Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Trends Endocrinol Metab ; 35(6): 466-477, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38861922

RESUMEN

The current paradigm for the insulin system focuses on the phenomenon of glucose-stimulated insulin secretion and insulin action on blood glucose control. This historical glucose-centric perspective may have introduced a conceptual bias in our understanding of insulin regulation. A body of evidence demonstrating that in vivo variations in blood glucose and insulin secretion can be largely dissociated motivated us to reconsider the fundamental design of the insulin system as a control system for metabolic homeostasis. Here, we propose that a minimal glucose-centric model does not accurately describe the physiological behavior of the insulin system and propose a new paradigm focusing on the effects of incretins, arguing that under fasting conditions, insulin is regulated by an adipoincretin effect.


Asunto(s)
Secreción de Insulina , Insulina , Humanos , Secreción de Insulina/fisiología , Insulina/metabolismo , Animales , Homeostasis/fisiología , Incretinas/metabolismo , Glucemia/metabolismo
2.
Cell Rep ; 43(5): 114132, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38656871

RESUMEN

Insulin-PI3K signaling controls insulin secretion. Understanding this feedback mechanism is crucial for comprehending how insulin functions. However, the role of adipocyte insulin-PI3K signaling in controlling insulin secretion in vivo remains unclear. Using adipocyte-specific PI3Kα knockout mice (PI3KαAdQ) and a panel of isoform-selective PI3K inhibitors, we show that PI3Kα and PI3Kß activities are functionally redundant in adipocyte insulin signaling. PI3Kß-selective inhibitors have no effect on adipocyte AKT phosphorylation in control mice but blunt it in adipocytes of PI3KαAdQ mice, demonstrating adipocyte-selective pharmacological PI3K inhibition in the latter. Acute adipocyte-selective PI3K inhibition increases serum free fatty acid (FFA) and potently induces insulin secretion. We name this phenomenon the adipoincretin effect. The adipoincretin effect operates in fasted mice with increasing FFA and decreasing glycemia, indicating that it is not primarily a control system for blood glucose. This feedback control system defines the rates of adipose tissue lipolysis and chiefly controls basal insulin secretion during fasting.


Asunto(s)
Adipocitos , Ayuno , Secreción de Insulina , Insulina , Ratones Noqueados , Fosfatidilinositol 3-Quinasas , Animales , Adipocitos/metabolismo , Insulina/metabolismo , Ratones , Ayuno/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Ácidos Grasos no Esterificados/metabolismo , Ácidos Grasos no Esterificados/sangre , Lipólisis , Masculino , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratones Endogámicos C57BL , Fosforilación , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología
3.
Cell Rep ; 43(4): 113960, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38507407

RESUMEN

GFRAL-expressing neurons actuate aversion and nausea, are targets for obesity treatment, and may mediate metformin effects by long-term GDF15-GFRAL agonism. Whether GFRAL+ neurons acutely regulate glucose and energy homeostasis is, however, underexplored. Here, we report that cell-specific activation of GFRAL+ neurons using a variety of techniques causes a torpor-like state, including hypothermia, the release of stress hormones, a shift from glucose to lipid oxidation, and impaired insulin sensitivity, glucose tolerance, and skeletal muscle glucose uptake but augmented glucose uptake in visceral fat. Metabolomic analysis of blood and transcriptomics of muscle and fat indicate alterations in ketogenesis, insulin signaling, adipose tissue differentiation and mitogenesis, and energy fluxes. Our findings indicate that acute GFRAL+ neuron activation induces endocrine and gluco- and thermoregulatory responses associated with nausea and torpor. While chronic activation of GFRAL signaling promotes weight loss in obesity, these results show that acute activation of GFRAL+ neurons causes hypothermia and hyperglycemia.


