RESUMEN
Insulin receptor substrates 1 and 2 (IRS-1 and IRS-2) are signaling adaptor proteins that participate in canonical pathways, where insulin cascade activation occurs, as well as in non-canonical pathways, in which phosphorylation of substrates is carried out by a diverse array of receptors including integrins, cytokines, steroid hormones, and others. IRS proteins are subject to a spectrum of post-translational modifications essential for their activation, encompassing phosphorylation events in distinct tyrosine, serine, and threonine residues. Tyrosine residue phosphorylation is intricately linked to the activation of the insulin receptor cascade and its interaction with SH2 domains within a spectrum of proteins, including PI3K. Conversely, serine residue phosphorylation assumes a different function, serving to attenuate the effects of insulin. In this review, we have identified over 50 serine residues within IRS-1 that have been reported to undergo phosphorylation orchestrated by a spectrum of kinases, thereby engendering the activation or inhibition of different signaling pathways. Furthermore, we delineate the phosphorylation of over 10 distinct tyrosine residues at IRS-1 or IRS-2 in response to insulin, a process essential for signal transduction and the subsequent activation of PI3K.
RESUMEN
Insulin signaling plays an important role in the development and progression of cancer since it is involved in proliferation and migration processes. It has been shown that the A isoform of the insulin receptor (IR-A) is often overexpressed, and its stimulation induces changes in the expression of the insulin receptor substrates (IRS-1 and IRS-2), which are expressed differently in the different types of cancer. We study the participation of the insulin substrates IRS-1 and IRS-2 in the insulin signaling pathway in response to insulin and their involvement in the proliferation and migration of the cervical cancer cell line. Our results showed that under basal conditions, the IR-A isoform was predominantly expressed. Stimulation of HeLa cells with 50 nM insulin led to the phosphorylation of IR-A, showing a statistically significant increase at 30 min (p ≤ 0.05). Stimulation of HeLa cells with insulin induces PI3K and AKT phosphorylation through the activation of IRS2, but not IRS1. While PI3K reached the highest level at 30 min after treatment (p ≤ 0.05), AKT had the highest levels from 15 min (p ≤ 0.05) and remained constant for 6 h. ERK1 and ERK2 expression was also observed, but only ERK2 was phosphorylated in a time-dependent manner, reaching a maximum peak 5 min after insulin stimulation. Although no effect on cell proliferation was observed, insulin stimulation of HeLa cells markedly promoted cell migration.
RESUMEN
BACKGROUND AND AIMS: Currently, treatments are being sought to improve the control of type II diabetes mellitus (T2DM), and inulin has been shown to be effective in reducing glucose levels and other metabolic control parameters. These effects on metabolic control may be associated with changes in the epigenetic modulation of genes of the insulin pathway. Therefore, our objective is to determine the effect of agave inulin in metabolic control parameters and in INS and IRS1 genes' methylation in T2DM patients. METHODS: This was a longitudinal experimental study with 67 Mexican participants who received an intervention of inulin agave (10 g daily) for 2 months. The methylation of the INS and IRS1 genes was determined by MSP. RESULTS: For the INS gene, we found a significant decrease in the proportions of T2DM patients with methylated DNA after inulin intervention (p = 0.0001). In contrast, the difference in the proportions of the unmethylated IRS1 gene before and after the inulin intervention was not significant (p = 0.79). On the other hand, we observed changes in the number of T2DM patients' recommended categories for metabolic control depending on the methylation of INS and IRS1 genes before and after treatment with inulin. CONCLUSION: For the first time, we report the modification in the methylation of two genes, INS and IRS1, of the insulin pathway and provide information on the possible relevant role of epigenetics as a key factor in positive changes in metabolic control parameters by inulin intake in T2DM patients.
Asunto(s)
Diabetes Mellitus Tipo 2 , Humanos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/genética , Inulina/metabolismo , Metilación , Insulina/metabolismo , México , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/metabolismoRESUMEN
BACKGROUND: Insulin resistance (IR) is a condition in which the response of organs to insulin is impaired. IR is an early marker of metabolic dysfunction. However, IR also appears in physiological contexts during critical developmental windows. The molecular mechanisms of physiological IR are largely unknown in both sexes. Sexual dimorphism in insulin sensitivity is observed since early stages of development. We propose that during periods of accelerated growth, such as around weaning, at postnatal day 20 (p20) in rats, the kinase S6K1 is overactivated and induces impairment of insulin signaling in its target organs. This work aimed to characterize IR at p20, determine its underlying mechanisms, and identify whether sexual dimorphism in physiological IR occurs during this stage. METHODS: We determined systemic insulin sensitivity through insulin tolerance tests, glucose tolerance tests, and blood glucose and insulin levels under fasting and fed conditions at p20 and adult male and female Wistar rats. Furthermore, we quantified levels of S6K1 phosphorylated at threonine 389 (T389) (active form) and its target IRS1 phosphorylated at serine 1101 (S1101) (inhibited form). In addition, we assessed insulin signal transduction by measuring levels of Akt phosphorylated at serine 473 (S473) (active form) in white adipose tissue and skeletal muscle through western blot. Finally, we determined the presence and function of GLUT4 in the plasma membrane by measuring the glucose uptake of adipocytes. Results were compared using two-way ANOVA (With age and sex as factors) and one-way ANOVA with post hoc Tukey's tests or t-student test in each corresponding case. Statistical significance was considered for P values < 0.05. RESULTS: We found that both male and female p20 rats have elevated levels of glucose and insulin, low systemic insulin sensitivity, and glucose intolerance. We identified sex- and tissue-related differences in the activation of insulin signaling proteins in p20 rats compared to adult rats. CONCLUSIONS: Male and female p20 rats present physiological insulin resistance with differences in the protein activation of insulin signaling. This suggests that S6K1 overactivation and the resulting IRS1 inhibition by phosphorylation at S1101 may modulate to insulin sensitivity in a sex- and tissue-specific manner. Video Abstract.
Insulin regulates the synthesis of carbohydrates, lipids and proteins differently between males, and females. One of its primary functions is maintaining adequate blood glucose levels favoring glucose entry in muscle and adipose tissue after food consumption. Insulin resistance (IR) is a condition in which the response of organs to insulin is impaired. IR is frequently associated with metabolic dysfunction such as inflammation, obesity, or type 2 diabetes. However, physiological IR develops in healthy individuals during periods of rapid growth, pregnancy, or aging by mechanisms not fully understood. We studied the postnatal development, specifically around weaning at postnatal day 20 (p20) of Wistar rats. In previous works, we identified insulin resistance during this period in male rats. This work aimed to characterize IR at p20, determine its underlying mechanisms, and identify whether sexual dimorphism in physiological IR occurs during this stage. We found that p20 rats of both sexes have elevated blood glucose and insulin levels, low systemic insulin sensitivity, and glucose intolerance. We identified differences in insulin-regulated protein activation (S6K1, IRS1, Akt, and GLUT4) between sexes in different tissues and adipose tissue depots. Studying these mechanisms and their differences between males and females is essential to understanding insulin actions and their relationship with the possible development of metabolic diseases in both sexes.
Asunto(s)
Resistencia a la Insulina , Animales , Glucemia/metabolismo , Femenino , Glucosa/metabolismo , Insulina/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Masculino , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , Serina/metabolismo , Caracteres Sexuales , Treonina/metabolismoRESUMEN
A pro-inflammatory environment is characteristic of obesity and polycystic ovary syndrome (PCOS). This environment through cytokines secretion negatively affects insulin action. Endometria from women with both conditions (obesity and PCOS) present high TNF-α level and altered insulin signaling. In addition, these patients present reproductive failures that could be associated to an abnormal endometrial function. Here, TNF-α and IL-6 effects on insulin signaling pathway were evaluated. Serum and endometrial IL-6, phospho-IRS1-S270 (inactive form) and phospho-IRS1-Y612 (active form) levels were evaluated in women with: Normal-Weight, Obesity and Obesity-PCOS. In endometrial cells under hyperandrogenic/hyperinsulinic conditions resembling PCOS, it was evaluated IL-6/TNF-α effects on phospho-IRS1-S270, phospho-IRS1-Y612, phospho-AKT-S473 levels, and S6K and JNK activation (IRS1-inactivating molecules). In obesity groups, diminution of IRS1-active form was observed, being more significantly in Obesity-PCOS; whereas, IRS1-inactive form increased in Obesity-PCOS. Serum and endometrial IL-6 were higher in Obesity-groups compared to Normal-Weight. In endometrial cells, TNF-α increases phospho-IRS1-S270, while IL-6 decreases phospho-IRS1-Y612. Importantly, TNF-α and IL-6 promote S6K and JNK activation; TNF-α increases and IL-6 decreases phospho-AKT-S473 levels. Thus, pro-inflammatory cytokines in endometrium could negatively influence insulin signaling by different mechanisms: TNF-α promotes activation of IRS1-inactivating kinases, whereas, IL-6 decreases IRS1 and AKT activation. Moreover, when obesity and PCOS are present the disruption of insulin signaling is aggravated. These effects could explain endometrial abnormal function and reproductive failures observed in women with obesity and PCOS.
Asunto(s)
Endometrio/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Interleucina-6/farmacología , Obesidad/metabolismo , Síndrome del Ovario Poliquístico/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Adulto , Línea Celular , Endometrio/efectos de los fármacos , Endometrio/patología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-6/metabolismo , Obesidad/patología , Fosforilación/efectos de los fármacos , Síndrome del Ovario Poliquístico/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Adulto JovenRESUMEN
BACKGROUND: Free fatty acid receptor 1 (FFAR1) is G-protein coupled receptor predominantly expressed in pancreatic ß-cells that is activated by a variety of free fatty acids (FFAs). Once activated, it promotes glucose-stimulated insulin secretion (GSIS). However, increased levels of FFAs lead to lipotoxicity, inducing loss of ß-cell function. FFAR1 plays a key role in the development of type 2 diabetes (T2D), and previous studies have indicated the importance of developing anti-diabetic therapies against FFAR1, although its role in the regulation of ß-cell function remains unclear. The present study investigated the role of FFAR1 under lipotoxic conditions using palmitic acid (PA). The rat insulinoma 1 clone 832/13 (INS-1 832/13) cell line was used as a model as it physiologically resembles native pancreatic ß-cells. Key players of the insulin signaling pathway, such as mTOR, Akt, IRS-1, and the insulin receptor (INSR1ß), were selected as candidates to be analyzed under lipotoxic conditions. RESULTS: We revealed that PA-induced lipotoxicity affected GSIS in INS-1 cells and negatively modulated the activity of both IRS-1 and Akt. Reduced phosphorylation of both IRS-1 S636/639 and Akt S473 was observed, in addition to decreased expression of both INSR1ß and FFAR1. Moreover, transient knockdown of FFAR1 led to a reduction in IRS-1 mRNA expression and an increase in INSR1ß mRNA. Finally, PA affected localization of FFAR1 from the cytoplasm to the perinucleus. CONCLUSIONS: In conclusion, our study suggests a novel regulatory involvement of FFAR1 in crosstalk with mTOR-Akt and IRS-1 signaling in ß-cells under lipotoxic conditions.
Asunto(s)
Células Secretoras de Insulina/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Ácido Palmítico/toxicidad , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Apoptosis , Línea Celular , Células Secretoras de Insulina/metabolismo , Ratas , Transducción de SeñalRESUMEN
The IGF1R/IRS1 signaling is activated in acute lymphoblastic leukemia (ALL) and can be targeted by the pharmacological inhibitors NT157 (IGF1R-IRS1/2 inhibitor) and OSI-906 (IGF1R/IR inhibitor). Here we investigate the cellular and molecular effects of NT157 and OSI-906 in ALL cells. NT157 and OSI-906 treatment reduced viability, proliferation and cell cycle progression in ALL cell lines. Similarly, in primary samples of patients with ALL, both OSI-906 and NT157 reduced viability, but only NT157 induced apoptosis. NT157 and OSI-906 did not show cytotoxicity in primary samples from healthy donor. NT157 and OSI-906 significantly decreased Jurkat cell migration, but did not modulate Namalwa migration. Consistent with the more potent effect of NT157 on cells, NT157 significantly modulated expression of 25 genes related to the MAPK signaling pathway in Jurkat cells, including oncogenes and tumor suppressor genes. Both compounds inhibited mTOR and p70S6K activity, but only NT157 inhibited AKT and 4-EBP1 activation. In summary, in ALL cells, NT157 has cytotoxic activity, whereas OSI-906 is cytostatic. NT157 has a stronger effect on ALL cells, and thus the direct inhibition of IRS1 may be a potential therapeutic target in ALL.
Asunto(s)
Antineoplásicos/farmacología , Imidazoles/farmacología , Proteínas Sustrato del Receptor de Insulina/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Pirazinas/farmacología , Pirogalol/análogos & derivados , Receptor IGF Tipo 1/antagonistas & inhibidores , Sulfonamidas/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Adulto , Anciano , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Proteínas Sustrato del Receptor de Insulina/metabolismo , Células Jurkat , Persona de Mediana Edad , Terapia Molecular Dirigida , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Pirogalol/farmacología , Receptor IGF Tipo 1/metabolismo , Transducción de Señal , Células Tumorales Cultivadas , Adulto JovenRESUMEN
BACKGROUND: Free fatty acid receptor 1 (FFAR1) is G-protein coupled receptor predominantly expressed in pancreatic ß-cells that is activated by a variety of free fatty acids (FFAs). Once activated, it promotes glucose-stimulated insulin secretion (GSIS). However, increased levels of FFAs lead to lipotoxicity, inducing loss of ß-cell function. FFAR1 plays a key role in the development of type 2 diabetes (T2D), and previous studies have indicated the importance of developing anti-diabetic therapies against FFAR1, although its role in the regulation of ß-cell function remains unclear. The present study investigated the role of FFAR1 under lipotoxic conditions using palmitic acid (PA). The rat insulinoma 1 clone 832/13 (INS-1 832/13) cell line was used as a model as it physiologically resembles native pancreatic ß-cells. Key players of the insulin signaling pathway, such as mTOR, Akt, IRS-1, and the insulin receptor (INSR1ß), were selected as candidates to be analyzed under lipotoxic conditions. RESULTS: We revealed that PA-induced lipotoxicity affected GSIS in INS-1 cells and negatively modulated the activity of both IRS-1 and Akt. Reduced phosphorylation of both IRS-1 S636/639 and Akt S473 was observed, in addition to decreased expression of both INSR1ß and FFAR1. Moreover, transient knockdown of FFAR1 led to a reduction in IRS-1 mRNA expression and an increase in INSR1ß; mRNA. Finally, PA affected localization of FFAR1 from the cytoplasm to the perinucleus. CONCLUSIONS: In conclusion, our study suggests a novel regulatory involvement of FFAR1 in crosstalk with mTOR-Akt and IRS-1 signaling in ß-cells under lipotoxic conditions.
Asunto(s)
Animales , Ratas , Ácido Palmítico/toxicidad , Receptores Acoplados a Proteínas G/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Transducción de Señal , Línea Celular , Apoptosis , Células Secretoras de Insulina/metabolismoRESUMEN
Obesity is accompanied by a low-grade inflammation state, characterized by increased proinflammatory cytokines levels such as tumor necrosis factor alpha (TNFα) and interleukin-1 beta (IL-1ß). In this regard, there exists a lack of studies in hepatic tissue about the role of TNFα receptor 1 (TNFR1) in the context of obesity and insulin resistance during the progression of nonalcoholic fatty liver disease (NAFLD). The aim of this work was to evaluate the effects of high-caloric feeding (HFD) (40% fat, for 16 weeks) on liver inflammation-induced apoptosis, insulin resistance, hepatic lipid accumulation and its progression toward nonalcoholic steatohepatitis (NASH) in TNFR1 knock-out and wild-type mice. Mechanisms involved in HFD-derived IL-1ß release and impairment of insulin signaling are still unknown, so we determined whether IL-1ß affects liver insulin sensitivity and apoptosis through TNFα receptor 1 (TNFR1)-dependent pathways. We showed that knocking out TNFR1 induces an enhanced IL-1ß plasmatic release upon HFD feed. This was correlated with higher hepatic and epididymal white adipose tissue mRNA levels. In vivo and in vitro assays confirmed an impairment in hepatic insulin signaling, in part due to IL-1ß-induced decrease of AKT activation and diminution of IRS1 levels, followed by an increase in inflammation, macrophage (resident and recruited) accumulation, hepatocyte apoptotic process and finally hepatic damage. In addition, TNFR1 KO mice displayed higher levels of pro-fibrogenic markers. TNFR1 signaling disruption upon an HFD leads to an accelerated progression from simple steatosis to a more severe phenotype with many NASH features, pointing out a key role of TNFR1 in NAFLD progression.
Asunto(s)
Dieta Alta en Grasa/efectos adversos , Enfermedad del Hígado Graso no Alcohólico/etiología , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Animales , Apoptosis/genética , Insulina/metabolismo , Resistencia a la Insulina , Interleucina-1beta/metabolismo , Hígado/metabolismo , Hígado/patología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/patología , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Transducción de SeñalRESUMEN
Sepsis survivors frequently develop late cognitive impairment. Because little is known on the mechanisms of post-septic memory deficits, there are no current effective approaches to prevent or treat such symptoms. Here, we subjected mice to severe sepsis induced by cecal ligation and puncture (CLP) and evaluated the sepsis-surviving animals in the open field, novel object recognition (NOR), and step-down inhibitory avoidance (IA) task at different times after surgery. Post-septic mice (30 days post-surgery) failed in the NOR and IA tests but exhibited normal performance when re-evaluated 45 days after surgery. Cognitive impairment in post-septic mice was accompanied by reduced hippocampal levels of proteins involved in synaptic plasticity, including synaptophysin, cAMP response element-binding protein (CREB), CREB phosphorylated at serine residue 133 (CREBpSer133), and GluA1 phosphorylated at serine residue 845 (GluA1pSer845). Expression of tumor necrosis factor α (TNF-α) was increased and brain insulin signaling was disrupted, as indicated by increased hippocampal IRS-1 phosphorylation at serine 636 (IRS-1pSer636) and decreased phosphorylation of IRS-1 at tyrosine 465 (IRS-1pTyr465), in the hippocampus 30 days after CLP. Phosphorylation of Akt at serine 473 (AktpSer473) and of GSK3 at serine 9 (GSK3ßpSer9) were also decreased in hippocampi of post-septic animals, further indicating that brain insulin signaling is disrupted by sepsis. We then treated post-septic mice with liraglutide, a GLP-1 receptor agonist with insulinotropic activity, or TDZD-8, a GSK3ß inhibitor, which rescued NOR memory. In conclusion, these results establish that hippocampal inflammation and disrupted insulin signaling are induced by sepsis and are linked to late memory impairment in sepsis survivors.
Asunto(s)
Encéfalo/metabolismo , Disfunción Cognitiva/metabolismo , Insulina/metabolismo , Sepsis/metabolismo , Transducción de Señal/fisiología , Animales , Encéfalo/patología , Disfunción Cognitiva/etiología , Disfunción Cognitiva/patología , Conducta Exploratoria/fisiología , Masculino , Ratones , Sepsis/complicaciones , Sepsis/patologíaRESUMEN
Insulin-like growth factor 1 (IGF1) and its receptor IGF1R regulate normal cell growth and contribute to cell transformation through activation of downstream signaling pathways. In fibroblast cells, insulin receptor substrate 1 (IRS1), through IGF1 signaling, was found to be the key protein for nuclear translocation of ß-catenin and MYC transcription activation. We herein investigated the IRS1/ß-catenin axis in acute lymphoblastic leukemia (ALL) cells. Samples were obtained from 45 patients with ALL and 13 healthy donors. ALL cell lines were used. Gene expression was measured by quantitative PCR. Protein expression, associations, and cellular localization were evaluated by immunoprecipitation, subcellular fractionation, and confocal microscopy. Cells were submitted to IGF1 stimulation and/or IGF1R pharmacological inhibition (OSI-906). IRS1, ß-catenin, and MYC mRNA expression were significantly elevated in ALL patients, compared to normal controls. MYC mRNA expression positively correlated with ß-catenin and IRS1. Increased age and MYC expression negatively affected overall survival by univariate analysis. Total and phospho-IGF1R and IRS1, MYC and ß-catenin protein expression were higher in ALL cells, compared to normal peripheral blood mononuclear cells (PBMC). IRS1 and ß-catenin were found to be colocalized in the nuclei and the cytoplasm of ALL cell lines, whereas both proteins were only slightly detected in the cytoplasm of normal PBMC. In Jurkat cells, a constitutive IRS1 and ß-catenin protein interaction were observed; OSI-906 treatment decreased IGF1R tyrosine phosphorylation, IRS1 expression and phosphorylation, nuclear translocation of ß-catenin, IRS1 and ß-catenin association, and MYC protein expression. In conclusion, the IRS1/ß-catenin axis is activated in ALL cells. J. Cell. Biochem. 118: 1774-1781, 2017. © 2016 Wiley Periodicals, Inc.
Asunto(s)
Proteínas Sustrato del Receptor de Insulina/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , beta Catenina/genética , Transporte Activo de Núcleo Celular/genética , Transporte Activo de Núcleo Celular/fisiología , Adolescente , Adulto , Western Blotting , Humanos , Imidazoles/farmacología , Inmunoprecipitación , Proteínas Sustrato del Receptor de Insulina/metabolismo , Microscopía Confocal , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Pirazinas/farmacología , ARN Mensajero/genética , Receptor IGF Tipo 1 , Receptores de Somatomedina/genética , Receptores de Somatomedina/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Adulto Joven , beta Catenina/metabolismoRESUMEN
Introducción: El sustrato del receptor de insulina 1 (IRS1) es un componente importante de la cascada de transducción de señales de la insulina y podría estar relacionado con los trastornos metabólicos asociados al síndrome metabólico. Objetivo: Evaluar el papel del polimorfismo Gly972Arg del gen IRS1 con factores de riesgo cardiometabólicos en niños pre-púberes. Metodología: Se estudiaron 279 niños con edades comprendidas entre 2-12 años, clasificados según los parámetros antropométricos y bioquímicos en: a) niños obesos sin RI (n=135), b) niños obesos con RI (n=80) y c) niños controles sanos (n=64). A cada niño se le realizó una extracción de sangre en ayunas y una postprandial, para determinar glicemia e insulina basal y postprandial, triglicéridos, colesterol total y fraccionado y la frecuencia genotípica del SNP Gly972Arg. Resultados: Se observó que 37,5% de los niños presentó RI; 9,6% hiperglicemia en ayunas; 27,3% hipertrigliceridemia y 50,46% bajos niveles de HDL-c. La frecuencia genotípica fue 89% genotipo Gly/Gly y 11% genotipo Gly/Arg. Se encontró diferencia significativa en la distribución de los diferentes genotipos del gen de IRS1 en los niños con sobrepeso/obesidad sin RI y niños con sobrepeso/obesidad con RI con respecto al grupo control (OR= 4,47; IC 95%=0,96-16,92; p < 0,05) y (OR= 4,43; IC 95%=0,93-21,00; p < 0,05) respectivamente. Conclusión: Se observó una asociación entre la presencia del genotipo Gly/Arg del gen IRS1 con sobrepeso/obesidad (factor de riesgo cardiometabólico) en los niños del estudio, presentando estos niños 4 veces más riesgos a presentar sobrepeso/obesidad que los niños con el genotipo Gly/Gly.
Introduction: Insulin receptor substrate 1 (IRS-1) is an important component of the insulin signal transduction cascade and could be related with metabolic disorders associated with metabolic syndrome (MS). Aim: Evaluate the role of the Gly972Arg polymorphism in the IRS1 gene in prepubertal children with cardiometabolic risk factors. Methods: We studied 279 children between 2-12 years of age, divided in groups 3 groups: a) obese children without insulin resistance (IR) (n=135), b) obese children with IR (n=80) and c) healthy children as controls (n=64). Basal and postprandial glucose, insulin, triglycerides, total and fractionated cholesterol and genotype frequency of the Gly972Arg SNP were determined in fasting and postprandial samples in each child. Results: 37.5% of the children had IR; in the fasting state, 9.6% had hyperglycemia, 27.3% hypertriglyceridemia and 50.46 % low HDL-C. The genotypic frequency was 89% for the Gly / Gly genotype and 11% Gly / Arg genotype. Significant difference was found in the distribution of the different genotypes of the IRS1 gene in children with overweight/obesity without IR and children with overweight/obesity with IR compared to the control group (OR = 4.47;CI 95% = 0.96-16.92; p <0.05) and (OR = 4.43; CI 95%= 0.93 - 21.00, p <0.05) respectively. Conclusion: Association between the presence of Gly/Arg genotype of the IRS1 gene with overweight/obesity (cardiometabolic risk factor) was observed in the studied children. These children were four times more likely to be overweight/obese than children with Gly / Gly genotype.
RESUMEN
Insulin acts in the hypothalamus, decreasing food intake (FI) by the IR/PI3K/Akt pathway. This pathway is impaired in obese animals and endoplasmic reticulum (ER) stress and low-grade inflammation are possible mechanisms involved in this impairment. Here, we highlighted the amygdala as an important brain region for FI regulation in response to insulin. This regulation was dependent on PI3K/AKT pathway similar to the hypothalamus. Insulin was able to decrease neuropeptide Y (NPY) and increase oxytocin mRNA levels in the amygdala via PI3K, which may contribute to hypophagia. Additionally, obese rats did not reduce FI in response to insulin and AKT phosphorylation was decreased in the amygdala, suggesting insulin resistance. Insulin resistance was associated with ER stress and low-grade inflammation in this brain region. The inhibition of ER stress with PBA reverses insulin action/signaling, decreases NPY and increases oxytocin mRNA levels in the amygdala from obese rats, suggesting that ER stress is probably one of the mechanisms that induce insulin resistance in the amygdala.
RESUMEN
Introducción: la diabetes mellitus tipo 2 es una enfermedad heterogénea y multifactorial, que está determinada por factores genéticos y no genéticos. El sustrato 1 del receptor de la insulina (IRS-1) cumple una función fundamental en la transmisión de la señal insulínica, por tanto sus variantes génicas constituyen blancos importantes en el estudio de la susceptibilidad genética a esta enfermedad en las diferentes poblaciones. Objetivo: explorar el papel de las variantes polimórficas Gly972Arg y Ala513Pro del gen IRS-1 en la susceptibilidad genética de la diabetes mellitus tipo 2 en un grupo de la población cubana.Métodos: se determinó la frecuencia de los polimorfismos Gly972Arg y Ala513Pro del IRS-1 en 499 ciudadanos cubanos, con un índice de masa corporal entre 22-30, con edades comprendidas entre los 40 y 70 años: de ellos 272 (54,5 por ciento) diabéticos y 227 (45,5 por ciento) no diabéticos.Resultados: la frecuencia del alelo Pro513 fue baja (1,2 por ciento) y similar para ambos grupos (1,1 por ciento vs. 1,3 por ciento para el grupo de diabéticos y el grupo control, respectivamente). La frecuencia del polimorfismo Gly972Arg fue de 16,2 por ciento, superior a la reportada para la mayoría de las poblaciones estudiadas. No se encontraron diferencias significativas en la frecuencia del alelo Arg972 entre el grupo de diabéticos y el grupo control (15,4 por ciento vs. 17,3 por ciento), ni cambios en los niveles de glucemia e insulinemia asociados a la presencia del alelo polimórfico Arg972.Conclusiones: en este grupo de sujetos de la población cubana, las variantes polimórficas Ala513Pro y Gly972Arg del gen IRS-1 no participan en la etiología de la diabetes mellitus tipo 2(AU)
Introduction: the type 2 diabetes mellitus is a heterogeneous and multifactor disease determined by genetic and no-genetic factors. The substrate 1 of insulin receptor (IRS-1) has a fundamental function in transmission of insulin signal, thus its genic variants are significant targets in study of genetic susceptibility to this disease in different populations. Objective: to explore the role of the Gly972Arg and Ala513PRo of IRS-1 gen polymorphous variants in the genetic susceptibility of type 2 diabetes mellitus in Cuban population. Methods: the frequency of above mentioned polymorphous variants in 499 Cuban citizens with a 22-30 body mass index aged between 40 to 70 including 272 diabetics (54,5 por ciento) and 227 non-diabetic (45,5 por ciento). Results: frequency of Pro513 allele was low (1,2 por ciento) and similar en both groups (1,1 por ciento versus 1,3 por ciento for diabetic group and the control one, respectively). Frequency of Gly972Arg polymorphism was of 16,2 por ciento, higher than that reported for most of study populations. There were not significant differences in frequency of Arg972 allele between the diabetic group and the control one (15,4 por ciento versus 17,3 por ciento). Also, there were not changes in glycemia and insulinemia levels associated to presence of polymorphous allele. Conclusions: in present group of Cuban population the above mentioned polymorphous variants are not involved in etiology of type 2 diabetes mellitus(AU)
Asunto(s)
Humanos , Frecuencia de los Genes/inmunología , Diabetes Mellitus/genética , Polimorfismo Genético/inmunologíaRESUMEN
Introducción: la diabetes mellitus tipo 2 es una enfermedad heterogénea y multifactorial, que está determinada por factores genéticos y no genéticos. El sustrato 1 del receptor de la insulina (IRS-1) cumple una función fundamental en la transmisión de la señal insulínica, por tanto sus variantes génicas constituyen blancos importantes en el estudio de la susceptibilidad genética a esta enfermedad en las diferentes poblaciones. Objetivo: explorar el papel de las variantes polimórficas Gly972Arg y Ala513Pro del gen IRS-1 en la susceptibilidad genética de la diabetes mellitus tipo 2 en un grupo de la población cubana.Métodos: se determinó la frecuencia de los polimorfismos Gly972Arg y Ala513Pro del IRS-1 en 499 ciudadanos cubanos, con un índice de masa corporal entre 22-30, con edades comprendidas entre los 40 y 70 años: de ellos 272 (54,5 por ciento) diabéticos y 227 (45,5 por ciento) no diabéticos.Resultados: la frecuencia del alelo Pro513 fue baja (1,2 por ciento) y similar para ambos grupos (1,1 por ciento vs. 1,3 por ciento para el grupo de diabéticos y el grupo control, respectivamente). La frecuencia del polimorfismo Gly972Arg fue de 16,2 por ciento, superior a la reportada para la mayoría de las poblaciones estudiadas. No se encontraron diferencias significativas en la frecuencia del alelo Arg972 entre el grupo de diabéticos y el grupo control (15,4 por ciento vs. 17,3 por ciento), ni cambios en los niveles de glucemia e insulinemia asociados a la presencia del alelo polimórfico Arg972.Conclusiones: en este grupo de sujetos de la población cubana, las variantes polimórficas Ala513Pro y Gly972Arg del gen IRS-1 no participan en la etiología de la diabetes mellitus tipo 2(AU)
Introduction: the type 2 diabetes mellitus is a heterogeneous and multifactor disease determined by genetic and no-genetic factors. The substrate 1 of insulin receptor (IRS-1) has a fundamental function in transmission of insulin signal, thus its genic variants are significant targets in study of genetic susceptibility to this disease in different populations. Objective: to explore the role of the Gly972Arg and Ala513PRo of IRS-1 gen polymorphous variants in the genetic susceptibility of type 2 diabetes mellitus in Cuban population. Methods: the frequency of above mentioned polymorphous variants in 499 Cuban citizens with a 22-30 body mass index aged between 40 to 70 including 272 diabetics (54,5 por ciento) and 227 non-diabetic (45,5 por ciento). Results: frequency of Pro513 allele was low (1,2 por ciento) and similar en both groups (1,1 por ciento versus 1,3 por ciento for diabetic group and the control one, respectively). Frequency of Gly972Arg polymorphism was of 16,2 por ciento, higher than that reported for most of study populations. There were not significant differences in frequency of Arg972 allele between the diabetic group and the control one (15,4 por ciento versus 17,3 por ciento). Also, there were not changes in glycemia and insulinemia levels associated to presence of polymorphous allele. Conclusions: in present group of Cuban population the above mentioned polymorphous variants are not involved in etiology of type 2 diabetes mellitus(AU)
Asunto(s)
Humanos , Polimorfismo Genético/inmunología , Predisposición Genética a la Enfermedad/epidemiología , Diabetes Mellitus Tipo 2/etiología , Índice de Masa CorporalRESUMEN
La diabetes tipo 2 (DT2) es elevada en Yucatán; 52% de los afectados presentan falla al tratamiento con sulfonilureas y metformina. Una posible explicación es por polimorfismos en los genes IRS1, CAPN10, PPARG2, involucrados en la disfunción de la célula b pancreática y respuesta baja a la acción de insulina. Se determinó la asociación de los polimorfismos Gly972Arg, SNP43 y Pro12Ala con el riesgo a la falla al tratamiento con sulfonilurea y metformina, en pacientes con DT2 de Yucatán, México. Se estudiaron ciento treinta y dos pacientes, clasificados con base al control de la hiperglucemia con sulfonilureas y metformina, en grupos de respondedores (HbA1c<8%) y no respondedores (HbA1c > 8%) al tratamiento. De cada sujeto, se obtuvieron datos demográficos, antropométricos, clínicos y metabólicos. Los polimorfismos se identificaron mediante el análisis del ADN por PCR/RFLP y PCR/OAL. Se calcularon las frecuencias genotípicas y alélicas y el equilibrio de Hardy-Weinberg. Se analizó estadísticamente con X² y regresión logística múltiple (Epi-Info 2000 y SPSS versión 12). Se observó diferencia significativa (p = 0,027) en el riesgo a la falla al tratamiento 4,69 veces mayor en sujetos obesos con genotipo AA SNP43, comparado con sujetos con genotipo GA: X² (OR= 4,69, IC: 1,15-20,59) y regresión logística múltiple, p= 0,048, (OR= 3,72, IC: 1,009-13,718). Se identificó interacción entre el genotipo AA y el IMC>27 (p=0,009). Los hallazgos sugieren que el polimorfismo SNP43 podría influir en la respuesta al tratamiento con sulfonilureas y metformina, con expresión dependiente de obesidad.
In Yucatán, 52% of patients with type 2 diabetes (DT2) present secondary failure to treatment associated with sulphonylurea and metformin. A possible explanation may be due to polymorphisms in the genes IRS1, CAPN10, PPARG2, which are involved in pancreatic b cell dysfunction and a poor response to the action of insulin. The association of the polymorphisms Gly972Arg, SNP43, and Pro12Ala, of the genes IRS1, CAPN10, PPARG2, with the risk of failure to sulphonylurea and metformin therapies was determinated in patients with DT2 in Yucatán, México. One hundred and thirty and two subjects with DT2 were classified in groups of responders (HbA1c< 8%) and non-responders (HbA1c> 8%) to the treatment, according to the control of hyperglucemia with sulphonylurea and metformin. Demographic, anthropometric and metabolic data were obtained from each subject. The polymorphisms were identified by means of DNA analysis by PCR/RFLP and PCR/OAL. Genotypic and allelic frequencies and the Hardy-Weinberg equilibrium were determined. Statistical analyses consisted of X² and multiple logistic regression tests (Epi-Info 2000 and SPSS version 12). Obese subjects carrying the genotype AA SNP43 showed 4.69 times more risk of failure to respond to treatment (p=0.027), when compared with subjects sharing GA genotype: X² (OR= 4.69, IC: 1.15-20.59) and multiple logistic regression, p= 0.048, (OR= 3.72, IC: 1.009-13.718). The interaction between genotype AA and the BMI> 27 showed also a significant difference (p=0.009). The findings suggest the fact that polymorphism SNP43 may influence the response to treatment with sulphonylurea and metformin, the expression being dependent on obesity.