Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 17.700
Filtrar
1.
Chem Pharm Bull (Tokyo) ; 72(4): 365-373, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38569867

RESUMO

Obesity is known to be associated with increased inflammation and dysregulated autophagy, both of which contribute to insulin resistance. Saikosaponin-A (SSA) has been reported to exhibit anti-inflammatory and lipid-lowering properties. In this research, we employed a combination of computational modeling and animal experiments to explore the effects of SSA. Male C57BL/6 mice were categorized into four groups: normal diet, high-fat diet (HFD), HFD + atorvastatin 10 mg/kg, and HFD + SSA 10 mg/kg. We conducted oral glucose and fat tolerance tests to assess metabolic parameters and histological changes. Furthermore, we evaluated the population of Kupffer cells (KCs) and examined gene expressions related to inflammation and autophagy. Computational analysis revealed that SSA displayed high binding affinity to tumor necrosis factor (TNF)-α, nuclear factor (NF)-κB, fibroblast growth factor 21 (FGF21), and autophagy-related 7 (ATG7). Animal study demonstrated that SSA administration improved fasting and postprandial glucose levels, homeostatic model assessment of insulin resistance (HOMA-IR) index, as well as triglyceride, free fatty acid, total cholesterol, low-density lipoprotein cholesterol (LDL-C)-cholesterol, and high-density lipoprotein cholesterol (HDL-C)-cholesterol levels in HFD-fed mice. Moreover, SSA significantly reduced liver weight and fat accumulation, while inhibiting the infiltration and M1 activation of KCs. At the mRNA level, SSA downregulated TNF-α and NF-κB expression, while upregulating FGF21 and ATG7 expression. In conclusion, our study suggests that SSA may serve as a therapeutic agent for addressing the metabolic complications associated with obesity. This potential therapeutic effect is attributed to the suppression of inflammatory cytokines and the upregulation of FGF21 and ATG7.


Assuntos
Experimentação Animal , Resistência à Insulina , Ácido Oleanólico/análogos & derivados , Saponinas , Camundongos , Masculino , Animais , Resistência à Insulina/fisiologia , Camundongos Endogâmicos C57BL , Obesidade/tratamento farmacológico , Fígado , Inflamação/metabolismo , Glucose/metabolismo , Colesterol , Dieta Hiperlipídica/efeitos adversos , Fator de Necrose Tumoral alfa/metabolismo , Insulina/metabolismo
2.
Physiol Rep ; 12(7): e15995, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38561245

RESUMO

Exercise has different effects on different tissues in the body, the sum of which may determine the response to exercise and the health benefits. In the present study, we aimed to investigate whether physical training regulates transcriptional network communites common to both skeletal muscle (SM) and subcutaneous adipose tissue (SAT). Eight such shared transcriptional communities were found in both tissues. Eighteen young overweight adults voluntarily participated in 7 weeks of combined strength and endurance training (five training sessions per week). Biopsies were taken from SM and SAT before and after training. Five of the network communities were regulated by training in SM but showed no change in SAT. One community involved in insulin- AMPK signaling and glucose utilization was upregulated in SM but downregulated in SAT. This diverging exercise regulation was confirmed in two independent studies and was also associated with BMI and diabetes in an independent cohort. Thus, the current finding is consistent with the differential responses of different tissues and suggests that body composition may influence the observed individual whole-body metabolic response to exercise training and help explain the observed attenuated whole-body insulin sensitivity after exercise training, even if it has significant effects on the exercising muscle.


Assuntos
Resistência à Insulina , Obesidade , Adulto , Humanos , Obesidade/metabolismo , Músculo Esquelético/metabolismo , Exercício Físico/fisiologia , Gordura Subcutânea/metabolismo , Insulina/metabolismo , Resistência à Insulina/fisiologia , Expressão Gênica , Tecido Adiposo/metabolismo
3.
Sci Rep ; 14(1): 7670, 2024 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-38561446

RESUMO

Dietary factors such as food texture affect feeding behavior and energy metabolism, potentially causing obesity and type 2 diabetes. We previously found that rats fed soft pellets (SPs) were neither hyperphagic nor overweight but demonstrated glucose intolerance, insulin resistance, and hyperplasia of pancreatic ß-cells. In the present study, we investigated the mechanism of muscle atrophy in rats that had been fed SPs on a 3-h time-restricted feeding schedule for 24 weeks. As expected, the SP rats were normal weight; however, they developed insulin resistance, glucose intolerance, and fat accumulation. In addition, skeletal muscles of SP rats were histologically atrophic and demonstrated disrupted insulin signaling. Furthermore, we learned that the muscle atrophy of the SP rats developed via the IL-6-STAT3-SOCS3 and ubiquitin-proteasome pathways. Our data show that the dietary habit of consuming soft foods can lead to not only glucose intolerance or insulin resistance but also muscle atrophy.