Asunto(s)
Glucosa , Hipotermia , Náusea , Neuronas , Letargo , Animales , Neuronas/metabolismo , Náusea/metabolismo , Hipotermia/metabolismo , Letargo/fisiología , Glucosa/metabolismo , Ratones , Masculino , Músculo Esquelético/metabolismo , Ratones Endogámicos C57BL , Insulina/metabolismo , Resistencia a la Insulina , Transducción de Señal
4.
Curr Top Microbiol Immunol ; 436: 311-336, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36243850

RESUMEN

The PI3K/AKT signaling module is recruited by several receptors implicated in maintaining tissue and metabolic homeostasis and signaling pathways controlling immune responses. Constitutive activation of PI3K/AKT signaling leads to tissue overgrowth and is frequently observed in cancer cells, whereas reduced PI3K/AKT signaling is associated with diabetes and growth defects. Thus, a critical roadblock to effective PI3K-targeted therapy comes from the crucial role of PI3K/AKT signaling in systemic metabolic homeostasis. This chapter describes the role of PI3K/AKT in insulin signaling and metabolic homeostasis and the interplay between insulin action and metabolic feedback loops that cause resistance to PI3K-targeted therapies. Furthermore, we provide examples of insulin-independent roles for PI3K/AKT in metabolic homeostasis, and some generalizations on the action of PI3K/AKT signaling at the interface of signaling and metabolism are derived. Finally, the specific roles for different class I PI3K isoforms in controlling systemic metabolic homeostasis and energy balance are discussed. We conclude that defining the functional specificities and redundancies of different class I PI3K isoforms in pathways driving disease and controlling metabolic homeostasis is fundamental to develop novel PI3K-targeted therapies.


Asunto(s)
Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Insulina/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
5.
FASEB J ; 36(9): e22512, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36001064

RESUMEN

The kinase IKKß controls pro-inflammatory gene expression, and its activity in the liver and leukocytes was shown to drive metabolic inflammation and insulin resistance in obesity. However, it was also proposed that liver IKKß signaling protects obese mice from insulin resistance and endoplasmic reticulum (ER) stress by increasing XBP1s protein stability. Furthermore, mice lacking IKKß in leukocytes display increased lethality to lipopolysaccharides. This study aims at improving our understanding of the role of IKKß signaling in obesity. We induced IKKß deletion in hematopoietic cells and liver of obese mice by Cre-LoxP recombination, using an INF-inducible system, or a liver-specific IKKß deletion in obese mice by adenovirus delivery of the Cre recombinase. The histopathological, immune, and metabolic phenotype of the mice was characterized. IKKß deletion in the liver and hematopoietic cells was not tolerated in mice with established obesity exposed to the TLR3 agonist poly(I:C) and exacerbated liver damage and ER-stress despite elevated XBP1s. By contrast, liver-specific ablation of IKKß in obese mice reduced steatosis and improved insulin sensitivity in association with increased XBP1s protein abundance and reduced expression of de-novo lipogenesis genes. We conclude that IKKß blockage in liver and leukocytes is not tolerated in obese mice exposed to TLR3 agonists. However, selective hepatic IKKß ablation improves fatty liver and insulin sensitivity in association with increased XBP1s protein abundance and reduced expression of lipogenic genes.


Asunto(s)
Hígado Graso , Resistencia a la Insulina , Animales , Hígado Graso/metabolismo , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Leucocitos/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/metabolismo , Receptor Toll-Like 3/metabolismo
6.
Nat Metab ; 4(1): 60-75, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35102341

RESUMEN

Fatty liver disease (FLD) is a growing health issue with burdening unmet clinical needs. FLD has a genetic component but, despite the common variants already identified, there is still a missing heritability component. Using a candidate gene approach, we identify a locus (rs71519934) at the Pleckstrin and Sec7 domain-containing 3 (PSD3) gene resulting in a leucine to threonine substitution at position 186 of the protein (L186T) that reduces susceptibility to the entire spectrum of FLD in individuals at risk. PSD3 downregulation by short interfering RNA reduces intracellular lipid content in primary human hepatocytes cultured in two and three dimensions, and in human and rodent hepatoma cells. Consistent with this, Psd3 downregulation by antisense oligonucleotides in vivo protects against FLD in mice fed a non-alcoholic steatohepatitis-inducing diet. Thus, translating these results to humans, PSD3 downregulation might be a future therapeutic option for treating FLD.


Asunto(s)
Susceptibilidad a Enfermedades , Hígado Graso/etiología , Hígado Graso/metabolismo , Regulación de la Expresión Génica , Factores de Intercambio de Guanina Nucleótido/genética , Alelos , Animales , Biomarcadores , Línea Celular , Hígado Graso/patología , Perfilación de la Expresión Génica , Variación Genética , Genotipo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Hepatocitos/metabolismo , Humanos , Hígado/metabolismo , Hígado/patología , Ratones , Polimorfismo de Nucleótido Simple , RNA-Seq , Ribonucleasas
7.
JHEP Rep ; 3(6): 100359, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34704005

RESUMEN

BACKGROUND & AIMS: Phosphatidylinositides-3 kinases (PI3Ks) are promising drug targets for cancer therapy, but blockage of PI3K-AKT signalling causes hyperglycaemia, hyperinsulinaemia, and liver damage in patients, and hepatocellular carcinoma (HCC) in mice. There are 4 PI3Ks: PI3Kα, PI3Kß, PI3Kδ, and PI3Kγ. The role of PI3Kγ in HCC is unknown. METHODS: We performed histopathological, metabolic, and molecular phenotyping of mice with genetic ablation of PI3Kγ using models where HCC was initiated by the carcinogen diethylnitrosamine (DEN) and promoted by dietary or genetic obesity (ob/ob). The role of PI3Kγ in leucocytes was investigated in mice lacking PI3Kγ in haematopoietic and endothelial cells. RESULTS: Loss of PI3Kγ had no effects on the development of DEN-induced HCC in lean mice. However, in mice injected with DEN and placed on an obesogenic diet, PI3Kγ ablation reduced tumour growth, which was associated with reduced insulinaemia, steatosis, and expression of inflammatory cytokines. ob/ob mice lacking PI3Kγ, and mice with diet-induced obesity lacking PI3Kγ in leucocytes and endothelial cells did not display improved insulin sensitivity, steatosis, metabolic inflammation, or reduced tumour growth. However, these mice showed a reduced number of tumours, reduced liver infiltration by neutrophils, and reduced hepatocyte proliferation acutely induced by DEN. CONCLUSIONS: Loss of PI3Kγ reduces tumour development in obesity-promoted HCC through multiple cell types and mechanisms that include improved insulinaemia, steatosis, and metabolic inflammation as well as the regulation of acute neutrophil infiltration and compensatory hepatocyte proliferation. PI3Kγ-selective inhibition may represent a novel therapeutic approach to reduce HCC initiation and slow HCC progression. LAY SUMMARY: Class-1 phosphatidylinositides-3 kinases (PI3Ks) are critical targets in cancer therapy, but complete inhibition of all isoforms causes liver damage, hyperglycaemia, and insulinaemia. Here we show that selective ablation of the PI3Kγ isoform dampens tumour initiation and growth in a mouse model of carcinogen-initiated and obesity-promoted hepatocellular carcinoma (HCC). The effect of PI3Kγ ablation on reduced tumour growth was explained by reduced tumour cell proliferation, which was associated with reduced insulin levels, liver lipids, and reduced expression of tumour-promoting cytokines. PI3Kγ ablation in leucocytes of obese mice had no effects on tumour size. However, it reduced tumour number in association with reduced carcinogen-induced neutrophil infiltration and hepatocyte proliferation in livers of obese mice. Inhibition of PI3Kγ may thus reduce HCC initiation and growth in obese subjects by a mechanism involving reduced metabolic stress and insulinaemia and reduced carcinogen-induced neutrophil infiltration to the fatty liver.

8.
EBioMedicine ; 65: 103264, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33712379

RESUMEN

BACKGROUND: It is commonly accepted that in obesity free fatty acids (FFA) cause insulin resistance and hyperglycemia, which drives hyperinsulinemia. However, hyperinsulinemia is observed in subjects with normoglycaemia and thus the paradigm above should be reevaluated. METHODS: We describe two studies: MD-Lipolysis, a case control study investigating the mechanisms of obesity-driven insulin resistance by a systemic metabolic analysis, measurements of adipose tissue lipolysis by microdialysis, and adipose tissue genomics; and POEM, a cohort study used for validating differences in circulating metabolites in relation to adiposity and insulin resistance observed in the MD-Lipolysis study. FINDINGS: In insulin-resistant obese with normal glycaemia from the MD-Lipolysis study, hyperinsulinemia was associated with elevated FFA. Lipolysis, assessed by glycerol release per adipose tissue mass or adipocyte surface, was similar between obese and lean individuals. Adipose tissue from obese subjects showed reduced expression of genes mediating catecholamine-driven lipolysis, lipid storage, and increased expression of genes driving hyperplastic growth. In the POEM study, FFA levels were specifically elevated in obese-overweight subjects with normal fasting glucose and high fasting levels of insulin and C-peptide. INTERPRETATION: In obese subjects with normal glycaemia elevated circulating levels of FFA at fasting are the major metabolic derangement candidate driving fasting hyperinsulinemia. Elevated FFA in obese with normal glycaemia were better explained by increased fat mass rather than by adipose tissue insulin resistance. These results support the idea that hyperinsulinemia and insulin resistance may develop as part of a homeostatic adaptive response to increased adiposity and FFA. FUNDING: Swedish-Research-Council (2016-02660); Diabetesfonden (DIA2017-250; DIA2018-384; DIA2020-564); Novo-Nordisk-Foundation (NNF17OC0027458; NNF19OC0057174); Cancerfonden (CAN2017/472; 200840PjF); Swedish-ALF-agreement (2018-74560).


Asunto(s)
Ácidos Grasos no Esterificados/metabolismo , Hiperinsulinismo/patología , Obesidad/patología , Tejido Adiposo/metabolismo , Estudios de Casos y Controles , Estudios de Cohortes , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/patología , Ácidos Grasos no Esterificados/sangre , Femenino , Regulación de la Expresión Génica , Glicerol/sangre , Glicerol/metabolismo , Humanos , Hiperinsulinismo/complicaciones , Insulina/sangre , Resistencia a la Insulina , Lipólisis , Masculino , Persona de Mediana Edad , Obesidad/complicaciones , Análisis de Componente Principal
9.
FASEB Bioadv ; 3(2): 94-107, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33615154

RESUMEN

The cJun N-terminal Kinases (JNK) emerged as a major link between obesity and insulin resistance, but their role in the loss of pancreatic ß-cell mass and function driving the progression from insulin resistance to type-2 diabetes and in the complications of diabetes was not investigated to the same extent. Furthermore, it was shown that pan-JNK inhibition exacerbates kidney damage in the db/db model of obesity-driven diabetes. Here we investigate the role of JNK1 in the db/db model of obesity-driven type-2 diabetes. Mice with systemic ablation of JNK1 (JNK1-/-) were backcrossed for more than 10 generations in db/+ C57BL/KS mice to generate db/db-JNK1-/- mice and db/db control mice. To define the role of JNK1 in the loss of ß-cell mass and function occurring during obesity-driven diabetes we performed comprehensive metabolic phenotyping, evaluated steatosis and metabolic inflammation, performed morphometric and cellular composition analysis of pancreatic islets, and evaluated kidney function in db/db-JNK1-/- mice and db/db controls. db/db-JNK1-/- mice and db/db control mice developed insulin resistance, fatty liver, and metabolic inflammation to a similar extent. However, db/db-JNK1-/- mice displayed better glucose tolerance and improved insulin levels during glucose tolerance test, higher pancreatic insulin content, and larger pancreatic islets with more ß-cells than db/db mice. Finally, albuminuria, kidney histopathology, kidney inflammation and oxidative stress in db/db-JNK1-/- mice and in db/db mice were similar. Our data indicate that selective JNK1 ablation improves glucose tolerance in db/db mice by reducing the loss of functional ß-cells occurring in the db/db mouse model of obesity-driven diabetes, without significantly affecting metabolic inflammation, steatosis, and insulin sensitivity. Furthermore, we have found that, differently from what previously reported for pan-JNK inhibitors, selective JNK1 ablation does not exacerbate kidney dysfunction in db/db mice. We conclude that selective JNK1 inactivation may have a superior therapeutic index than pan-JNK inhibition in obesity-driven diabetes.

10.
Sci Rep ; 10(1): 12031, 2020 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-32694512

RESUMEN

Hepatoma cell lines are widely used to model the hepatocyte for insulin signaling and fatty liver disease. However, a direct comparison of insulin action in primary hepatocytes and in hepatoma cell lines is needed to validate this model and to better understand liver cancer. Here we have investigated insulin signaling, gluconeogenic gene expression, glucose production, and fatty acid synthase abundance in primary hepatocytes and in HepG2, Hepa 1-6, and McARH7777 hepatoma cell lines. Differences in the electrophoretic profiles of protein extracts from human and mouse primary hepatocytes and the hepatoma cells lines are shown. Compared to primary hepatocytes, hepatoma cells showed high basal phosphorylation of AKT at Thr 308 and constitutively activated RAS-MAPK signaling, which were resistant to the dominant negative Ras mutant H-Ras17N. Hepatoma cell lines also showed defective expression of gluconeogenic enzymes, insulin unresponsive GSK phosphorylation, and marginal glucose production. Hepatoma cells also showed lower protein levels of fatty acid synthase and a largely distinct protein electrophoresis profile from hepatocytes but similar between different hepatoma lines. We conclude that hepatoma cell lines do not accurately model the hepatocyte for insulin action but may be valuable tools to investigate the proteomic changes conferring to hepatocellular carcinoma its peculiar metabolisms.


Asunto(s)
Glucosa/metabolismo , Hepatocitos/metabolismo , Insulina/metabolismo , Fenotipo , Animales , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Humanos , Neoplasias Hepáticas/metabolismo , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Proteínas ras/metabolismo
11.
Cell Metab ; 29(6): 1400-1409.e5, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-30982732

RESUMEN

Phosphatidylinositol-3-kinase (PI3K) activity is aberrant in tumors, and PI3K inhibitors are investigated as cancer therapeutics. PI3K signaling mediates insulin action in metabolism, but the role of PI3K isoforms in insulin signaling remains unresolved. Defining the role of PI3K isoforms in insulin signaling is necessary for a mechanistic understanding of insulin action and to develop PI3K inhibitors with optimal therapeutic index. We show that insulin-driven PI3K-AKT signaling depends on redundant PI3Kα and PI3Kß activities, whereas PI3Kδ and PI3Kγ are largely dispensable. We have also found that RAS activity promotes AKT phosphorylation in insulin-stimulated hepatocytes and that promotion of insulin-driven AKT phosphorylation by RAS depends on PI3Kα. These findings reveal the detailed mechanism by which insulin activates AKT, providing an improved mechanistic understanding of insulin signaling. This improved model for insulin signaling predicts that isoform-selective PI3K inhibitors discriminating between PI3Kα and PI3Kß should be dosed below their hyperglycemic threshold to achieve isoform selectivity.


Asunto(s)
Hepatocitos/metabolismo , Insulina/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas ras/fisiología , Animales , Células Cultivadas , Células HEK293 , Hepatocitos/efectos de los fármacos , Humanos , Insulina/metabolismo , Insulina/farmacología , Isoenzimas/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosfatidilinositol 3-Quinasas/genética , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteínas ras/genética
12.
FASEB J ; 32(1): 319-329, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28904022

RESUMEN

PI3Kγ has emerged as a promising target for the treatment of obesity and insulin resistance; however, previous studies have indicated that PI3Kγ activity in pancreatic ß cells is required for normal insulin secretion in response to glucose. Hence, a possible deterioration of insulin secretion capacity in patients who are predisposed to the failure of pancreatic ß-cell function is a major concern for the pharmacologic inhibition of PI3Kγ. To address this issue, we investigated the effects of PI3Kγ ablation in db/db diabetic mice, a genetic model of obesity-driven ß-cell failure and diabetes. Mice that lacked PI3Kγ were backcrossed into db/+ mice C57BL/KS (>10 generations) to obtain db/db-PI3Kγ-/- mice. db/db-PI3Kγ-/- mice and control db/db mice were phenotyped for glucose homeostasis, insulin sensitivity, insulin secretion, steatosis, metabolic inflammation, pancreatic islet morphometry, islet cellular composition, and inflammation. Pancreatic ß-cell apoptosis and proliferation were also evaluated. db/db-PI3Kγ -/- mice and control db/db mice developed similar body weight, steatosis, glycemia, and insulin levels after a glucose load; however, db/db-PI3Kγ-/- mice displayed improved insulin tolerance, higher levels of fasting serum insulin, and lower pancreatic insulin content. In db/db-PI3Kγ-/- mice, the number of adipose tissue macrophages was similar to control, but displayed reduced adipose tissue neutrophils and M2-polarized adipose tissue gene expression. Finally, db/db-PI3Kγ-/- mice have more pancreatic ß cells and larger islets than db/db mice, despite displaying similar islet inflammation. This phenotype could be explained by reduced ß-cell apoptosis in db/db-PI3Kγ-/- mice compared with control db/db mice. Our results are consistent with the concept that the beneficial action of PI3Kγ ablation in obesity-driven glucose intolerance is largely a result of its leptin-dependent effects on adiposity and, to a lesser extent, the promotion of adipose tissue neutrophil recruitment and M1 polarization of gene expression. Of importance, our data challenge the concept that PI3Kγ is required for insulin secretion in response to glucose in vivo, and indicate that PI3Kγ ablation protects db/db mice from ß-cell apoptosis and improves fasting insulin levels. We conclude that PI3Kγ inhibition in obese patients who are predisposed to ß-cell failure is not expected to produce adverse effects on insulin secretion.-Breasson, L., Sardi, C., Becattini, B., Zani, F., Solinas, G. PI3Kγ ablation does not promote diabetes in db/db mice, but improves insulin sensitivity and reduces pancreatic ß-cell apoptosis.


Asunto(s)
Diabetes Mellitus Experimental/etiología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Tejido Adiposo/patología , Animales , Apoptosis , Glucemia/metabolismo , Fosfatidilinositol 3-Quinasa Clase Ib/deficiencia , Fosfatidilinositol 3-Quinasa Clase Ib/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Expresión Génica , Transportador de Glucosa de Tipo 2/genética , Insulina/sangre , Insulina/genética , Resistencia a la Insulina/genética , Resistencia a la Insulina/fisiología , Células Secretoras de Insulina/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Neutrófilos/patología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Obesidad/metabolismo , Obesidad/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo
13.
Sci Signal ; 10(488)2017 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-28720716

RESUMEN

The phosphoinositide 3-kinase γ (PI3Kγ) plays a major role in leukocyte recruitment during acute inflammation and has been proposed to inhibit classical macrophage activation by driving immunosuppressive gene expression. PI3Kγ plays an important role in diet-induced obesity and insulin resistance. In seeking to determine the underlying molecular mechanisms, we showed that PI3Kγ action in high-fat diet-induced inflammation and insulin resistance depended largely on its role in the control of adiposity, which was due to PI3Kγ activity in a nonhematopoietic cell type. However, PI3Kγ activity in leukocytes was required for efficient neutrophil recruitment to adipose tissue. Neutrophil recruitment was correlated with proinflammatory gene expression in macrophages in adipose tissue, which triggered insulin resistance early during the development of obesity. Our data challenge the concept that PI3Kγ is a general suppressor of classical macrophage activation and indicate that PI3Kγ controls macrophage gene expression by non-cell-autonomous mechanisms, the outcome of which is context-dependent.


Asunto(s)
Tejido Adiposo/patología , Fosfatidilinositol 3-Quinasa Clase Ib/fisiología , Dieta Alta en Grasa/efectos adversos , Inflamación/prevención & control , Resistencia a la Insulina , Leucocitos/enzimología , Obesidad/complicaciones , Animales , Perfilación de la Expresión Génica , Inflamación/etiología , Leucocitos/patología , Metabolismo de los Lípidos , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/fisiopatología , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo
14.
Mol Metab ; 6(2): 174-184, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28180059

RESUMEN

BACKGROUND: The cJun-N-terminal-kinase (JNK) plays a central role in the cell stress response, with outcomes ranging from cell death to cell proliferation and survival, depending on the specific context. JNK is also one of the most investigated signal transducers in obesity and insulin resistance, and studies have identified new molecular mechanisms linking obesity and insulin resistance. Emerging evidence indicates that whereas JNK1 and JNK2 isoforms promote the development of obesity and insulin resistance, JNK3 activity protects from excessive adiposity. Furthermore, current evidence indicates that JNK activity within specific cell types may, in specific stages of disease progression, promote cell tolerance to the stress associated with obesity and type-2 diabetes. SCOPE OF REVIEW: This review provides an overview of the current literature on the role of JNK in the progression from obesity to insulin resistance, NAFLD, type-2 diabetes, and diabetes complications. MAJOR CONCLUSION: Whereas current evidence indicates that JNK1/2 inhibition may improve insulin sensitivity in obesity, the role of JNK in the progression from insulin resistance to diabetes, and its complications is largely unresolved. A better understanding of the role of JNK in the stress response to obesity and type-2 diabetes, and the development of isoform-specific inhibitors with specific tissue distribution will be necessary to exploit JNK as possible drug target for the treatment of type-2 diabetes.


Asunto(s)
Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , Animales , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Humanos , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Obesidad/metabolismo , Estrés Oxidativo/fisiología , Transducción de Señal , Estrés Fisiológico/fisiología
15.
Oncotarget ; 8(63): 106145-106146, 2017 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-29290924
16.
FASEB J ; 30(9): 3124-32, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27230858

RESUMEN

Obesity and insulin resistance are associated with oxidative stress, which may be implicated in the progression of obesity-related diseases. The kinase JNK1 has emerged as a promising drug target for the treatment of obesity and type 2 diabetes. JNK1 is also a key mediator of the oxidative stress response, which can promote cell death or survival, depending on the magnitude and context of its activation. In this article, we describe a study in which the long-term effects of JNK1 inactivation on glucose homeostasis and oxidative stress in obese mice were investigated for the first time. Mice lacking JNK1 (JNK1(-/-)) were fed an obesogenic high-fat diet (HFD) for a long period. JNK1(-/-) mice fed an HFD for the long term had reduced expression of antioxidant genes in their skin, more skin oxidative damage, and increased epidermal thickness and inflammation compared with the effects in control wild-type mice. However, we also observed that the protection from obesity, adipose tissue inflammation, steatosis, and insulin resistance, conferred by JNK1 ablation, was sustained over a long period and was paralleled by decreased oxidative damage in fat and liver. We conclude that compounds targeting JNK1 activity in brain and adipose tissue, which do not accumulate in the skin, may be safer and most effective.-Becattini, B., Zani, F., Breasson, L., Sardi, C., D'Agostino, V. G., Choo, M.-K., Provenzani, A., Park, J. M., Solinas, G. JNK1 ablation in mice confers long-term metabolic protection from diet-induced obesity at the cost of moderate skin oxidative damage.


Asunto(s)
Grasas de la Dieta/efectos adversos , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Obesidad/inducido químicamente , Piel/patología , Envejecimiento , Animales , Grasas de la Dieta/administración & dosificación , Hígado Graso/inducido químicamente , Hígado Graso/metabolismo , Inflamación/metabolismo , Ratones , Ratones Noqueados , Proteína Quinasa 8 Activada por Mitógenos/genética , Estrés Oxidativo
17.
Mol Metab ; 2(3): 292-305, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24049741

RESUMEN

The interplay between hepatic glycogen metabolism and blood glucose levels is a paradigm of the rhythmic nature of metabolic homeostasis. Here we show that mice lacking a functional PER2 protein (Per2 (Brdm1) ) display reduced fasting glycemia, altered rhythms of hepatic glycogen accumulation, and altered rhythms of food intake. Per2 (Brdm1) mice show reduced hepatic glycogen content and altered circadian expression during controlled fasting and refeeding. Livers from Per2 (Brdm1) mice display reduced glycogen synthase protein levels during refeeding, and increased glycogen phosphorylase activity during fasting. The latter is explained by PER2 action on the expression of the adapter proteins PTG and GL, which target the protein phosphatase-1 to glycogen to decrease glycogen phosphorylase activity. Finally, PER2 interacts with genomic regions of Gys2, PTG, and G L . These results indicate an important role for PER2 in the hepatic transcriptional response to feeding and acute fasting that promotes glucose storage to liver glycogen.

18.
Proc Natl Acad Sci U S A ; 108(42): E854-63, 2011 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-21949398

RESUMEN

Obesity is associated with a chronic low-grade inflammation, and specific antiinflammatory interventions may be beneficial for the treatment of type 2 diabetes and other obesity-related diseases. The lipid kinase PI3Kγ is a central proinflammatory signal transducer that plays a major role in leukocyte chemotaxis, mast cell degranulation, and endothelial cell activation. It was also reported that PI3Kγ activity within hematopoietic cells plays an important role in obesity-induced inflammation and insulin resistance. Here, we show that protection from insulin resistance, metabolic inflammation, and fatty liver in mice lacking functional PI3Kγ is largely consequent to their leaner phenotype. We also show that this phenotype is largely based on decreased fat gain, despite normal caloric intake, consequent to increased energy expenditure. Furthermore, our data show that PI3Kγ action on diet-induced obesity depends on PI3Kγ activity within a nonhematopoietic compartment, where it promotes energetic efficiency for fat mass gain. We also show that metabolic modulation by PI3Kγ depends on its lipid kinase activity and might involve kinase-independent signaling. Thus, PI3Kγ is an unexpected but promising drug target for the treatment of obesity and its complications.


Asunto(s)
Tejido Adiposo Blanco/enzimología , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Resistencia a la Insulina/fisiología , Obesidad/enzimología , Termogénesis/fisiología , Animales , Fosfatidilinositol 3-Quinasa Clase Ib/deficiencia , Fosfatidilinositol 3-Quinasa Clase Ib/genética , Dieta Alta en Grasa/efectos adversos , Hígado Graso/enzimología , Hígado Graso/etiología , Hígado Graso/prevención & control , Inflamación/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Obesidad/etiología , Obesidad/prevención & control , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Esterol Esterasa/metabolismo , Delgadez/enzimología
19.
Proc Natl Acad Sci U S A ; 105(43): 16809-13, 2008 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-18922779

RESUMEN

JNK is a stress-activated protein kinase that modulates pathways implicated in a variety of disease states. JNK-interacting protein-1 (JIP1) is a scaffolding protein that enhances JNK signaling by creating a proximity effect between JNK and upstream kinases. A minimal peptide region derived from JIP1 is able to inhibit JNK activity both in vitro and in cell. We report here a series of small molecules JIP1 mimics that function as substrate competitive inhibitors of JNK. One such compound, BI-78D3, dose-dependently inhibits the phosphorylation of JNK substrates both in vitro and in cell. In animal studies, BI-78D3 not only blocks JNK dependent Con A-induced liver damage but also restores insulin sensitivity in mouse models of type 2 diabetes. Our findings open the way for the development of protein kinase inhibitors targeting substrate specific docking sites, rather than the highly conserved ATP binding sites. In view of its favorable inhibition profile, selectivity, and ability to function in the cellular milieu and in vivo, BI-78D3 represents not only a JNK inhibitor, but also a promising stepping stone toward the development of an innovative class of therapeutics.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Dioxanos/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Tiazoles/farmacología , Animales , Unión Competitiva , Enfermedad Hepática Inducida por Sustancias y Drogas , Diabetes Mellitus Experimental/tratamiento farmacológico , Resistencia a la Insulina , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Hepatopatías/prevención & control , Ratones , Imitación Molecular , Fosforilación/efectos de los fármacos , Unión Proteica , Transducción de Señal
20.
Proc Natl Acad Sci U S A ; 103(33): 12602-6, 2006 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16891420

RESUMEN

Bcl-2 family proteins play a crucial role in tissue homeostasis and apoptosis (programmed cell death). Bid is a proapoptotic member of the Bcl-2 family, promoting cell death when activated by caspase-8. Following an NMR-based approach (structure-activity relationships by interligand NOE) we were able to identify two chemical fragments that bind on the surface of Bid. Covalent linkage of the two fragments led to high-affinity bidentate derivatives. In vitro and in-cell assays demonstrate that the compounds prevent tBid translocation to the mitochondrial membrane and the subsequent release of proapoptotic stimuli and inhibit neuronal apoptosis in the low micromolar range. Therefore, by using a rational chemical-biology approach, we derived antiapoptotic compounds that may have a therapeutic potential for disorders associated with Bid activation, e.g., neurodegenerative diseases, cerebral ischemia, or brain trauma.


Asunto(s)
Proteína Proapoptótica que Interacciona Mediante Dominios BH3 , Ligandos , Resonancia Magnética Nuclear Biomolecular/métodos , Animales , Apoptosis/fisiología , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/antagonistas & inhibidores , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/química , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Células Cultivadas , Ratones , Mitocondrias/metabolismo , Modelos Moleculares , Estructura Molecular , Neuronas/citología , Neuronas/metabolismo , Ratas , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA