Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
1.
SciELO Preprints; ago. 2024.
Preprint en Inglés | SciELO Preprints | ID: pps-9605

RESUMEN

Introduction: The liver plays a critical role in glucose and lipid homeostasis. Insulin resistance (IR) has been increasingly recognized as a primary etiological factor in metabolic disorders. Hepatic insulin resistance (HIR) is a specific manifestation of IR characterized by the liver's reduced responsiveness to insulin despite elevated circulating insulin levels. Objective: This review aims to elucidate the role of HIR in the pathogenesis of metabolic disorders, focusing on its relationship with metabolic dysfunction-associated fatty liver disease (MAFLD) and type 2 diabetes mellitus (T2DM). Methods: A comprehensive literature search was conducted to explore the underlying mechanisms of HIR, its clinical implications, and its association with MAFLD and T2DM. Results: HIR is characterized by impaired insulin-mediated glucose uptake and increased hepatic glucose output. This metabolic dysfunction contributes to the development of hepatic steatosis, dyslipidemia, and insulin resistance in peripheral tissues. The interplay between HIR and lipogenesis is important in the progression of MAFLD and its association with T2DM, and could be described as a hepatic equivalent of T2DM. Conclusion: The understanding of a T2DM-like condition in the liver is decisive for developing more targeted and effective treatments.


Introdução: O fígado desempenha um papel crítico na homeostase da glicose e lipídios. A resistência à insulina (RI) tem sido cada vez mais reconhecida como um fator etiológico primário em distúrbios metabólicos. A resistência à insulina hepática (RIH) é uma manifestação específica da RI caracterizada pela redução da resposta hepática à insulina, apesar dos níveis elevados de insulina circulante. Objetivo: Este manuscrito visa avaliar o papel da RIH na patogênese dos distúrbios metabólicos, com foco em sua relação com a doença hepática gordurosa associada à disfunção metabólica (DHGDM) e o diabetes mellitus tipo 2 (DMT2). Métodos: Foi realizada uma busca abrangente na literatura para explorar os mecanismos subjacentes da RIH, suas implicações clínicas e sua associação com a DHGDM e o DMT2. Resultados: A RIH é caracterizada pela redução da captação de glicose mediada pela insulina e pelo aumento da produção hepática de glicose. Essa disfunção metabólica contribui para o desenvolvimento de esteatose hepática, dislipidemia e RI em tecidos periféricos. A interação entre RIH e lipogênese é importante na progressão da DHGDM e sua associação com o DMT2, podendo ser descrita como um equivalente hepático do DMT2. Conclusão: A compreensão de uma condição semelhante ao DMT2 no fígado é decisiva para o desenvolvimento de tratamentos mais direcionados e eficazes.

2.
Adv Sci (Weinh) ; : e2403451, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38970167

RESUMEN

Statins, the first-line medication for dyslipidemia, are linked to an increased risk of type 2 diabetes. But exactly how statins cause diabetes is yet unknown. In this study, a developed short-term statin therapy on hyperlipidemia mice show that hepatic insulin resistance is a cause of statin-induced diabetes. Statin medication raises the expression of progesterone and adiponectin receptor 9 (PAQR9) in liver, which inhibits insulin signaling through degradation of protein phosphatase, Mg2+/Mn2+ dependent 1 (PPM1α) to activate ERK pathway. STIP1 homology and U-box containing protein 1 (STUB1) is found to mediate ubiquitination of PPM1α promoted by PAQR9. On the other hand, decreased activity of hepatocyte nuclear factor 4 alpha (HNF4α) seems to be the cause of PAQR9 expression under statin therapy. The interventions on PAQR9, including deletion of PAQR9, caloric restriction and HNF4α activation, are all effective treatments for statin-induced diabetes, while liver specific over-expression of PPM1α is another possible tactic. The results reveal the importance of HNF4α-PAQR9-STUB1-PPM1α axis in controlling the statin-induced hepatic insulin resistance, offering a fresh insight into the molecular mechanisms underlying statin therapy.

3.
Diabetes Res Clin Pract ; 214: 111788, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39032659

RESUMEN

AIM: Our study aimed to analyze how hepatic insulin resistance (IR) influences the efficacy of 48 weeks of metformin treatment in newly diagnosed type 2 diabetes patients. METHODS: We chose 291 participants who were allocated to a 48-week metformin treatment in the "Metformin and Acarbose in Chinese as initial Hypoglycemic treatment" (MARCH) trial and calculated their hepatic insulin resistance indexes (HIRI). We equally grouped the subjects into tertiles: low, medium, and high HIRI groups based on baseline HIRI; Low, medium, and high ΔHIRI groups based on the decreasing extent of HIRI after a 48-week metformin treatment. RESULTS: Multiple linear regression showed that baseline HIRI was positively associated with the rising degree of Matsuda index and the falling range of fasting blood glucose, fasting insulin, homeostasis model assessment of insulin resistance (HOMA-IR), and HIRI. Furthermore, baseline fasting insulin, homeostatic model assessment of ß cell function (HOMA-ß), HOMA-IR, and HIRI were positively associated with the decreasing extent of HIRI, while baseline Matsuda index had a negative association with the falling extent of HIRI. CONCLUSIONS: Patients with higher levels of hepatic IR obtained better curative effects from metformin in terms of glycemic control, insulin saving, insulin sensitivity enhancement, and IR improvement. Higher fasting blood glucose, fasting insulin, HOMA-ß, IR, and lower Matsuda index were indicators of better hepatic IR improvement.