Assuntos
Diabetes Mellitus Tipo 2 , Intolerância à Glucose , Resistência à Insulina , Ratos , Animais , Resistência à Insulina/fisiologia , Intolerância à Glucose/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Insulina/metabolismo , Atrofia Muscular/etiologia , Atrofia Muscular/metabolismo , Músculo Esquelético/metabolismo , Dieta , Dieta Hiperlipídica
4.
J Diabetes ; 16(4): e13540, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38599845

RESUMO

OBJECTIVE: Mitochondria-associated membranes (MAMs) serve pivotal functions in hepatic insulin resistance (IR). Our aim was to explore the potential role of MAMs in mitigating hepatic IR through exercise and to compare the effects of different intensities of exercise on hepatic MAMs formation in high-fat diet (HFD) mice. METHODS: Male C57BL/6J mice were fed an HFD and randomly assigned to undergo supervised high-intensity interval training (HIIT) or moderate-intensity continuous training (MICT). IR was evaluated using the serum triglyceride/high-density lipoprotein cholesterol ratio (TG/HDL-C), glucose tolerance test (GTT), and insulin tolerance test (ITT). Hepatic steatosis was observed using hematoxylin-eosin (H&E) and oil red O staining. The phosphatidylinositol 3-kinase/protein kinase B/glycogen synthase kinase 3 beta (PI3K-AKT-GSK3ß) signaling pathway was assessed to determine hepatic IR. MAMs were evaluated through immunofluorescence (colocalization of voltage-dependent anion-selective channel 1 [VDAC1] and inositol 1,4,5-triphosphate receptor [IP3R]). RESULTS: After 8 weeks on an HFD, there was notable inhibition of the hepatic PI3K/Akt/GSK3ß signaling pathway, accompanied by a marked reduction in hepatic IP3R-VDAC1 colocalization levels. Both 8-week HIIT and MICT significantly enhanced the hepatic PI3K/Akt/GSK3ß signaling and colocalization levels of IP3R-VDAC1 in HFD mice, with MICT exhibiting a stronger effect on hepatic MAMs formation. Furthermore, the colocalization of hepatic IP3R-VDAC1 positively correlated with the expression levels of phosphorylation of protein kinase B (p-AKT) and phosphorylation of glycogen synthase kinase 3 beta (p-GSK3ß), while displaying a negative correlation with serum triglyceride/high-density lipoprotein cholesterol levels. CONCLUSION: The reduction in hepatic MAMs formation induced by HFD correlates with the development of hepatic IR. Both HIIT and MICT effectively bolster hepatic MAMs formation in HFD mice, with MICT demonstrating superior efficacy. Thus, MAMs might wield a pivotal role in exercise-induced alleviation of hepatic IR.


Assuntos
Treinamento Intervalado de Alta Intensidade , Resistência à Insulina , Masculino , Camundongos , Animais , Resistência à Insulina/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Glicogênio Sintase Quinase 3 beta , Fosfatidilinositol 3-Quinases , Dieta Hiperlipídica/efeitos adversos , 60482 , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Triglicerídeos , Lipoproteínas HDL , Colesterol
5.
J Alzheimers Dis ; 98(4): 1169-1179, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38607755

RESUMO

Alzheimer's disease (AD) is a complex neurodegenerative disorder characterized by the accumulation of neurofibrillary tangles and amyloid-ß plaques. Recent research has unveiled the pivotal role of insulin signaling dysfunction in the pathogenesis of AD. Insulin, once thought to be unrelated to brain function, has emerged as a crucial factor in neuronal survival, synaptic plasticity, and cognitive processes. Insulin and the downstream insulin signaling molecules are found mainly in the hippocampus and cortex. Some molecules responsible for dysfunction in insulin signaling are GSK-3ß, Akt, PI3K, and IRS. Irregularities in insulin signaling or insulin resistance may arise from changes in the phosphorylation levels of key molecules, which can be influenced by both stimulation and inactivity. This, in turn, is believed to be a crucial factor contributing to the development of AD, which is characterized by oxidative stress, neuroinflammation, and other pathological hallmarks. Furthermore, this route is known to be indirectly influenced by Nrf2, NF-κB, and the caspases. This mini-review delves into the intricate relationship between insulin signaling and AD, exploring how disruptions in this pathway contribute to disease progression. Moreover, we examine recent advances in drug delivery systems designed to target insulin signaling for AD treatment. From oral insulin delivery to innovative nanoparticle approaches and intranasal administration, these strategies hold promise in mitigating the impact of insulin resistance on AD. This review consolidates current knowledge to shed light on the potential of these interventions as targeted therapeutic options for AD.


Assuntos
Doença de Alzheimer , Resistência à Insulina , Humanos , Doença de Alzheimer/patologia , Insulina/metabolismo , Resistência à Insulina/fisiologia , Glicogênio Sintase Quinase 3 beta , Peptídeos beta-Amiloides/metabolismo , Sistemas de Liberação de Medicamentos
6.
Mol Biol Rep ; 51(1): 516, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38622329

RESUMO

BACKGROUND: Resveratrol has received much attention due to its beneficial effects including antioxidant activity. The purpose of this study was to investigate the therapeutic effects of resveratrol treatment on oxidative stress and insulin resistance in the skeletal muscle of high-fat diet (HFD)-fed animals. METHODS AND RESULTS: A total of 30 six-week-old C57BL/6J mice were randomly allocated to three groups (10 animals in each group): The control group in which mice were fed a normal chow diet (NCD); the HFD group in which mice were fed an HFD for 26 weeks; and the HFD-resveratrol group in which HFD was replaced by a resveratrol supplemented-HFD (400 mg/kg diet) after 10 weeks of HFD feeding. At the end of this period, gastrocnemius muscle samples were examined to determine insulin resistance and the oxidative status in the presence of HFD and resveratrol. Resveratrol supplementation in HFD-fed mice reduced body and adipose tissue weight, improved insulin sensitivity, and decreased oxidative stress as indicated by lower malonaldehyde (MDA) levels and higher total antioxidant capacity. The supplement also increased the expression and activity of antioxidative enzymes in gastrocnemius muscle and modulated Nrf2 and Keap1 expression levels. CONCLUSIONS: These results suggest that resveratrol is effective in improving the antioxidant defense system of the skeletal muscle in HFD-fed mice, indicating its therapeutic potential to combat diseases associated with insulin resistance and oxidative stress.


Assuntos
Antioxidantes , Resistência à Insulina , Camundongos , Animais , Antioxidantes/metabolismo , Resveratrol/farmacologia , Resveratrol/metabolismo , Resistência à Insulina/fisiologia , Dieta Hiperlipídica/efeitos adversos , Fator 2 Relacionado a NF-E2/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Camundongos Endogâmicos C57BL , Músculo Esquelético/metabolismo , Transdução de Sinais , Insulina/metabolismo
7.
Gynecol Endocrinol ; 40(1): 2341701, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38622970

RESUMO

OBJECTIVE: To evaluate the effects of alpha lipoic acid (ALA) on hormonal and metabolic parameters in a group of overweight/obese Polycystic Ovary Syndrome (PCOS) patients. METHODS: This was a retrospective study in which thirty-two overweight/obese patients with PCOS (n = 32) not requiring hormonal treatment were selected from the database of the ambulatory clinic of the Gynecological Endocrinology Center at the University of Modena and Reggio Emilia, Italy. The hormonal profile, routine exams and insulin and C-peptide response to oral glucose tolerance test (OGTT) were evaluated before and after 12 weeks of complementary treatment with ALA (400 mg/day). Hepatic Insulin Extraction (HIE) index was also calculated. RESULTS: ALA administration significantly improved insulin sensitivity and decreased ALT and AST plasma levels in all subjects, though no changes were observed on reproductive hormones. When PCOS patients were subdivided according to the presence or absence of familial diabetes background, the higher effects of ALA were observed in the former group that showed AST and ALT reduction and greater HIE index decrease. CONCLUSION: ALA administration improved insulin sensitivity in overweight/obese PCOS patients, especially in those with familial predisposition to diabetes. ALA administration improved both peripheral sensitivity to insulin and liver clearance of insulin. Such effects potentially decrease the risk of nonalcoholic fat liver disease and diabetes in PCOS patients.


Assuntos
Diabetes Mellitus , Resistência à Insulina , Hepatopatia Gordurosa não Alcoólica , Síndrome do Ovário Policístico , Ácido Tióctico , Feminino , Humanos , Insulina , Sobrepeso/complicações , Sobrepeso/tratamento farmacológico , Síndrome do Ovário Policístico/complicações , Síndrome do Ovário Policístico/tratamento farmacológico , Ácido Tióctico/uso terapêutico , Resistência à Insulina/fisiologia , Hepatopatia Gordurosa não Alcoólica/complicações , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Hepatopatia Gordurosa não Alcoólica/metabolismo , Estudos Retrospectivos , Obesidade/complicações , Obesidade/tratamento farmacológico
8.
Nutrients ; 16(6)2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38542793

RESUMO

Protein intake reportedly increases the risk of diabetes; however, the results have been inconsistent. Diabetes in adulthood may be attributed to early life dietary amino acid composition. This study aimed to investigate the association between amino acid composition and glycemic biomarkers in adolescents. Dietary intake was assessed using a food frequency questionnaire, and fasting glucose and insulin levels were measured in 1238 eighth graders. The homeostatic model assessment (HOMA) indices (insulin resistance and ß-cell function) were calculated. Anthropometrics were measured and other covariates were obtained from a questionnaire. Amino acid composition was isometric log transformed according to the compositional data analysis, which was used as explanatory variables in multivariate linear regression models for glucose, insulin, and HOMA indices. Only the association between glucose and leucine was significant. In replacement of other amino acids with leucine, an increase of 0.1% of total amino acids correlated with a lower glucose level (-1.02 mg/dL). One-to-one substitution of leucine for isoleucine or methionine decreased glucose (-2.98 and -2.28 mg/dL, respectively). Associations with other biomarkers were not observed. In conclusion, compositional data analysis of amino acids revealed an association only with glucose in adolescents; however, the results of this study should be verified in other populations.


Assuntos
Diabetes Mellitus , Resistência à Insulina , Humanos , Adolescente , Leucina , Japão , Glicemia/metabolismo , Insulina , Resistência à Insulina/fisiologia , Aminoácidos , Glucose , Biomarcadores
9.
Toxicol In Vitro ; 97: 105802, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38431059

RESUMO

BACKGROUND: An etiology of palmitic acid (PA) induced insulin resistance (IR) is complex for which two mechanisms are proposed namely ROS induced JNK activation and lipid induced protein kinase-C (PKCε) activation. However, whether these mechanisms act alone or in consortium is not clear. METHODS AND RESULTS: In this study, we have characterized PA induced IR in liver cells. These cells were treated with different concentrations of PA for either 8 or 16 h. Insulin responsiveness of cells treated with PA for 8 h was found to be same as that of control. However, cells treated with PA for 16 h, showed increased glucose output both in the presence and in absence of insulin only at higher concentrations, indicating development of IR. In these, both JNK and PKCε were activated in response to increased ROS and lipid accumulation, respectively. Activated JNK and PKCε phosphorylated IRS1 at Ser-307 resulting in inhibition of AKT which in turn inactivated GSK3ß, leading to reduced glycogen synthase activity. Inhibition of AKT also reduced insulin suppression of hepatic gluconeogenesis by activating Forkhead box protein O1 (FOXO1) and increased expression of the gluconeogenic enzymes and their transcription factors. CONCLUSION: Thus, our data clearly demonstrate that both these mechanisms work simultaneously and more importantly, identified a threshold of HepG2 cells, which when crossed led to the pathological state of IR in response to PA.


Assuntos
Resistência à Insulina , Humanos , Resistência à Insulina/fisiologia , Ácido Palmítico/toxicidade , Células Hep G2 , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Glucose/metabolismo , Insulina/metabolismo , Fígado/metabolismo
10.
J Ovarian Res ; 17(1): 69, 2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38539230

RESUMO

Polycystic ovary syndrome (PCOS) is a common reproductive and metabolic condition in women of childbearing age and a major cause of anovulatory infertility. The pathophysiology of PCOS is complex. Recent studies have reported that apart from hyperandrogenism, insulin resistance, systemic chronic inflammation, and ovarian dysfunction, gut microbiota dysbiosis is also involved in PCOS development and may aggravate inflammation and metabolic dysfunction, forming a vicious cycle. As naturally occurring plant secondary metabolites, polyphenols have been demonstrated to have anticancer, antibacterial, vasodilator, and analgesic properties, mechanistically creating putative bioactive, low-molecular-weight metabolites in the human gut. Here, we summarize the role of gut microbiota dysbiosis in the development of PCOS and demonstrate the ability of different polyphenols - including anthocyanin, catechins, and resveratrol - to regulate gut microbes and alleviate chronic inflammation, thus providing new insights that may assist in the development of novel therapeutic strategies to treat women with PCOS.


Assuntos
Microbioma Gastrointestinal , Hiperandrogenismo , Resistência à Insulina , Síndrome do Ovário Policístico , Feminino , Humanos , Síndrome do Ovário Policístico/complicações , Microbioma Gastrointestinal/fisiologia , Polifenóis/farmacologia , Polifenóis/uso terapêutico , Disbiose/complicações , Resistência à Insulina/fisiologia , Inflamação/tratamento farmacológico , Inflamação/metabolismo
11.
Int J Mol Sci ; 25(6)2024 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-38542170

RESUMO

The communication between mitochondria and the endoplasmic reticulum (ER) is facilitated by a dynamic membrane structure formed by protein complexes known as mitochondria-associated membranes (MAMs). The structural and functional integrity of MAMs is crucial for insulin signal transduction, relying heavily on their regulation of intracellular calcium homeostasis, lipid homeostasis, mitochondrial quality control, and endoplasmic reticulum stress (ERS). This article reviews recent research findings, suggesting that exercise may promote the remodeling of MAMs structure and function by modulating the expression of molecules associated with their structure and function. This, in turn, restores cellular homeostasis and ultimately contributes to the amelioration of insulin resistance (IR). These insights provide additional possibilities for the study and treatment of insulin resistance-related metabolic disorders such as obesity, diabetes, fatty liver, and atherosclerosis.


Assuntos
Resistência à Insulina , Humanos , Resistência à Insulina/fisiologia , 60482 , Mitocôndrias/metabolismo , Retículo Endoplasmático/metabolismo , Homeostase , Estresse do Retículo Endoplasmático/fisiologia
12.
Mol Metab ; 82: 101905, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38431218

RESUMO

OBJECTIVE: Kallistatin (KST), also known as SERPIN A4, is a circulating, broadly acting human plasma protein with pleiotropic properties. Clinical studies in humans revealed reduced KST levels in obesity. The exact role of KST in glucose and energy homeostasis in the setting of insulin resistance and type 2 diabetes is currently unknown. METHODS: Kallistatin mRNA expression in human subcutaneous white adipose tissue (sWAT) of 47 people with overweight to obesity of the clinical trial "Comparison of Low Fat and Low Carbohydrate Diets With Respect to Weight Loss and Metabolic Effects (B-SMART)" was measured. Moreover, we studied transgenic mice systemically overexpressing human KST (hKST-TG) and wild type littermate control mice (WT) under normal chow (NCD) and high-fat diet (HFD) conditions. RESULTS: In sWAT of people with overweight to obesity, KST mRNA increased after diet-induced weight loss. On NCD, we did not observe differences between hKST-TG and WT mice. Under HFD conditions, body weight, body fat and liver fat content did not differ between genotypes. Yet, during intraperitoneal glucose tolerance tests (ipGTT) insulin excursions and HOMA-IR were lower in hKST-TG (4.42 ± 0.87 AU, WT vs. 2.20 ± 0.27 AU, hKST-TG, p < 0.05). Hyperinsulinemic euglycemic clamp studies with tracer-labeled glucose infusion confirmed improved insulin sensitivity by higher glucose infusion rates in hKST-TG mice (31.5 ± 1.78 mg/kg/min, hKST-TG vs. 18.1 ± 1.67 mg/kg/min, WT, p < 0.05). Improved insulin sensitivity was driven by reduced hepatic insulin resistance (clamp hepatic glucose output: 7.7 ± 1.9 mg/kg/min, hKST-TG vs 12.2 ± 0.8 mg/kg/min, WT, p < 0.05), providing evidence for direct insulin sensitizing effects of KST for the first time. Insulin sensitivity was differentially affected in skeletal muscle and adipose tissue. Mechanistically, we observed reduced Wnt signaling in the liver but not in skeletal muscle, which may explain the effect. CONCLUSIONS: KST expression increases after weight loss in sWAT from people with obesity. Furthermore, human KST ameliorates diet-induced hepatic insulin resistance in mice, while differentially affecting skeletal muscle and adipose tissue insulin sensitivity. Thus, KST may be an interesting, yet challenging, therapeutic target for patients with obesity and insulin resistance.


Assuntos
Diabetes Mellitus Tipo 2 , Resistência à Insulina , Doenças não Transmissíveis , Serpinas , Humanos , Camundongos , Animais , Glucose/metabolismo , Resistência à Insulina/fisiologia , Serpinas/genética , Sobrepeso , Insulina/metabolismo , Obesidade/metabolismo , Camundongos Transgênicos , Dieta Hiperlipídica/efeitos adversos , Homeostase , Redução de Peso , RNA Mensageiro/metabolismo
13.
Front Endocrinol (Lausanne) ; 15: 1297841, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38481448

RESUMO

Objective: To evaluate the effects of behavioral intervention for polycystic ovary syndrome (PCOS). Methods: Electronic databases were searched, including Pubmed, Medline, EMBASE, and the Cochrane Central Register of Controlled Trials from inception to 1 April 2023. Inclusion criteria for this study required a diagnosis of PCOS. Interventions of interest included behavioral intervention and routine treatment compared with routine treatment. The studies included in the analysis were designed as randomized controlled trials (RCTs). We conducted meta-analyses following the recommended guidelines. The data was analyzed using either the random effects model or fixed effects model. The results of the studies were expressed as either mean differences (MD) or standardized mean differences (SMD) along with their corresponding 95% confidence intervals (CIs). Results: Eight RCTs were identified, including data from 744 patients (415 in the intervention group and 329 in the control group). The results indicate an improvement in the effectiveness of behavioral interventions for weight loss (MD: -1.07; 95% CI: -2.1 to 0.03; I2 = 0%; P=0.04), body mass index (BMI) (MD: -1.12; 95% CI: -1.92 to -0.33; I2 = 73%; P=0.006), waist circumference (MD: -3.97; 95% CI: -5.64 to -2.29; I2 = 0%; P<0.00001), quality of life about weight (MD: 0.58; 95% CI: 0.15 to 1.02; I2 = 0%; P=0.008), depression (SMD: -1.12; 95% CI: -2.35 to -0.07; I2 = 92%; P=0.04), and triglycerides (MD: -0.16; 95% CI: -0.27 to -0.05; I2 = 27%; P=0.004). However, there were no significant differences in menstrual cycles, hirsutism, emotions, and infertility. The study also found that behavioral interventions had no significant effect on systolic and diastolic blood pressure, high-density lipoprotein, low-density lipoprotein, homeostasis model assessment of insulin resistance, testosterone, total cholesterol, fasting glucose, fasting insulin, hemoglobin A1C, and sex hormone binding globulin. Conclusion: Behavioral intervention supplementation contributes to weight loss, reduction in BMI and waist circumference, and improvement in depression among patients with PCOS. However, no significant improvement was observed in the biochemical index and quality of life. The long-term effects of behavioral intervention for PCOS remain unclear due to limitations in the quality of the studies involved and the short duration of treatment. Systematic Review Registration: https://www.crd.york.ac.uk/PROSPERO, identifier CRD42023442875.


Assuntos
Resistência à Insulina , Síndrome do Ovário Policístico , Feminino , Humanos , Síndrome do Ovário Policístico/metabolismo , Hirsutismo , Resistência à Insulina/fisiologia , Qualidade de Vida , Redução de Peso
14.
Biochem Biophys Res Commun ; 705: 149742, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38460438

RESUMO

l-norleucine, an isomer of leucine, stimulates the anabolic process of insulin. However, it is not known if and how it improves insulin sensitivity and insulin resistance. This experiment describes the generation of an insulin resistance model using high glucose-induced cells and the administration of 1.0 mmol/L l-norleucine for 48 h, to observe the effects on metabolism and gene expression in skeletal muscle cells. The results showed that l-norleucine significantly increased mitochondrial ATP content, decreased the amount of reactive oxygen species (ROS) and promoted the expression of mitochondrial generation-related genes TFAM, AMPK, PGC-1α in cells under high glucose treatment; at the same time, l-norleucine also increased glucose uptake, suggesting that l-norleucine increased insulin sensitivity and improved insulin resistance. This study suggesting that l-norleucine improves insulin resistance by ameliorating oxidative stress damage of mitochondria, improving mitochondrial function, and improving insulin sensitivity in skeletal muscle cell caused by high glucose, rather than by altering mitochondrial efficiency.


Assuntos
Resistência à Insulina , Humanos , Resistência à Insulina/fisiologia , Músculo Esquelético/metabolismo , Mitocôndrias/metabolismo , Insulina/metabolismo , Norleucina/metabolismo , Norleucina/farmacologia , Glucose/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Mitocôndrias Musculares/metabolismo
15.
Cardiovasc Diabetol ; 23(1): 97, 2024 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-38493102

RESUMO

BACKGROUND: Tissue-specific insulin resistance (IR) predominantly in muscle (muscle IR) or liver (liver IR) has previously been linked to distinct fasting metabolite profiles, but postprandial metabolite profiles have not been investigated in tissue-specific IR yet. Given the importance of postprandial metabolic impairments in the pathophysiology of cardiometabolic diseases, we compared postprandial plasma metabolite profiles in response to a high-fat mixed meal between individuals with predominant muscle IR or liver IR. METHODS: This cross-sectional study included data from 214 women and men with BMI 25-40 kg/m2, aged 40-75 years, and with predominant muscle IR or liver IR. Tissue-specific IR was assessed using the muscle insulin sensitivity index (MISI) and hepatic insulin resistance index (HIRI), which were calculated from the glucose and insulin responses during a 7-point oral glucose tolerance test. Plasma samples were collected before (T = 0) and after (T = 30, 60, 120, 240 min) consumption of a high-fat mixed meal and 247 metabolite measures, including lipoproteins, cholesterol, triacylglycerol (TAG), ketone bodies, and amino acids, were quantified using nuclear magnetic resonance spectroscopy. Differences in postprandial plasma metabolite iAUCs between muscle and liver IR were tested using ANCOVA with adjustment for age, sex, center, BMI, and waist-to-hip ratio. P-values were adjusted for a false discovery rate (FDR) of 0.05 using the Benjamini-Hochberg method. RESULTS: Sixty-eight postprandial metabolite iAUCs were significantly different between liver and muscle IR. Liver IR was characterized by greater plasma iAUCs of large VLDL (p = 0.004), very large VLDL (p = 0.002), and medium-sized LDL particles (p = 0.026), and by greater iAUCs of TAG in small VLDL (p = 0.025), large VLDL (p = 0.003), very large VLDL (p = 0.002), all LDL subclasses (all p < 0.05), and small HDL particles (p = 0.011), compared to muscle IR. In liver IR, the postprandial plasma fatty acid (FA) profile consisted of a higher percentage of saturated FA (p = 0.013), and a lower percentage of polyunsaturated FA (p = 0.008), compared to muscle IR. CONCLUSION: People with muscle IR or liver IR have distinct postprandial plasma metabolite profiles, with more unfavorable postprandial metabolite responses in those with liver IR compared to muscle IR.


Assuntos
Resistência à Insulina , Masculino , Humanos , Feminino , Resistência à Insulina/fisiologia , Estudos Transversais , Triglicerídeos , Ácidos Graxos/metabolismo , Fígado/metabolismo , Músculos/metabolismo , Período Pós-Prandial/fisiologia
16.
Afr J Reprod Health ; 28(2): 43-54, 2024 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-38425153

RESUMO

Polycystic ovary syndrome (PCOS), as an endocrine and metabolic disorder, affects approximately 6% -20% of women of childbearing age. This study aims to assess the therapeutic effects of Metformin combined with vitamin D in PCOS patients. Eight databases were searched to obtain randomized controlled trials, both domestically and internationally, on the effects of Metformin combined with vitamin D in patients with PCOS. Data analysis was performed using RevMan 5.3 software. Nine studies were ultimately included in this meta-analysis. Six studies reported the homeostatic model assessment for insulin resistance of the test group and the control group, which was significantly lower (SMD: -0.23; 95% Cl: -0.42,-0.04; P<0.05) than the control group, body mass index (BMI) (SMD: -1.86; 95% Cl: -2.77,-0.96; P<0.01), Serum 25 (OH) D (SMD: 14.28; 95% Cl: 12.26,16.29; P<0.01), testosterone (SMD: -0.11; 95% Cl: -0.15,-0.07; P<0.01) and regulated menstrual cycles (OR: 1.27; 95% Cl: 0.99,1.63; P=0.063). Our meta-analysis of nine trials demonstrates significant reductions in insulin resistance, BMI, and testosterone levels, along with increased serum vitamin D levels and improved menstrual cycle regulation after Metformin and vitamin D treatment. These findings suggest the potential of this combined therapy in managing the multifaceted aspects of PCOS.


Le syndrome des ovaires polykystiques (SOPK), en tant que trouble endocrinien et métabolique, touche environ 6 à 20 % des femmes en âge de procréer. Cette étude vise à évaluer les effets thérapeutiques de la metformine associée à la vitamine D chez les patients atteints du SOPK. Huit bases de données ont été consultées pour obtenir des essais contrôlés randomisés, tant au niveau national qu'international, sur les effets de la metformine associée à la vitamine D chez les patients atteints du SOPK. L'analyse des données a été réalisée à l'aide du logiciel RevMan 5.3. Neuf études ont finalement été incluses dans cette méta-analyse. Six études ont rapporté l'évaluation du modèle homéostatique pour la résistance à l'insuline du groupe test et du groupe témoin, qui était significativement inférieure (DMS : -0,23 ; IC à 95 % : -0,42, -0,04 ; P <0,05) par rapport au groupe témoin, la masse corporelle indice (IMC) (DMS : -1,86 ; Cl 95 % : -2,77, -0,96 ; P<0,01), Sérum 25 (OH) D (SMD : 14,28 ; Cl 95 % : 12,26,16,29 ; P<0,01), testostérone (DMS : -0,11 ; Cl à 95 % : -0,15, -0,07 ; P<0,01) et cycles menstruels régulés (OR : 1,27 ; Cl à 95 % : 0,99, 1,63 ; P=0,063). Notre méta-analyse de neuf essais démontre des réductions significatives de la résistance à l'insuline, de l'IMC et des taux de testostérone, ainsi qu'une augmentation des taux sériques de vitamine D et une amélioration de la régulation du cycle menstruel après un traitement à la metformine et à la vitamine D. Ces résultats suggèrent le potentiel de cette thérapie combinée dans la gestion des aspects multiformes du SOPK.


Assuntos
Metformina , Síndrome do Ovário Policístico , Vitamina D , Feminino , Humanos , Hipoglicemiantes/uso terapêutico , Resistência à Insulina/fisiologia , Metformina/uso terapêutico , Síndrome do Ovário Policístico/tratamento farmacológico , Testosterona/metabolismo , Vitamina D/uso terapêutico
17.
Nutrients ; 16(5)2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38474874

RESUMO

AIM: Meat is commonly consumed in India; however, in comparison to Western settings, it is eaten in relatively lower quantities and with minimal processing. The association between meat intake and cardio-metabolic diseases (CMDs) and their risk factors in India is currently uncertain. We examined whether meat intake is associated with risk factors for CMDs and the measures of subclinical atherosclerosis in urbanising villages in southern India. METHODS: We conducted a cross-sectional analysis of 6012 adults (52.3% male) participating in the Andhra Pradesh Children and Parents' Study (APCAPS), which is a large prospective, intergenerational cohort study in Southern India that began with the long-term follow-up of the Hyderabad Nutrition Trial (1987-1990). We used cross-sectional data from the third wave of data collection conducted in 2010-2012, where total meat intake was assessed using 100-item, semi-quantitative validated food frequency questionnaires (FFQ). The FFQs were validated using multiple weighed 24 h dietary recalls. The main predictor, 'total meat intake', was calculated as the sum of chicken, red meat, and fish consumption. The risk factors for CMDs [systolic blood pressure (SBP), diastolic blood pressure (DBP), body mass index (BMI), waist circumference (WC), fasting glucose, total cholesterol, homeostasis model assessment insulin resistance (HOMA-IR), total cholesterol, low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol, triglycerides, and C-reactive protein] and measures of subclinical atherosclerosis [Carotid Intima-Media Thickness, Pulse Wave Velocity, and Augmentation Index] were assessed using standardised clinical procedures. Stratified by gender, the association of meat intake with the risk factors of CMDs and measures of subclinical atherosclerosis was examined using linear multilevel models with random intercept at the household level. RESULTS: The mean (SD) age of the male (n = 3128) and female participants (n = 2828) was 34.09 years (15.55) and 34.27 years (12.73), respectively. The median (IQR) intake of meat was 17.79 g/day (8.90, 30.26) in males and 8.90 g/day (4.15, 18.82) in females. In males, a 10 g increase in total meat intake/1000 Kcal/day was positively associated with DBP, BMI, WC, total cholesterol, LDL-C, and triglycerides, whereas in females, a 10 g increase in total meat intake/1000 Kcal/day was positively associated with SBP, DBP, fasting glucose, HOMA-IR, total cholesterol, LDL-C, and triglycerides. There was no relationship between meat consumption and measures of subclinical atherosclerosis. CONCLUSIONS: Meat intake had a linear positive association with CMD risk factors among the relatively younger Indian population who were consuming meat at lower levels compared to their European counterparts.


Assuntos
Aterosclerose , Doenças Cardiovasculares , Resistência à Insulina , Adulto , Criança , Humanos , Masculino , Feminino , Estudos Transversais , LDL-Colesterol , Espessura Intima-Media Carotídea , Estudos de Coortes , Estudos Prospectivos , Análise de Onda de Pulso , Doenças Cardiovasculares/etiologia , Fatores de Risco , Triglicerídeos , Resistência à Insulina/fisiologia , Glucose , Carne , Aterosclerose/complicações , Índice de Massa Corporal
18.
Exp Clin Endocrinol Diabetes ; 132(3): 152-161, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38513652

RESUMO

BACKGROUND: Type 2 diabetes mellitus (T2DM) is a commonly observed complication associated with obesity. The effect of fibroblast growth factor 19 (FGF19), a promising therapeutic agent for metabolic disorders, on pancreatic ß cells in obesity-associated T2DM remains poorly understood. METHODS: Human pancreatic ß cells were cultured with high glucose (HG) and palmitic acid (PA), followed by treatment with FGF19. The cell proliferation, apoptosis, and insulin secretion were evaluated by CCK-8, qRT-PCR, ELISA, flow cytometry, and western blotting. The expression of the insulin receptor substrate (IRS)/glucose transporter (GLUT) pathway was evaluated. The interaction between FGF19 and IRS1 was predicted using the STRING database and verified by co-immunoprecipitation and immunofluorescence. The regulatory effects of the IRS1/GLUT4 pathway on human pancreatic ß cells were assessed by overexpressing IRS1 and silencing IRS1 and GLUT4. RESULTS: HG+PA treatment reduced the human pancreatic ß cell proliferation and insulin secretion and promoted cell apoptosis. However, FGF19 treatment restored these alterations and significantly increased the expressions of IRS1, GLUT1, and GLUT4 in the IRS/GLUT pathway. Furthermore, FGF19 and IRS1 were found to interact. IRS1 overexpression partially promoted the proliferation of pancreatic ß cells and insulin secretion through GLUT4. Additionally, the silencing of IRS1 or GLUT4 attenuated the therapeutic effects of FGF19. CONCLUSION: In conclusion, FGF19 partly promoted the proliferation and insulin secretion of human pancreatic ß cells and inhibited apoptosis by upregulating the IRS1/GLUT4 pathway. These findings establish a theoretical framework for the clinical utilization of FGF19 in the treatment of obesity-associated T2DM.


Assuntos
Diabetes Mellitus Tipo 2 , Fatores de Crescimento de Fibroblastos , Transportador de Glucose Tipo 1 , Proteínas Substratos do Receptor de Insulina , Secreção de Insulina , Células Secretoras de Insulina , Obesidade , Humanos , Proliferação de Células/efeitos dos fármacos , Diabetes Mellitus Tipo 2/complicações , Fatores de Crescimento de Fibroblastos/farmacologia , Proteínas Substratos do Receptor de Insulina/metabolismo , Resistência à Insulina/fisiologia , Secreção de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Obesidade/etiologia , Obesidade/terapia , Ácido Palmítico/metabolismo , Ácido Palmítico/farmacologia , Transportador de Glucose Tipo 1/metabolismo , Linhagem Celular Tumoral , Glucose/metabolismo , Glucose/farmacologia
19.
Sci Rep ; 14(1): 7113, 2024 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-38532054

RESUMO

Type 2 diabetes (T2D) can cause severe cardiac complications at functional, histologic and molecular levels. These pathological complications could be mediated by ATP-releasing channels such as Panx1 and ATP receptors, in particular P2X7. The aim of our study was to investigate the effect of high-intensity interval training (HIIT) on T2D-induced cardiac complications at the functional, histopathological and molecular levels, with a particular focus on ATP-releasing channels. 48 male Wistar rats at the age of 8 weeks were randomly allocated into four groups: control (Con), Diabetes (T2D), Training (TR), and Diabetes + Training (T2D + TR). T2D was induced by a high-fat diet plus a low dose (35 mg/kg) of STZ administration. Rats in the TR and T2D + TR groups underwent an 8-weeks training program involving intervals ranging from 80 to 100% of their maximum running speed (Vmax), with 4-10 intervals per session. Protein expression of Interleukin 1ß (IL1ß), Interleukin 10 (IL-10), Pannexin 1 (Panx1), P2X7R (purinergic P2X receptor 7), NLRP1 (NLR Family Pyrin Domain Containing 1), BAX, and Bcl2 were measured in the heart tissue. Additionally, we assessed heart function, histopathological changes, as well as insulin resistance using the homeostasis model assessment of insulin resistance (HOMA-IR). In contrast to the T2D group, HIIT led to increased protein expression of Bcl2 and IL-10 in the heart. It also resulted in improvements in systolic and diastolic blood pressures, heart rate, ± dp/dt (maximum and minimum changes in left ventricular pressure), while reducing protein expression of IL-1ß, Panx1, P2X7R, NLRP1, and BAX levels in the heart. Furthermore, left ventricular diastolic pressure (LVDP) was reduced (P ≤ 0.05). Moreover, heart lesion scores increased with T2D but decreased with HIIT, along with a reduction in fibrosis percentage (P ≤ 0.05). The results of this study suggest that the cardioprotective effects of HIIT on the diabetic heart may be mediated by the modulation of ATP-releasing channels. This modulation may lead to a reduction in inflammation and apoptosis, improve cardiac function, and attenuate cardiac injury and fibrosis.


Assuntos
Diabetes Mellitus Tipo 2 , Treinamento Intervalado de Alta Intensidade , Resistência à Insulina , Masculino , Ratos , Animais , Resistência à Insulina/fisiologia , Interleucina-10 , Proteína X Associada a bcl-2 , Ratos Wistar , Fibrose , Trifosfato de Adenosina
20.
J Transl Med ; 22(1): 309, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38532480

RESUMO

BACKGROUND: Dihydromyricetin (DHM), a flavonoid compound of natural origin, has been identified in high concentrations in ampelopsis grossedentata and has a broad spectrum of biological and pharmacological functions, particularly in regulating glucose and lipid metabolism. The objective of this research was to examine how DHM affected nonalcoholic fatty liver disease (NAFLD) and its underlying mechanisms involved in the progression of NAFLD in a rat model subjected to a high-fat diet (HFD). Additionally, the study examines the underlying mechanisms in a cellular model of steatohepatitis using palmitic acid (PA)-treated HepG2 cells, with a focus on the potential correlation between autophagy and hepatic insulin resistance (IR) in the progress of NAFLD. METHODS: SD rats were exposed to a HFD for a period of eight weeks, followed by a treatment with DHM (at doses of 50, 100, and 200 mg·kg-1·d-1) for additional six weeks. The HepG2 cells received a 0.5 mM PA treatment for 24 h, either alone or in conjunction with DHM (10 µM). The histopathological alterations were assessed by the use of Hematoxylin-eosin (H&E) staining. The quantification of glycogen content and lipid buildup in the liver was conducted by the use of PAS and Oil Red O staining techniques. Serum lipid and liver enzyme levels were also measured. Autophagic vesicle and autolysosome morphology was studied using electron microscopy. RT-qPCR and/or western blotting techniques were used to measure IR- and autophagy-related factors levels. RESULTS: The administration of DHM demonstrated efficacy in ameliorating hepatic steatosis, as seen in both in vivo and in vitro experimental models. Moreover, DHM administration significantly increased GLUT2 expression, decreased G6Pase and PEPCK expression, and improved IR in the hepatic tissue of rats fed a HFD and in cells exhibiting steatosis. DHM treatment elevated Beclin 1, ATG 5, and LC3-II levels in hepatic steatosis models, correlating with autolysosome formation. The expression of AMPK levels and its downstream target PGC-1α, and PPARα were decreased in HFD-fed rats and PA-treated hepatocytes, which were reversed through DHM treatment. AMPK/ PGC-1α and PPARα knockdown reduced the impact of DHM on hepatic autophagy, IR and accumulation of hepatic lipid. CONCLUSIONS: Our findings revealed that AMPK/ PGC-1α, PPARα-dependent autophagy pathways in the pathophysiology of IR and hepatic steatosis has been shown, suggesting that DHM might potentially serve as a promising treatment option for addressing this disease.


Assuntos
Flavonóis , Resistência à Insulina , Hepatopatia Gordurosa não Alcoólica , Ratos , Animais , Camundongos , Hepatopatia Gordurosa não Alcoólica/patologia , PPAR alfa/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Resistência à Insulina/fisiologia , Ratos Sprague-Dawley , Fígado/patologia , Metabolismo dos Lipídeos , Ácido Palmítico/metabolismo , Ácido Palmítico/farmacologia , Ácido Palmítico/uso terapêutico , Autofagia , Dieta Hiperlipídica , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...