted cubic spline (RCS) modeling to assess the association between the METS-IR and hypertension, as well as systolic blood pressure (SBP) and diastolic blood pressure (DBP). Results: This study enrolled 7,721 adults aged ≥20 years, 2,926 (34.03%) of whom was diagnosed as hypertension. After adjusting for all potential covariates, an increased METS-IR (log2 conversion, denoted as log2METS-IR) was independently associated with a higher prevalence of hypertension (odd ratio [OR] 3.99, 95% confidence interval [CI] 3.19~5.01). The OR for hypertension in subjects with the highest quartile of METS-IR was 3.89-fold (OR 3.89, 95% CI 3.06~4.94) higher than that in those with the lowest quartile of METS-IR. This positive correlation became more significant as METS-IR increased (p for trend < 0.001). Log2METS-IR was significantly correlated with increase in SBP (ß 6.75, 95% CI 5.65~7.85) and DBP (ß 5.59, 95% CI 4.75~6.43) in a fully adjusted model. Consistent results were obtained in subgroup analyses. Hypertension, SBP and DBP all exhibited a non-linear increase with the rise in METS-IR. The minimal threshold for the beneficial association of METS-IR with hypertension, SBP and DBP were all identified to be 46.88. Conclusion: The findings of this study revealed a significant positive association between METS-IR and hypertension among US adults, suggesting METS-IR as a potential tool for assessing hypertension risk.


Subject(s)
Hypertension , Insulin Resistance , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult , Blood Pressure/physiology , Cross-Sectional Studies , Metabolic Syndrome/metabolism , Nutrition Surveys , Prevalence , Risk Factors , United States
20.
Sci Rep ; 14(1): 11313, 2024 05 17.
Article in English | MEDLINE | ID: mdl-38760452

ABSTRACT

Physical activity promotes various metabolic benefits by balancing pro and anti-inflammatory adipokines. Recent studies suggest that asprosin might be involved in progression of metabolic syndrome (MetS), however, the underlying mechanisms have not been understood yet. This study aimed to evaluate the effects of high-intensity interval training (HIIT), moderate-intensity continuous training (MICT), and further detraining on MetS indices, insulin resistance, serum and the liver levels of asprosin, and AMP-activated protein kinase (AMPK) pathway in menopause-induced MetS model of rats. A total of 64 Wistar rats were used in this study and divided into eight groups: Sham1, OVX1 (ovariectomized), Sham2, OVX2, OVX + HIIT, OVX + MICT, OVX + HIIT + Det (detraining), and OVX + MICT + Det. Animals performed the protocols, and then serum concentrations of asprosin, TNF-α, insulin, fasting blood glucose, and lipid profiles (TC, LDL, TG, and HDL) were assessed. Additionally, the liver expression of asprosin, AMPK, and P-AMPK was measured by western blotting. Both HIIT and MICT caused a significant decrease in weight, waist circumference, BMI (P = 0.001), and serum levels of glucose, insulin, asprosin (P = 0.001), triglyceride, total cholesterol, low-density lipoprotein (LDL), and TNF-α (P = 0.001), but an increase in the liver AMPK, P-AMPK, and P-AMPK/AMPK (P = 0.001), compared with OVX2 noexercised group. MICT was superior to HIIT in reducing serum asprosin, TNF-a, TG, LDL (P = 0.001), insulin, fasting blood glucose, HOMA-IR, and QUEKI index (P = 0.001), but an increase in the liver AMPK, and p-AMPK (P = 0.001). Although after two months of de-training almost all indices returned to the pre exercise values (P < 0.05). The findings suggest that MICT effectively alleviates MetS induced by menopause, at least partly through the activation of liver signaling of P-AMPK and the reduction of asprosin and TNF-α. These results have practical implications for the development of exercise interventions targeting MetS in menopausal individuals, emphasizing the potential benefits of MICT in mitigating MetS-related complications.


Subject(s)
AMP-Activated Protein Kinases , Disease Models, Animal , Fibrillin-1 , Metabolic Syndrome , Physical Conditioning, Animal , Rats, Wistar , Signal Transduction , Animals , Fibrillin-1/metabolism , Metabolic Syndrome/metabolism , Metabolic Syndrome/therapy , Rats , Female , AMP-Activated Protein Kinases/metabolism , High-Intensity Interval Training/methods , Liver/metabolism , Insulin Resistance , Blood Glucose/metabolism , Insulin/blood , Insulin/metabolism , Peptide Fragments/blood , Peptide Fragments/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...