Asunto(s)
Glucemia , Diabetes Mellitus Tipo 2 , Hipoglucemiantes , Resistencia a la Insulina , Hígado , Metformina , Humanos , Metformina/uso terapéutico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/sangre , Resistencia a la Insulina/fisiología , Masculino , Femenino , Persona de Mediana Edad , Hipoglucemiantes/uso terapéutico , Hígado/metabolismo , Hígado/efectos de los fármacos , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Glucemia/análisis , Adulto , Anciano , Resultado del Tratamiento , Insulina
4.
Cardiovasc Toxicol ; 24(8): 747-756, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38851663

RESUMEN

Metabolic dysfunction associated-steatotic liver disease (MASLD)/metabolic dysfunction-associated steatohepatitis (MASH) is the liver manifestation of metabolic syndrome, which is characterized by insulin resistance, hyperglycemia, hypertension, dyslipidemia, and/or obesity. Environmental pollutant exposure has been recently identified as a risk factor for developing MASH. Heterocyclic amines (HCAs) are mutagens generated when cooking meat at high temperatures or until well-done. Recent epidemiological studies reported that dietary HCA exposure may be linked to insulin resistance and type II diabetes, and we recently reported that HCAs induce insulin resistance and glucose production in human hepatocytes. However, no previous studies have examined the effects of HCAs on hepatic lipid homeostasis. In the present study, we assessed the effects of two common HCAs, MeIQx (2-amino-3, 8-dimethylimidazo [4, 5-f] quinoxaline) and PhIP (2-amino-1-methyl-6-phenylimidazo[4, 5-b] pyridine), on lipid homeostasis in cryopreserved human hepatocytes. Exposure to a single concentration of 25 µM MeIQx or PhIP in human hepatocytes led to dysregulation of lipid homeostasis, typified by significant increases in lipid droplets and triglycerides. PhIP significantly increased expression of lipid droplet-associated genes, PNPLA3 and HSD17B13, and both HCAs significantly increased PLIN2. Exposure to MeIQx or PhIP also significantly increased expression of several key genes involved in lipid synthesis, transport and metabolism, including FASN, DGAT2, CPT1A, SCD, and CD36. Furthermore, both MeIQx and PhIP significantly increased intracellular cholesterol and decreased expression of PON1 which is involved in cholesterol efflux. Taken together, these results suggest that HCAs dysregulate lipid production, metabolism, and storage. The current study demonstrates, for the first time, that HCA exposure may lead to fat accumulation in hepatocytes, which may contribute to hepatic insulin resistance and MASH.


Asunto(s)
Criopreservación , Hepatocitos , Homeostasis , Metabolismo de los Lípidos , Humanos , Hepatocitos/metabolismo , Hepatocitos/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Homeostasis/efectos de los fármacos , Células Cultivadas , Mutágenos/toxicidad , Triglicéridos/metabolismo
5.
J Diabetes ; 16(4): e13540, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38599845

RESUMEN

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.


Asunto(s)
Entrenamiento de Intervalos de Alta Intensidad , Resistencia a la Insulina , Masculino , Ratones , Animales , Resistencia a la Insulina/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Fosfatidilinositol 3-Quinasas , Dieta Alta en Grasa/efectos adversos , Membranas Asociadas a Mitocondrias , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Triglicéridos , Lipoproteínas HDL , Colesterol
6.
Br J Pharmacol ; 181(7): 967-986, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37940413

RESUMEN

BACKGROUND AND PURPOSE: Defective insulin signalling and dysfunction of the endoplasmic reticulum (ER), driven by excessive lipid accumulation in the liver, is a characteristic feature in the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Thromboxane A2 (TXA2 ), an arachidonic acid metabolite, is significantly elevated in obesity and plays a crucial role in hepatic gluconeogenesis and adipose tissue macrophage polarization. However, the role of liver TXA2 /TP receptors in insulin resistance and lipid metabolism is largely unknown. EXPERIMENTAL APPROACH: TP receptor knockout (TP-/- ) mice were generated and fed a high-fat diet for 16 weeks. Insulin sensitivity, ER stress responses and hepatic lipid accumulation were assessed. Furthermore, we used primary hepatocytes to dissect the mechanisms by which the TXA2 /TP receptor axis regulates insulin signalling and hepatocyte lipogenesis. KEY RESULTS: TXA2 was increased in diet-induced obese mice, and depletion of TP receptors in adult mice improved systemic insulin resistance and hepatic steatosis. Mechanistically, we found that the TXA2 /TP receptor axis disrupts insulin signalling by activating the Ca2+ /calcium calmodulin-dependent kinase II γ (CaMKIIγ)-protein kinase RNA-like endoplasmic reticulum kinase (PERK)-C/EBP homologous protein (Chop)-tribbles-like protein 3 (TRB3) axis in hepatocytes. In addition, our results revealed that the TXA2 /TP receptor axis directly promoted lipogenesis in primary hepatocytes and contributed to Kupffer cell inflammation. CONCLUSIONS AND IMPLICATIONS: The TXA2 /TP receptor axis facilitates insulin resistance through Ca2+ /CaMKIIγ to activate PERK-Chop-TRB3 signalling. Inhibition of hepatocyte TP receptors improved hepatic steatosis and inflammation. The TP receptor is a new therapeutic target for NAFLD and metabolic syndrome.


Asunto(s)
Resistencia a la Insulina , Insulinas , Enfermedad del Hígado Graso no Alcohólico , Animales , Ratones , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Receptores de Tromboxano A2 y Prostaglandina H2/metabolismo , Receptores de Tromboxano A2 y Prostaglandina H2/uso terapéutico , Tromboxano A2/metabolismo , Tromboxano A2/uso terapéutico , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Hígado/metabolismo , Hepatocitos/metabolismo , Inflamación/metabolismo , Estrés del Retículo Endoplásmico , Insulinas/metabolismo , Dieta Alta en Grasa , Ratones Endogámicos C57BL
7.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-1039897

RESUMEN

Background Arsenic exposure is a common and important environmental and occupational hazardous factor in China, and arsenic-induced insulin resistance (IR) has attracted widespread attention as a negative health outcome to the population. Objective To explore part of the mechanism of hepatic IR induced by arsenic exposure based on the peroxisome proliferators-activated receptors γ (PPARγ)/ glucose transporter 4 (GLUT4) pathway, and to investigate potential effects of Ginkgo biloba extract (GBE) on hepatic IR induced by arsenic exposure and associated mechanism of action. Methods The target of drug action was predicted by network pharmacology and verified by in vivo and in vitro experiments. In vivo experiments: 48 SPF C57BL/6J male mice were divided into 4 groups, including control group, 50 mg·L−1 NaAsO2 model group (NaAsO2), 50 mg·L−1 NaAsO2+10 mg·kg−1 GBE intervene group (NaAsO2+GBE), and 10 mg·kg−1 GBE group (GBE), 12 mice in each group. The animals were given free access to purified water containing 50 mg·L−1 NaAsO2, or given intraperitoneal injection of normal saline containing 10 mg·kg−1 GBE once per week. After 6 months of exposure, blood glucose detection, intraperitoneal glucose tolerance test (IPGTT), and insulin tolerance test (ITT) were performed. Serum and liver tissues were collected after the mice were neutralized, liver histopathological sections were obtained, serum insulin levels, liver tissue glycogen content, glucose content were detected by enzyme linked immunosorbent assay (ELISA), and the expression of PPARγ and GLUT4 proteins was detected by Western blot (WB). In vitro experiments: HepG2 cells were divided into 4 groups, including control group, 8 μmol·L−1 NaAsO2 group (NaAsO2), 8 μmol·L−1 NaAsO2 + 200 mg·L−1 GBE intervene group (NaAsO2+GBE), and 200 mg·L−1 GBE group (GBE). The levels of glycogen and glucose were detected by ELISA, and the expression of PPARγ and GLUT4 proteins was detected by WB. Results A strong binding effect between GBE and PPARγ was revealed by network pharmacology. In in vivo experiments, the NaAsO2 group exhibited an elevated blood glucose compared to the control group, and the NaAsO2+GBE group showed a decreased blood glucose compared to the NaAsO2 group (P<0.01). The histopathological sections indicated severe liver structural damage in the arsenic exposure groups (NaAsO2 group and NaAsO2+GBE group), with varying staining intensity, partial liver cell necrosis, and diffuse red blood cell appearance. Both results of in vitro and in vivo experiments showed a decrease in glycogen synthesis and glucose uptake in the NaAsO2 groups compared to the control groups, which was alleviated in the NaAsO2+GBE group (P<0.01). The results of WB revealed inhibited PPARγ expression and reduced GLUT4 levels on the cell membrane, and all these changes were alleviated in the NaAsO2+GBE group (P<0.01). Conclusion This study findings suggest that GBE antagonizes arsenic exposure-induced hepatic IR by regulating the PPARγ/GLUT4 pathway, indicating that GBE has a protective effect on arsenic exposure-induced hepatic IR, and PPARγ may be a potential therapeutic target for arsenic exposure-induced hepatic IR.

8.
Eur J Appl Physiol ; 124(5): 1425-1437, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38100040

RESUMEN

PURPOSE: Impaired insulin sensitivity is central in the etiology of type 2 diabetes in people with obesity. The effectiveness of resistance training (RE) alone in improving insulin sensitivity in people with obesity is undetermined. This study aimed to determine the influence of obesity on insulin sensitivity responses to RE. METHODS: Nineteen sedentary men were allocated to Lean (BMI 22.7 ± 2.5 kg m-2; n = 10) or Obese group (BMI 33.2 ± 3.2 kg m-2; n = 9). Participants were evaluated before and after a 10-week supervised progressive RE (3 sets of 10 repetition maximum (RM), 3 d/wk) for insulin sensitivity indexes using an oral glucose tolerance test, body composition using anthropometrics, and strength using 1RM. RESULTS: Groups were matched at baseline for all variables except for body composition and absolute strength. Body fat was not changed in both groups. Matsuda insulin sensitivity index, hepatic insulin resistance, and insulin area under the curve improved by 64.3 ± 61.9 unit, - 58.2 ± 102.9 unit, 2.3 ± 4.1 unit, and - 721.6 ± 858.2 µU/ml, respectively, only in the Lean group. The increased 1RM% for leg press was greater in the Lean (49.5 ± 18.7%) than in the Obese (31.5 ± 13.9), but not different for bench press (18.0 ± 9.1% vs. 16.4 ± 6.0%, respectively). CONCLUSION: Sustained obesity precludes insulin sensitivity improvements and attenuates strength gains in response to progressive RE. Additional strategies such as caloric restriction might be necessary for RE to improve insulin sensitivity, particularly at high levels of obesity.


Asunto(s)
Resistencia a la Insulina , Obesidad , Entrenamiento de Fuerza , Humanos , Masculino , Resistencia a la Insulina/fisiología , Entrenamiento de Fuerza/métodos , Obesidad/fisiopatología , Obesidad/terapia , Adulto , Fuerza Muscular/fisiología , Composición Corporal/fisiología
9.
Biomed Environ Sci ; 36(11): 1045-1058, 2023 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-38098324

RESUMEN

Objective: In this study, the combined effect of two stressors, namely, electromagnetic fields (EMFs) from mobile phones and fructose consumption, on hypothalamic and hepatic master metabolic regulators of the AMPK/SIRT1-UCP2/FOXO1 pathway were elucidated to delineate the underlying molecular mechanisms of insulin resistance. Methods: Weaned Wistar rats (28 days old) were divided into 4 groups: Normal, Exposure Only (ExpO), Fructose Only (FruO), and Exposure and Fructose (EF). Each group was provided standard laboratory chow ad libitum for 8 weeks . Additionally, the control groups, namely, the Normal and FruO groups, had unrestricted access to drinking water and fructose solution (15%), respectively. Furthermore, the respective treatment groups, namely, the ExpO and EF groups, received EMF exposure (1,760 MHz, 2 h/day x 8 weeks). In early adulthood, mitochondrial function, insulin receptor signaling, and oxidative stress signals in hypothalamic and hepatic tissues were assessed using western blotting and biochemical analysis. Result: In the hypothalamic tissue of EF, SIRT1, FOXO 1, p-PI3K, p-AKT, Complex III, UCP2, MnSOD, and catalase expressions and OXPHOS and GSH activities were significantly decreased ( P < 0.05) compared to the Normal, ExpO, and FruO groups. In hepatic tissue of EF, the p-AMPKα, SIRT1, FOXO1, IRS1, p-PI3K, Complex I, II, III, IV, V, UCP2, and MnSOD expressions and the activity of OXPHOS, SOD, catalase, and GSH were significantly reduced compared to the Normal group ( P < 0.05). Conclusion: The findings suggest that the combination of EMF exposure and fructose consumption during childhood and adolescence in Wistar rats disrupts the closely interlinked and multi-regulated crosstalk of insulin receptor signals, mitochondrial OXPHOS, and the antioxidant defense system in the hypothalamus and liver.


Asunto(s)
Teléfono Celular , Fructosa , Humanos , Ratas , Animales , Adulto , Ratas Wistar , Fructosa/metabolismo , Catalasa , Receptor de Insulina/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Campos Electromagnéticos/efectos adversos , Sirtuina 1/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína Forkhead Box O1/metabolismo , Proteína Desacopladora 2
10.
Nutrients ; 15(21)2023 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-37960269

RESUMEN

The etiology of numerous metabolic disorders is characterized by hepatic insulin resistance (IR). Uncertainty surrounds miR-34a's contribution to high-fat-induced hepatic IR and its probable mechanism. The role and mechanism of miR-34a and its target gene ENO3 in high-fat-induced hepatic IR were explored by overexpressing/suppressing miR-34a and ENO3 levels in in vivo and in vitro experiments. Moreover, as a human hepatic IR model, the miR-34a/ENO3 pathway was validated in patients with non-alcoholic fatty liver disease (NAFLD). The overexpression of hepatic miR-34a lowered insulin signaling and altered glucose metabolism in hepatocytes. In contrast, reducing miR-34a expression significantly reversed hepatic IR indices induced by palmitic acid (PA)/HFD. ENO3 was identified as a direct target gene of miR-34a. Overexpression of ENO3 effectively inhibited high-fat-induced hepatic IR-related indices both in vitro and in vivo. Moreover, the expression patterns of members of the miR-34a/ENO3 pathway in the liver tissues of NAFLD patients was in line with the findings of both cellular and animal studies. A high-fat-induced increase in hepatic miR-34a levels attenuates insulin signaling and impairs glucose metabolism by suppressing the expression of its target gene ENO3, ultimately leading to hepatic IR. The miR-34a/ENO3 pathway may be a potential therapeutic target for hepatic IR and related metabolic diseases.


Asunto(s)
Resistencia a la Insulina , MicroARNs , Enfermedad del Hígado Graso no Alcohólico , Animales , Humanos , Ratones , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , MicroARNs/genética , MicroARNs/metabolismo , Hígado/metabolismo , Insulina/metabolismo , Glucosa/metabolismo , Ratones Endogámicos C57BL
11.
Nutr Metab (Lond) ; 20(1): 51, 2023 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-37996895

RESUMEN

OBJECTIVE: To characterize potential mechanisms of fisetin on hepatic insulin resistance (IR) using network pharmacology and in vitro validation. METHODS: Putative targets of fisetin were retrieved from the Traditional Chinese Medicine Systems Pharmacology database, whereas the potential genes of hepatic IR were obtained from GeneCards database. A protein-protein interaction (PPI) network was constructed according to the intersection targets of fisetin and hepatic IR using the Venn diagram. The biological functions and potential pathways related to genes were determined using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. Cell experiments were also conducted to further verify the mechanism of fisetin on hepatic IR. RESULTS: A total of 118 potential targets from fisetin were associated with hepatic IR. The areas of nodes and corresponding degree values of TP53, AKT1, TNF, IL6, CASP3, CTNNB1, JUN, SRC, epidermal growth factor receptor (EGFR), and HSP90AA1 were larger and could be easily found in the PPI network. Furthermore, GO analysis revealed that these key targets were significantly involved in multiple biological processes that participated in oxidative stress and serine/threonine kinase activity. KEGG enrichment analysis showed that the PI3K/AKT signaling pathway was a significant pathway involved in hepatic IR. Our in vitro results demonstrated that fisetin treatment increased the expressions of EGFR and IRS in HepG2 and L02 cells under normal or IR conditions. Western blot results revealed that p-AKT/AKT levels were significantly up-regulated, suggesting that fisetin was involved in the PI3K/AKT signaling pathway to regulate insulin signaling. CONCLUSION: We explored the pharmacological actions and the potential molecular mechanism of fisetin in treating hepatic IR from a holistic perspective. Our study lays a theoretical foundation for the development of fisetin for type 2 diabetes.

12.
J Gastroenterol Hepatol ; 38(10): 1847-1854, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37646384

RESUMEN

BACKGROUND AND AIM: There have been several reports that some probiotics improve non-alcoholic fatty liver disease (NAFLD); however, many studies have involved cocktail therapies. We evaluated whether heat-killed Lactobacillus brevis SBL88 (L. brevis SBL88) monotherapy improves the clinical features of NAFLD. METHODS: The NAFLD model was induced in mice fed a high-fat diet (HFD) (HFD mice) or HFD + 1% heat-killed L. brevis SBL88 (SBL mice) for 16 weeks. Histopathological liver findings were analyzed. To evaluate the gut microbiota, a modified terminal restriction fragment length polymorphism analysis of the feces was performed. RNA sequencing in the liver was performed with Ion Proton™. To investigate the direct effects of heat-killed L. brevis SBL88, an in vitro study was performed. RESULTS: Histopathological findings revealed that fat droplets in the liver were significantly reduced in SBL mice; however, terminal restriction fragment length polymorphism did not show alterations in the gut microbiota between HFD mice and SBL mice. RNA sequencing and pathway analysis revealed that the regulation of lipid and insulin metabolism was affected. The mRNA expression of insulin receptor substrate 2 (IRS-2) was significantly higher in SBL mice, whereas the expression of IRS-1 was not significantly different. Phospho-IRS-2 expression was also significantly increased in SBL mice. In addition, an in vitro study revealed significant alterations in IRS-2 and forkhead box protein O1 expression levels. CONCLUSION: SBL mice exhibited partially improved selective hepatic insulin resistance. Our data suggest that heat-killed L. brevis SBL88 could attenuate the clinical features of NAFLD that are not mediated by alterations in the gut microbiota.


Asunto(s)
Microbioma Gastrointestinal , Resistencia a la Insulina , Levilactobacillus brevis , Enfermedad del Hígado Graso no Alcohólico , Animales , Ratones , Enfermedad del Hígado Graso no Alcohólico/terapia , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Resistencia a la Insulina/genética , Microbioma Gastrointestinal/genética , Calor , Hígado/patología , Dieta Alta en Grasa/efectos adversos , Ratones Endogámicos C57BL
14.
Front Endocrinol (Lausanne) ; 14: 1193373, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37396181

RESUMEN

Diabetes mellitus is a metabolic disease clinically-characterized as acute and chronic hyperglycemia. It is emerging as one of the common conditions associated with incident liver disease in the US. The mechanism by which diabetes drives liver disease has become an intense topic of discussion and a highly sought-after therapeutic target. Insulin resistance (IR) appears early in the progression of type 2 diabetes (T2D), particularly in obese individuals. One of the co-morbid conditions of obesity-associated diabetes that is on the rise globally is referred to as non-alcoholic fatty liver disease (NAFLD). IR is one of a number of known and suspected mechanism that underlie the progression of NAFLD which concurrently exhibits hepatic inflammation, particularly enriched in cells of the innate arm of the immune system. In this review we focus on the known mechanisms that are suspected to play a role in the cause-effect relationship between hepatic IR and hepatic inflammation and its role in the progression of T2D-associated NAFLD. Uncoupling hepatic IR/hepatic inflammation may break an intra-hepatic vicious cycle, facilitating the attenuation or prevention of NAFLD with a concurrent restoration of physiologic glycemic control. As part of this review, we therefore also assess the potential of a number of existing and emerging therapeutic interventions that can target both conditions simultaneously as treatment options to break this cycle.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Enfermedad del Hígado Graso no Alcohólico , Humanos , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Inflamación/complicaciones
15.
J Biol Chem ; 299(5): 104678, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37028764

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is one of the most common liver diseases worldwide. Although the involvement of chronic overnutrition, systemic inflammation, and insulin resistance in the development of NAFLD is well-established, however, the associations among these remain to be elucidated. Several studies have reported that chronic overnutrition, such as excessive consumption of fats (high-fat diet, HFD), can cause insulin resistance and inflammation. However, the mechanisms by which HFD exerts inflammation and thereby promotes insulin resistance and intrahepatic fat accumulation remain poorly understood. Here, we show that HFD induces the expression of hepatic serine/threonine kinase 38 (STK38), which further induces systemic inflammation leading to insulin resistance. Notably, ectopic expression of STK38 in mouse liver leads to lean NAFLD phenotype with hepatic inflammation, insulin resistance, intrahepatic lipid accumulation, and hypertriglyceridemia in mice fed on a regular chow diet. Further, depletion of hepatic STK38 in HFD-fed mice remarkably reduces proinflammation, improves hepatic insulin sensitivity, and decreases hepatic fat accumulation. Mechanistically, two critical stimuli are elicited by STK38 action. For one stimulus, STK38 binds to Tank-Binding protein Kinase 1 and induces Tank-Binding protein Kinase 1 phosphorylation to promote NF-κß nuclear translocation that mobilizes the release of proinflammatory cytokines and eventually leads to insulin resistance. The second, stimulus involves intrahepatic lipid accumulation by enhanced de novo lipogenesis via reducing the AMPK-ACC signaling axis. These findings identify STK38 as a novel nutrient-sensitive proinflammatory and lipogenic factor in maintaining hepatic energy homeostasis, and it provides a promising target for hepatic and immune health.


Asunto(s)
Dieta Alta en Grasa , Enfermedad del Hígado Graso no Alcohólico , Proteínas Serina-Treonina Quinasas , Animales , Ratones , Dieta Alta en Grasa/efectos adversos , Inflamación/metabolismo , Resistencia a la Insulina/fisiología , Lípidos , Lipogénesis/genética , Hígado/metabolismo , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Hipernutrición , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Serina/metabolismo
16.
Toxicol Appl Pharmacol ; 462: 116424, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36775252

RESUMEN

The NLRP3 inflammasome plays an important role in the pathogenesis of numerous inflammation-related diseases. Benzyl isothiocyanate (BITC) is rich in cruciferous vegetables and possesses potent antioxidant, anti-inflammatory, anti-cancer, and anti-obesogenic properties. In this study, we investigated the role of the NLRP3 inflammasome in the protection by BITC against steatohepatitis and insulin resistance. A mouse model of high-fat/cholesterol/cholic acid diet (HFCCD)-induced steatohepatitis, LPS/nigericin-stimulated primary Kupffer cells, and IL-1ß treated primary hepatocytes were used. BITC attenuated LPS/nigericin-induced activation of the NLRP3 inflammasome by enhancing protein kinase A-dependent NLRP3 ubiquitination, which increased the degradation of NLRP3 and reduced IL-1ß secretion in Kupffer cells. In hepatocytes, BITC pretreatment reversed the IL-1ß-induced decrease in the phosphorylation of IR, AKT, and GSK3ß in response to insulin. After 12 weeks of HFCCD feeding, increases in blood alanine aminotransferase (ALT) and glucose levels were ameliorated by BITC. Hepatic IL-1ß production, macrophage infiltration, and collagen expression induced by HFCCD were also mitigated by BITC. BITC suppresses activation of the NLRP3 inflammasome in Kupffer cells by enhancing the PKA-dependent ubiquitination of NLRP3, which leads to suppression of IL-1ß production and subsequently ameliorates hepatic inflammation and insulin resistance.


Asunto(s)
Resistencia a la Insulina , Enfermedad del Hígado Graso no Alcohólico , Ratones , Animales , Inflamasomas/metabolismo , Macrófagos del Hígado , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Nigericina/metabolismo , Lipopolisacáridos/farmacología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Colesterol/metabolismo , Dieta Alta en Grasa , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Ratones Endogámicos C57BL
17.
Mol Metab ; 69: 101675, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36682412

RESUMEN

OBJECTIVES: Metabolic-associated fatty liver disease (MAFLD) is the most common chronic liver disease that can range from hepatic steatosis to non-alcoholic steatohepatitis (NASH), which can lead to fibrosis and cirrhosis. Recently, ketogenic diet (KD), a low carbohydrate diet, gained popularity as a weight-loss approach, although it has been reported to induce hepatic insulin resistance and steatosis in animal model systems via an undefined mechanism. Herein, we investigated the KD metabolic benefits and its contribution to the pathogenesis of NASH. METHODS: Using metabolic, biochemical and omics approaches, we identified the effects of a KD on NASH and investigated the mechanisms by which KD induces hepatic insulin resistance and steatosis. RESULTS: We demonstrate that KD can induce fibrosis and NASH regardless of body weight loss compared to high-fat diet (HFD) fed mice at thermoneutrality. At ambient temperature (23 °C), KD-fed mice develop a severe hepatic injury, inflammation, and steatosis. In addition, KD increases liver cholesterol, IL-6, and p-JNK and aggravates diet induced-glucose intolerance and hepatic insulin resistance compared to HFD. Pharmacological inhibition of IL-6 and JNK reverses KD-induced glucose intolerance, and hepatic steatosis and restores insulin sensitivity. CONCLUSIONS: Our studies uncover a new mechanism for KD-induced hepatic insulin resistance and NASH potentially via IL-6-JNK signaling and provide a new NASH mouse model.


Asunto(s)
Intolerancia a la Glucosa , Resistencia a la Insulina , Enfermedad del Hígado Graso no Alcohólico , Ratones , Animales , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Intolerancia a la Glucosa/etiología , Interleucina-6 , Dieta Alta en Grasa , Dieta Baja en Carbohidratos
18.
Pharmacol Res Perspect ; 11(1): e01043, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36585794

RESUMEN

Diabetes is a metabolic disorder with an increasing global prevalence. Somatostatin (SST), a peptide hormone, regulates hormone secretion via five SST receptor (SSTR) subtypes (SSTR1-5) in a tissue-specific manner. As SSTR5 is expressed in pancreatic ß-cells and intestinal L-cells, studies have suggested that SSTR5 regulates glucose tolerance through insulin and incretin secretion, thereby having a prominent role in diabetes. Moreover, SSTR5 knockout (KO) mice display enhanced insulin sensitivity; however, the underlying mechanism has not been clarified. Therefore, in this study, we investigate the effect of SSTR5 blockade on insulin resistance and the target organ using SSTR5 KO mice and a selective SSTR5 antagonist (compound-1). High-fat diet (HFD)-fed SSTR5 KO mice exhibited significantly lower homeostasis model assessment of insulin resistance (HOMA-IR) than HFD-fed wild-type mice. Two-week oral administration of compound-1 dose-dependently and significantly reduced changes in the levels of glycosylated hemoglobin (GHb), plasma glucose, plasma insulin, and HOMA-IR in male KK-Ay /Ta Jcl mice (KK-Ay mice), a model of obese type 2 diabetes with severe insulin resistance. Additionally, compound-1 significantly increased the glucose infusion rate while decreasing hepatic glucose production in male KK-Ay mice, as evidenced by hyperinsulinemic-euglycemic clamp analyses. In addition, compound-1 ameliorated the insulin-induced Akt phosphorylation suppression by octreotide in the liver of male C57BL/6J mice. Collectively, our results demonstrate that selective SSTR5 inhibition can improve insulin sensitivity by enhancing liver insulin action; thus, selective SSTR5 antagonists represent potentially novel therapeutic agents for type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Masculino , Ratones , Animales , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Ratones Endogámicos C57BL , Insulina/metabolismo , Glucosa/metabolismo , Hígado/metabolismo , Ratones Noqueados
19.
Artículo en Inglés | WPRIM (Pacífico Occidental) | ID: wpr-1007880

RESUMEN

OBJECTIVE@#In this study, the combined effect of two stressors, namely, electromagnetic fields (EMFs) from mobile phones and fructose consumption, on hypothalamic and hepatic master metabolic regulators of the AMPK/SIRT1-UCP2/FOXO1 pathway were elucidated to delineate the underlying molecular mechanisms of insulin resistance.@*METHODS@#Weaned Wistar rats (28 days old) were divided into 4 groups: Normal, Exposure Only (ExpO), Fructose Only (FruO), and Exposure and Fructose (EF). Each group was provided standard laboratory chow ad libitum for 8 weeks . Additionally, the control groups, namely, the Normal and FruO groups, had unrestricted access to drinking water and fructose solution (15%), respectively. Furthermore, the respective treatment groups, namely, the ExpO and EF groups, received EMF exposure (1,760 MHz, 2 h/day x 8 weeks). In early adulthood, mitochondrial function, insulin receptor signaling, and oxidative stress signals in hypothalamic and hepatic tissues were assessed using western blotting and biochemical analysis.@*RESULT@#In the hypothalamic tissue of EF, SIRT1, FOXO 1, p-PI3K, p-AKT, Complex III, UCP2, MnSOD, and catalase expressions and OXPHOS and GSH activities were significantly decreased ( P < 0.05) compared to the Normal, ExpO, and FruO groups. In hepatic tissue of EF, the p-AMPKα, SIRT1, FOXO1, IRS1, p-PI3K, Complex I, II, III, IV, V, UCP2, and MnSOD expressions and the activity of OXPHOS, SOD, catalase, and GSH were significantly reduced compared to the Normal group ( P < 0.05).@*CONCLUSION@#The findings suggest that the combination of EMF exposure and fructose consumption during childhood and adolescence in Wistar rats disrupts the closely interlinked and multi-regulated crosstalk of insulin receptor signals, mitochondrial OXPHOS, and the antioxidant defense system in the hypothalamus and liver.


Asunto(s)
Humanos , Ratas , Animales , Adulto , Ratas Wistar , Fructosa/metabolismo , Catalasa , Receptor de Insulina/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Campos Electromagnéticos/efectos adversos , Sirtuina 1/metabolismo , Teléfono Celular , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína Forkhead Box O1/metabolismo , Proteína Desacopladora 2
20.
Nutrients ; 14(24)2022 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-36558381

RESUMEN

Type 2 diabetes mellitus (T2DM) is a systemic metabolic disorder characterized by insulin deficiency and insulin resistance. Recently, it has become a significant threat to public health. Polygonatum sibiricum saponin (PSS) has potential hypoglycemic effects, but its specific mechanism needs further study. In this study, PSS significantly decreased the level of blood glucose, water intake, and the organ index in diabetic mice. Meanwhile, PSS effectively reduced the content of total triglyceride (TG), total cholesterol (TCHO), low-density lipoprotein cholesterol (LDL-C), alanine aminotransferase (ALT), and aspartate aminotransferase (AST) in the blood, and increased the content of high-density lipoprotein cholesterol (HDL-C). This suggests that PSS could reduce the content of blood lipids and initially improve the damage of hepatocytes. We found that PSS alleviated hepatic insulin resistance, repaired islet beta cells, and enabled insulin to play its biological role normally. It also improved oral glucose tolerance and abated serum lipopolysaccharide (LPS) and glycosylated hemoglobin (HbA1c) levels in T2DM mice. Furthermore, studies have found that PSS increased the content of phosphorylated protein kinase B (AKT), thereby promoting the effect of glucose transporter 4 (GLUT-4), and activating glycogen synthase kinase 3beta (GSK-3ß) and glycogen synthase (GS) proteins to promote hepatic glycogen synthesis. Finally, we found that PSS could promote the growth of beneficial bacteria such as Bifidobacterium and Lactobacillus, reduce the growth of harmful bacteria such as Enterococcus and Enterobacter, and preliminarily improve the composition of important bacteria in the intestine. These studies indicate that PSS has an excellent hypoglycemic effect, which provides a potential new treatment for T2DM and guidance for more in-depth research.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Polygonatum , Ratones , Animales , Hipoglucemiantes/farmacología , Hipoglucemiantes/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Glucógeno/metabolismo , Hígado/metabolismo , Insulina , Glucemia/metabolismo , Colesterol/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA