Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 486(7404): 549-53, 2012 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-22722857

RESUMEN

Obesity and type-2 diabetes have increased markedly over the past few decades, in parallel. One of the major links between these two disorders is chronic, low-grade inflammation. Prolonged nutrient excess promotes the accumulation and activation of leukocytes in visceral adipose tissue (VAT) and ultimately other tissues, leading to metabolic abnormalities such as insulin resistance, type-2 diabetes and fatty-liver disease. Although invasion of VAT by pro-inflammatory macrophages is considered to be a key event driving adipose-tissue inflammation and insulin resistance, little is known about the roles of other immune system cell types in these processes. A unique population of VAT-resident regulatory T (Treg) cells was recently implicated in control of the inflammatory state of adipose tissue and, thereby, insulin sensitivity. Here we identify peroxisome proliferator-activated receptor (PPAR)-γ, the 'master regulator' of adipocyte differentiation, as a crucial molecular orchestrator of VAT Treg cell accumulation, phenotype and function. Unexpectedly, PPAR-γ expression by VAT Treg cells was necessary for complete restoration of insulin sensitivity in obese mice by the thiazolidinedione drug pioglitazone. These findings suggest a previously unknown cellular mechanism for this important class of thiazolidinedione drugs, and provide proof-of-principle that discrete populations of Treg cells with unique functions can be precisely targeted to therapeutic ends.


Asunto(s)
Tejido Adiposo/citología , PPAR gamma/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo , Tejido Adiposo/inmunología , Tejido Adiposo/patología , Animales , Diferenciación Celular , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Epidídimo/citología , Epidídimo/inmunología , Factores de Transcripción Forkhead/metabolismo , Expresión Génica , Hipoglucemiantes/farmacología , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Resistencia a la Insulina/fisiología , Recuento de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/metabolismo , Obesidad/patología , Fenotipo , Pioglitazona , ARN Mensajero/genética , ARN Mensajero/metabolismo , Linfocitos T Reguladores/efectos de los fármacos , Tiazolidinedionas/farmacología , Transcripción Genética
2.
Diabetes Obes Metab ; 19(10): 1458-1462, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28295931

RESUMEN

OBJECTIVE: To assess long-term efficacy and safety of salsalate to improve glycemia in persons with diabetes risk, who are overweight with statin-treated, stable coronary heart disease. METHODS: Glycemic status was assessed in 192 persons without diabetes at baseline in a pre-specified secondary analysis from Targeting INflammation Using SALsalate in CardioVascular Disease (TINSAL-CVD), a multi-center, double-masked, randomized (1:1), placebo-controlled, parallel clinical trial. RESULTS: Participants were mostly Caucasian males, age 60±7 years, BMI 31.4±3.0 kg/m2 , fasting glucose 92.8±11.0 mg/dL, and HbA1c 5.8±0.3%. Reductions in mean fasting glucose -5.70 mg/dL (95%CI: -7.44 to -3.97 mg/dL, P<0.001), HbA1c -0.11% (95%CI: -0.210 to -0.002%, P=0.046) and glycated serum protein -81.8 µg/mL (95%CI: -93.7 to -69.9 µg/mL, P<0.001) were demonstrated in salsalate compared to placebo-assigned groups over 30 months. Reductions in fasting glucose and glycated serum protein were greater with salsalate compared to placebo in participants with prediabetes compared to a normoglycemic sub-group (Pinteraction =0.018). Salsalate lowered total white blood cell counts (mean difference -0.7x103 /µL, 95%CI: -1.0 to -0.4 x103 /µL, P<0.001) and increased adiponectin (mean difference 1.8 µg/mL, 95%CI: 0.9 to 2.6 µg/mL, P<0.001) and albuminurea (16.7 µg/mg, 95%CI: 6.4 to 27.1 µg/mg, P<0.001) compared to placebo, consistent with previous results for patients with type 2 diabetes taking salsalate for shorter times. CONCLUSIONS: Salsalate improves glycemia in obese persons at increased risk for diabetes, and hence may decrease risk of incident type 2 diabetes. Salsalate may inform new therapeutic approaches for diabetes prevention, but renal safety may limit clinical utility.


Asunto(s)
Glucemia/efectos de los fármacos , Enfermedades Cardiovasculares/tratamiento farmacológico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Sobrepeso/sangre , Sobrepeso/tratamiento farmacológico , Estado Prediabético/sangre , Estado Prediabético/tratamiento farmacológico , Salicilatos/uso terapéutico , Adulto , Anciano , Glucemia/metabolismo , Enfermedades Cardiovasculares/sangre , Enfermedades Cardiovasculares/complicaciones , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Sobrepeso/complicaciones , Placebos , Estado Prediabético/complicaciones , Factores de Riesgo , Resultado del Tratamiento
3.
J Biol Chem ; 289(28): 19769-77, 2014 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-24867955

RESUMEN

Complementary surfaces are buried when peptide hormones, growth factors, or cytokines bind and activate cellular receptors. Although these extended surfaces provide high affinity and specificity to the interactions, they also present great challenges to the design of small molecules that might either mimic or antagonize the process. We show that the insulin receptor (IR) and downstream signals can be activated by targeting a site outside of its ligand-binding domain. A 24-residue peptide having the IR transmembrane (TM) domain sequence activates IR, but not related growth factor receptors, through specific interactions with the receptor TM domain. Like insulin-dependent activation, IR-TM requires that IR have a competent ATP-binding site and kinase activation loop. IR-TM also activates mutated receptors from patients with severe insulin resistance, which do not respond to insulin. These results show that IR can be activated through a pathway that bypasses its canonical ligand-binding domain.


Asunto(s)
Resistencia a la Insulina , Receptor de Insulina/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Humanos , Ratones , Mutación , Células 3T3 NIH , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptor de Insulina/genética
4.
Crit Care Med ; 42(7): e501-9, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24933061

RESUMEN

OBJECTIVE: Diaphragmatic weakness and acute respiratory failure are common in sepsis. Nuclear factor-κB acts as a general coordinator of the systemic inflammatory response, but its role within the diaphragm itself during sepsis is unknown. We investigated the potential protective effect upon the diaphragm of inhibiting nuclear factor-κB only within muscle fibers during acute endotoxemia. DESIGN: Prospective study in experimental animals. SETTING: University research laboratory. INTERVENTIONS: Wild-type and transgenic (muscle-specific IκBα super-repressor) mice with skeletal muscle-specific inhibition of the classical nuclear factor-κB pathway were subjected to acute endotoxemia. Muscle-specific ubiquitin ligases (muscle RING-finger protein 1 and atrogin-1), caspase-3 activity, inhibitor of apoptosis proteins, proinflammatory cytokines (interleukin-1ß, monocyte chemoattractant protein-1, and tumor necrosis factor-α), and diaphragmatic contractility were evaluated after 24 hours. MEASUREMENTS AND MAIN RESULTS: In wild-type mice, endotoxemia significantly increased proinflammatory cytokines (fold-change messenger RNA: interleukin-1ß = 7.6, monocyte chemoattractant protein-1 = 15.3, and tumor necrosis factor-α = 2.2) and proteolysis effectors (fold-change messenger RNA: muscle RING-finger protein 1 = 5.7, atrogin-1 = 2.8; caspase-3 activity elevated by 28%) in the diaphragm, while reducing its force-generating capacity by 38%. In nonendotoxemic muscle-specific IκBα super-repressor diaphragms, caspase-3 activity was unexpectedly increased by 40% above basal wild-type levels and inhibitors of apoptosis proteins were down-regulated, but force production remained normal. In muscle-specific IκBα super-repressor mice subjected to endotoxemia, proinflammatory cytokines, muscle RING-finger protein 1, and atrogin-1 were not significantly increased above their basal levels, and diaphragmatic weakness and further increases in caspase-3 activity were completely prevented. CONCLUSIONS: These results suggest that nuclear factor-κB signaling within skeletal muscle fibers is a key pathway leading to diaphragmatic weakness during acute endotoxemia, most likely via effects on multiple inflammatory mediators. In addition, inhibition of nuclear factor-κB signaling within diaphragm muscle fibers has complex effects on caspase-3 activation, which could have implications for the treatment of sepsis-induced diaphragmatic dysfunction.


Asunto(s)
Diafragma/metabolismo , Endotoxemia/fisiopatología , Inflamación/metabolismo , FN-kappa B/antagonistas & inhibidores , Animales , Apoptosis/fisiología , Caspasa 3/biosíntesis , Citocinas/biosíntesis , Ratones , Ratones Transgénicos , Contracción Muscular/fisiología , Fibras Musculares Esqueléticas , Ubiquitina-Proteína Ligasas/metabolismo
5.
Ann Intern Med ; 159(1): 1-12, 2013 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-23817699

RESUMEN

BACKGROUND: Short-duration studies show that salsalate improves glycemia in type 2 diabetes mellitus (T2DM). OBJECTIVE: To assess 1-year efficacy and safety of salsalate in T2DM. DESIGN: Placebo-controlled, parallel trial; computerized randomization and centralized allocation, with patients, providers, and researchers blinded to assignment. (ClinicalTrials.gov: NCT00799643). SETTING: 3 private practices and 18 academic centers in the United States. PATIENTS: Persons aged 18 to 75 years with fasting glucose levels of 12.5 mmol/L or less (≤225 mg/dL) and hemoglobin A1c (HbA1c) levels of 7.0% to 9.5% who were treated for diabetes. INTERVENTION: 286 participants were randomly assigned (between January 2009 and July 2011) to 48 weeks of placebo (n = 140) or salsalate, 3.5 g/d (n = 146), in addition to current therapies, and 283 participants were analyzed (placebo, n = 137; salsalate, n = 146). MEASUREMENTS: Change in hemoglobin A1c level (primary outcome) and safety and efficacy measures. RESULTS: The mean HbA1c level over 48 weeks was 0.37% lower in the salsalate group than in the placebo group (95% CI, -0.53% to -0.21%; P < 0.001). Glycemia improved despite more reductions in concomitant diabetes medications in salsalate recipients than in placebo recipients. Lower circulating leukocyte, neutrophil, and lymphocyte counts show the anti-inflammatory effects of salsalate. Adiponectin and hematocrit levels increased more and fasting glucose, uric acid, and triglyceride levels decreased with salsalate, but weight and low-density lipoprotein cholesterol levels also increased. Urinary albumin levels increased but reversed on discontinuation; estimated glomerular filtration rates were unchanged. LIMITATION: Trial duration and number of patients studied were insufficient to determine long-term risk-benefit of salsalate in T2DM. CONCLUSION: Salsalate improves glycemia in patients with T2DM and decreases inflammatory mediators. Continued evaluation of mixed cardiorenal signals is warranted.


Asunto(s)
Antiinflamatorios no Esteroideos/administración & dosificación , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Salicilatos/administración & dosificación , Adolescente , Adulto , Anciano , Antiinflamatorios no Esteroideos/efectos adversos , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/fisiopatología , Femenino , Hemoglobina Glucada/metabolismo , Humanos , Riñón/efectos de los fármacos , Riñón/fisiopatología , Masculino , Persona de Mediana Edad , Salicilatos/efectos adversos , Método Simple Ciego , Adulto Joven
6.
Arterioscler Thromb Vasc Biol ; 32(8): 1771-6, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22815343

RESUMEN

Obesity and its comorbidities, including type 2 diabetes mellitus and cardiovascular disease, are associated with a state of chronic low-grade inflammation that can be detected both systemically and within specific tissues. Areas of active investigation focus on the molecular bases of metabolic inflammation and potential pathogenic roles in insulin resistance, diabetes, and cardiovascular disease. An increased accumulation of macrophages occurring in obese adipose tissue has emerged as a key process in metabolic inflammation. Recent studies have also begun to unravel the heterogeneity of adipose tissue macrophages, and their physical and functional interactions with adipocytes, endothelial cells, and other immune cells within the adipose tissue microenvironment. Translating the information gathered from experimental models of insulin resistance and diabetes into meaningful therapeutic interventions is a tantalizing goal with long-term global health implications. In this context, ongoing clinical studies are testing the effects of targeting inflammation systemically on metabolic and cardiovascular outcomes.


Asunto(s)
Inflamación/complicaciones , Resistencia a la Insulina , Síndrome Metabólico/complicaciones , Tejido Adiposo Blanco/fisiología , Animales , Antiinflamatorios/uso terapéutico , Enfermedad Crónica , Humanos , Leucocitos/fisiología , Macrófagos/fisiología , Obesidad/complicaciones , Pérdida de Peso
7.
Proc Natl Acad Sci U S A ; 107(1): 240-5, 2010 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-20007767

RESUMEN

Lipodystrophy and obesity are opposites in terms of a deficiency versus excess of adipose tissue mass, yet these conditions are accompanied by similar metabolic consequences, including insulin resistance, dyslipidemia, hepatic steatosis, and increased risk for diabetes and atherosclerosis. Hepatic and myocellular steatosis likely contribute to metabolic dysregulation in both states. Inflammation and macrophage infiltration into adipose tissue also appear to participate in the pathogenesis of obesity-induced insulin resistance, but their contributions to lipodystrophy-induced insulin resistance have not been evaluated. We used aP2-nSREBP-1c transgenic (Tg) mice, an established model of lipodystrophy, to ask this question. Circulating cytokine elevations suggested systemic inflammation but even more dramatic was the number of infiltrating macrophages in all white and brown adipose tissue depots of the Tg mice; in contrast, there was no evidence of inflammatory infiltrates or responses in any other tissue including liver. Despite there being overt evidence of adipose tissue inflammation, antiinflammatory strategies including salicylate treatment and genetic suppression of myeloid NF-kappaB signaling that correct insulin resistance in obesity were ineffective in the lipodystrophic mice. We further showed that adipose tissue macrophages (ATMs) in lipodystrophy and obesity are very different in terms of activation state, gene expression patterns, and response to lipopolysaccharide. Although ATMs are even more abundant in lipodystrophy than in obesity, they have distinct phenotypes and likely roles in tissue remodeling, but do not appear to be involved in the pathogenesis of insulin resistance.


Asunto(s)
Tejido Adiposo/citología , Inflamación/inmunología , Lipodistrofia , Macrófagos/citología , Tejido Adiposo/inmunología , Tejido Adiposo/patología , Tejido Adiposo/trasplante , Animales , Apoptosis/fisiología , Modelos Animales de Enfermedad , Humanos , Resistencia a la Insulina/fisiología , Lipodistrofia/inmunología , Lipodistrofia/patología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Ratones Transgénicos , Obesidad/inmunología , Obesidad/patología , Fenotipo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Trasplante de Tejidos
8.
Am J Respir Cell Mol Biol ; 47(3): 288-97, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22538866

RESUMEN

Disease exacerbations and muscle wasting comprise negative prognostic factors of chronic obstructive pulmonary disease (COPD). Transient systemic inflammation and malnutrition have been implicated in skeletal muscle wasting after acute exacerbations of COPD. However, the interactions between systemic inflammation and malnutrition in their contributions to muscle atrophy, as well as the molecular basis underlying the transition of systemic inflammation to muscle atrophy, remain unresolved. Pulmonary inflammation was induced in mice by an intratracheal instillation of LPS to model acute disease exacerbation. Systemic inflammation, nutritional intake, and body and muscle weights were determined. Muscle inflammatory signaling and atrophy signaling were examined, and the effect of the muscle-specific inactivation of NF-κB on muscle atrophy was assessed in genetically modified mice. The intratracheal LPS instillation was followed by markedly elevated circulating cytokine concentrations and NF-κB activation in extrapulmonary tissues, including skeletal muscle. The administration of intratracheal LPS increased the expression of muscle E3 ubiquitin ligases, which govern muscle proteolysis, in particular MuRF1, and caused a rapid loss of muscle mass. Reduced food intake only partly accounted for the observed muscle atrophy, and did not activate NF-κB in muscle. Rather, plasma transfer experiments revealed the presence of NF-κB-signaling and atrophy-signaling properties in the circulation of intratracheal LPS-treated mice. The genetic inhibition of muscle NF-κB activity suppressed intratracheal LPS-induced MuRF1 expression and resulted in a significant sparing of muscle tissue. Systemic inflammation and malnutrition contribute to the muscle wasting induced by acute pulmonary inflammation via distinct mechanisms, and muscle NF-κB activation is required for the transition from inflammatory to muscle atrophy signaling.


Asunto(s)
Músculo Esquelético/patología , Atrofia Muscular , FN-kappa B/metabolismo , Neumonía/patología , Animales , Expresión Génica , Lipopolisacáridos/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Neumonía/metabolismo , Transducción de Señal
9.
J Biol Chem ; 286(20): 18130-8, 2011 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-21454505

RESUMEN

We have solved the x-ray crystal structures of the RabGAP domains of human TBC1D1 and human TBC1D4 (AS160), at 2.2 and 3.5 Å resolution, respectively. Like the yeast Gyp1p RabGAP domain, whose structure was solved previously in complex with mouse Rab33B, the human TBC1D1 and TBC1D4 domains both have 16 α-helices and no ß-sheet elements. We expected the yeast Gyp1p RabGAP/mouse Rab33B structure to predict the corresponding interfaces between cognate mammalian RabGAPs and Rabs, but found that residues were poorly conserved. We further tested the relevance of this model by Ala-scanning mutagenesis, but only one of five substitutions within the inferred binding site of the TBC1D1 RabGAP significantly perturbed catalytic efficiency. In addition, substitution of TBC1D1 residues with corresponding residues from Gyp1p did not enhance catalytic efficiency. We hypothesized that biologically relevant RabGAP/Rab partners utilize additional contacts not described in the yeast Gyp1p/mouse Rab33B structure, which we predicted using our two new human TBC1D1 and TBC1D4 structures. Ala substitution of TBC1D1 Met(930), corresponding to a residue outside of the Gyp1p/Rab33B contact, substantially reduced catalytic activity. GLUT4 translocation assays confirmed the biological relevance of our findings. Substitutions with lowest RabGAP activity, including catalytically dead RK and Met(930) and Leu(1019) predicted to perturb Rab binding, confirmed that biological activity requires contacts between cognate RabGAPs and Rabs beyond those in the yeast Gyp1p RabGAP/mouse Rab33B structure.


Asunto(s)
Proteínas Activadoras de GTPasa/química , Transportador de Glucosa de Tipo 4 , Modelos Moleculares , Animales , Cristalografía por Rayos X , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Humanos , Ratones , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Relación Estructura-Actividad , Proteínas de Unión al GTP rab/química , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo
10.
Am J Physiol Lung Cell Mol Physiol ; 302(1): L103-10, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22003096

RESUMEN

Loss of diaphragm muscle strength in inflammatory lung disease contributes to mortality and is associated with diaphragm fiber atrophy. Ubiquitin (Ub) 26S-proteasome system (UPS)-dependent protein breakdown, which mediates muscle atrophy in a number of physiological and pathological conditions, is elevated in diaphragm muscle of patients with chronic obstructive pulmonary disease. Nuclear factor kappa B (NF-κB), an essential regulator of many inflammatory processes, has been implicated in the regulation of poly-Ub conjugation of muscle proteins targeted for proteolysis by the UPS. Here, we test if NF-κB activation in diaphragm muscle and subsequent protein degradation by the UPS are required for pulmonary inflammation-induced diaphragm atrophy. Acute pulmonary inflammation was induced in mice by intratracheal lipopolysaccharide instillation. Fiber cross-sectional area, ex vivo tyrosine release, protein poly-Ub conjugation, and inflammatory signaling were determined in diaphragm muscle. The contribution of NF-κB or the UPS to diaphragm atrophy was assessed in mice with intact or genetically repressed NF-κB signaling or attenuated poly-Ub conjugation, respectively. Acute pulmonary inflammation resulted in diaphragm atrophy measured by reduced muscle fiber cross-sectional area. This was accompanied by diaphragm NF-κB activation, and proteolysis, measured by tyrosine release from the diaphragm. Poly-Ub conjugation was increased in diaphragm, as was the expression of muscle-specific E3 Ub ligases. Genetic suppression of poly-Ub conjugation prevented inflammation-induced diaphragm muscle atrophy, as did muscle-specific inhibition of NF-κB signaling. In conclusion, the present study is the first to demonstrate that diaphragm muscle atrophy, resulting from acute pulmonary inflammation, requires NF-κB activation and UPS-mediated protein degradation.


Asunto(s)
Diafragma/metabolismo , Atrofia Muscular/metabolismo , FN-kappa B/metabolismo , Poliubiquitina/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Animales , Diafragma/patología , Diafragma/fisiopatología , Modelos Animales de Enfermedad , Humanos , Inflamación/metabolismo , Inflamación/fisiopatología , Masculino , Ratones , Ratones Transgénicos , Atrofia Muscular/etiología , Atrofia Muscular/fisiopatología , Complejo de la Endopetidasa Proteasomal/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/complicaciones , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Transducción de Señal/fisiología
11.
Nat Med ; 11(2): 183-90, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15685173

RESUMEN

We show that NF-kappaB and transcriptional targets are activated in liver by obesity and high-fat diet (HFD). We have matched this state of chronic, subacute 'inflammation' by low-level activation of NF-kappaB in the liver of transgenic mice, designated LIKK, by selectively expressing constitutively active IKK-b in hepatocytes. These mice exhibit a type 2 diabetes phenotype, characterized by hyperglycemia, profound hepatic insulin resistance, and moderate systemic insulin resistance, including effects in muscle. The hepatic production of proinflammatory cytokines, including IL-6, IL-1beta and TNF-alpha, was increased in LIKK mice to a similar extent as induced by HFD in in wild-type mice. Parallel increases were observed in cytokine signaling in liver and mucscle of LIKK mice. Insulin resistance was improved by systemic neutralization of IL-6 or salicylate inhibition of IKK-beta. Hepatic expression of the IkappaBalpha superrepressor (LISR) reversed the phenotype of both LIKK mice and wild-type mice fed an HFD. These findings indicate that lipid accumulation in the liver leads to subacute hepatic 'inflammation' through NF-kappaB activation and downstream cytokine production. This causes insulin resistance both locally in liver and systemically.


Asunto(s)
Hepatocitos/fisiología , Resistencia a la Insulina/fisiología , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Antiinflamatorios no Esteroideos/metabolismo , Metabolismo de los Hidratos de Carbono , Citocinas/genética , Citocinas/metabolismo , Grasas de la Dieta/metabolismo , Femenino , Hepatocitos/patología , Humanos , Quinasa I-kappa B , Insulina/metabolismo , Interleucina-6/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , FN-kappa B/genética , Obesidad/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Ratas , Salicilatos/metabolismo , Transducción de Señal/fisiología
12.
Cell Death Differ ; 29(7): 1423-1432, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35017647

RESUMEN

Apoptotic cells are rapidly engulfed and removed by phagocytes after displaying cell surface eat-me signals. Among many phospholipids, only phosphatidylserine (PS) is known to act as an eat-me signal on apoptotic cells. Using unbiased proteomics, we identified externalized phosphatidylinositides (PIPs) as apoptotic eat-me signals recognized by CD14+ phagocytes. Exofacial PIPs on the surfaces of early and late-apoptotic cells were observed in patches and blebs using anti-PI(3,4,5)P3 antibody, AKT- and PLCδ PH-domains, and CD14 protein. Phagocytosis of apoptotic cells was blocked either by masking exofacial PIPs or by CD14 knockout in phagocytes. We further confirmed that exofacial PIP+ thymocytes increased dramatically after in vivo irradiation and that exofacial PIP+ cells represented more significant populations in tissues of Cd14-/- than WT mice, especially after induction of apoptosis. Our findings reveal exofacial PIPs to be previously unknown cell death signals recognized by CD14+ phagocytes.


Asunto(s)
Fagocitosis , Transducción de Señal , Animales , Apoptosis/fisiología , Ratones , Fagocitos/metabolismo , Fagocitosis/fisiología , Fosfatidilserinas/metabolismo , Transducción de Señal/fisiología
13.
Cell Metab ; 4(5): 337-8, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17084707

RESUMEN

In this issue of Cell Metabolism, Semenkovich and his colleagues show that ATM, a protein well known for its roles in the cellular response to DNA breaks, may also be linked to metabolic and cardiovascular diseases (Schneider et al., 2006). ATM seemingly does this by inhibiting JNK, a stress kinase involved in inflammation with related effects in insulin resistance and atherosclerosis. In an interesting twist, the authors show that chloroquine, an antimalarial drug, also activates ATM, which inhibits JNK, and improves insulin sensitivity and cardiovascular effects. These findings provide potential new insights into the pathogenesis and treatment of metabolic syndrome.


Asunto(s)
Proteínas de Ciclo Celular/efectos de los fármacos , Cloroquina/uso terapéutico , Proteínas de Unión al ADN/efectos de los fármacos , Síndrome Metabólico/tratamiento farmacológico , Ratones Noqueados/genética , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Supresoras de Tumor/efectos de los fármacos , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Aterosclerosis/tratamiento farmacológico , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Macrófagos/efectos de los fármacos , Ratones , Mutación , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo
14.
J Lipid Res ; 52(5): 942-50, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21357939

RESUMEN

Low-grade inflammation in different tissues, including activation of the nuclear factor κB pathway in liver, is involved in metabolic disorders such as type 2 diabetes and cardiovascular diseases (CVDs). In this study, we investigated the relation between chronic hepatocyte-specific overexpression of IkB kinase (IKK)-ß and hypertriglyceridemia, an important risk factor for CVD, by evaluating whether activation of IKK-ß only in the hepatocyte affects VLDL-triglyceride (TG) metabolism directly. Transgenic overexpression of constitutively active human IKK-ß specifically in hepatocytes of hyperlipidemic APOE*3-Leiden mice clearly induced hypertriglyceridemia. Mechanistic in vivo studies revealed that the hypertriglyceridemia was caused by increased hepatic VLDL-TG production rather than a change in plasma VLDL-TG clearance. Studies in primary hepatocytes showed that IKK-ß overexpression also enhances TG secretion in vitro, indicating a direct relation between IKK-ß activation and TG production within the hepatocyte. Hepatic lipid analysis and hepatic gene expression analysis of pathways involved in lipid metabolism suggested that hepatocyte-specific IKK-ß overexpression increases VLDL production not by increased steatosis or decreased FA oxidation, but most likely by carbohydrate-responsive element binding protein-mediated upregulation of Fas expression. These findings implicate that specific activation of inflammatory pathways exclusively within hepatocytes induces hypertriglyceridemia. Furthermore, we identify the hepatocytic IKK-ß pathway as a possible target to treat hypertriglyceridemia.


Asunto(s)
Apolipoproteína E3/metabolismo , Hepatocitos/metabolismo , Quinasa I-kappa B/metabolismo , Lipoproteínas VLDL/biosíntesis , Hígado/metabolismo , Triglicéridos/biosíntesis , Animales , Apolipoproteínas E/metabolismo , Western Blotting , Células Cultivadas , Humanos , Hipertrigliceridemia/genética , Hipertrigliceridemia/metabolismo , Quinasa I-kappa B/genética , Masculino , Ratones
15.
Am J Physiol Endocrinol Metab ; 301(6): E1099-107, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21862721

RESUMEN

Systemic inflammation is strongly involved in the pathophysiology of the metabolic syndrome, a cluster of metabolic risk factors that includes hypertriglyceridemia. Aspirin treatment lowers inflammation via inhibition of NF-κB activity but also reduces hypertriglyceridemia in humans. The aim of this study was to investigate the mechanism by which aspirin improves hypertriglyceridemia. Human apolipoprotein CI (apoCI)-expressing mice (APOC1 mice), an animal model with elevated plasma triglyceride (TG) levels, as well as normolipidemic wild-type (WT) mice were fed a high-fat diet (HFD) and treated with aspirin. Aspirin treatment reduced hepatic NF-κB activity in HFD-fed APOC1 and WT mice, and in addition, aspirin decreased plasma TG levels (-32%, P < 0.05) in hypertriglyceridemic APOC1 mice. This TG-lowering effect could not be explained by enhanced VLDL-TG clearance, but aspirin selectively reduced hepatic production of VLDL-TG in both APOC1 (-28%, P < 0.05) and WT mice (-33%, P < 0.05) without affecting VLDL-apoB production. Aspirin did not alter hepatic expression of genes involved in FA oxidation, lipogenesis, and VLDL production but decreased the incorporation of plasma-derived FA by the liver into VLDL-TG (-24%, P < 0.05), which was independent of hepatic expression of genes involved in FA uptake and transport. We conclude that aspirin improves hypertriglyceridemia by decreasing VLDL-TG production without affecting VLDL particle production. Therefore, the inhibition of inflammatory pathways by aspirin could be an interesting target for the treatment of hypertriglyceridemia.


Asunto(s)
Aspirina/farmacología , Dieta Alta en Grasa , Hipertrigliceridemia/prevención & control , Lipoproteínas VLDL/metabolismo , Triglicéridos/metabolismo , Animales , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/uso terapéutico , Apolipoproteína C-I/genética , Aspirina/uso terapéutico , Dieta Alta en Grasa/efectos adversos , Regulación hacia Abajo/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Hipertrigliceridemia/sangre , Hipertrigliceridemia/etiología , Hipertrigliceridemia/metabolismo , Lipoproteínas VLDL/sangre , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , FN-kappa B/metabolismo , Triglicéridos/sangre
16.
Clin Chem ; 57(2): 162-7, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21098138

RESUMEN

BACKGROUND: Chronic inflammation may participate in the pathogenesis of insulin resistance, type 2 diabetes, and cardiovascular disease and may be a common denominator that links obesity to these disease states. CONTENT: Epidemiologic studies have linked inflammatory biomarkers to incident diabetes and cardiovascular disease risk. Cellular and animal studies have provided support to the idea that inflammation mediates these disease processes, providing impetus to pharmacologically target these pathways for disease treatment and prevention. We review clinical strategies to target inflammation, with a focus on the antiinflammatory and antihyperglycemic effects of salicylates. SUMMARY: The evolving concept of diet-induced obesity driving insulin resistance, type 2 diabetes, and cardiovascular disease through immunologic processes provides new opportunities for the use of antiinflammatory strategies to correct the metabolic consequences of excess adiposity.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Tejido Adiposo/fisiopatología , Animales , Enfermedades Cardiovasculares/complicaciones , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/inmunología , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/inmunología , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Resistencia a la Insulina , Obesidad/complicaciones , Obesidad/tratamiento farmacológico , Obesidad/inmunología , Salicilatos/uso terapéutico
17.
Ann Intern Med ; 152(6): 346-57, 2010 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-20231565

RESUMEN

BACKGROUND: Salsalate, a nonacetylated prodrug of salicylate, has been shown to decrease blood glucose concentration in small studies. OBJECTIVE: To compare the efficacy and safety of salsalate at different doses in patients with type 2 diabetes. DESIGN: Parallel randomized trial with computer-generated randomization and centralized allocation. Patients and investigators, including those assessing outcomes and performing analyses, were masked to group assignment. (ClinicalTrials.gov registration number: NCT00392678) SETTING: 3 private practices and 14 universities in the United States. PATIENTS: Persons aged 18 to 75 years with fasting plasma glucose concentrations of 12.5 mmol/L or less (< or = 225 mg/dL) and hemoglobin A1c (HbA1c) levels of 7.0% to 9.5% treated by diet, exercise, and oral medication at stable doses for at least 8 weeks. INTERVENTION: After a 4-week, single-masked run-in period, patients were randomly assigned to receive placebo or salsalate in dosages of 3.0, 3.5, or 4.0 g/d for 14 weeks (27 patients each) in addition to their current therapy. MEASUREMENTS: Change in HbA1c was the primary outcome. Adverse effects and changes in measures of coronary risk and renal function were secondary outcomes. RESULTS: Higher proportions of patients in the 3 salsalate treatment groups experienced decreases in HbA1c levels of 0.5% or more from baseline (P = 0.009). Mean HbA1c changes were -0.36% (P = 0.02) at 3.0 g/d, -0.34% (P = 0.02) at 3.5 g/d, and -0.49% (P = 0.001) at 4.0 g/d compared with placebo. Other markers of glycemic control also improved in the 3 salsalate groups, as did circulating triglyceride and adiponectin concentrations. Mild hypoglycemia was more common with salsalate; documented events occurred only in patients taking sulfonylureas. Urine albumin concentrations increased in all salsalate groups compared with placebo. The drug was otherwise well tolerated. LIMITATION: The number of patients studied and the trial duration were insufficient to warrant recommending the use of salsalate for type 2 diabetes at this time. CONCLUSION: Salsalate lowers HbA1c levels and improves other markers of glycemic control in patients with type 2 diabetes and may therefore provide a new avenue for treatment. Renal and cardiac safety of the drug require further evaluation. PRIMARY FUNDING SOURCE: National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/administración & dosificación , Profármacos/administración & dosificación , Salicilatos/administración & dosificación , Adolescente , Adulto , Anciano , Albuminuria/inducido químicamente , Método Doble Ciego , Femenino , Enfermedades Gastrointestinales/inducido químicamente , Hemoglobina Glucada/metabolismo , Humanos , Hipoglucemiantes/efectos adversos , Lípidos/sangre , Masculino , Persona de Mediana Edad , Profármacos/efectos adversos , Salicilatos/efectos adversos , Acúfeno/inducido químicamente , Adulto Joven
18.
Proc Natl Acad Sci U S A ; 105(41): 15878-83, 2008 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-18840680

RESUMEN

Aire induces ectopic expression of peripheral tissue antigens (PTAs) in thymic medullary epithelial cells, which promotes immunological tolerance. Beginning with a broad screen of histone peptides, we demonstrate that the mechanism by which this single factor controls the transcription of thousands of genes involves recognition of the amino-terminal tail of histone H3, but not of other histones, by one of Aire's plant homeodomain (PHD) fingers. Certain posttranslational modifications of H3 tails, notably dimethylation or trimethylation at H3K4, abrogated binding by Aire, whereas others were tolerated. Similar PHD finger-H3 tail-binding properties were recently reported for BRAF-histone deacetylase complex 80 and DNA methyltransferase 3L; sequence alignment, molecular modeling, and biochemical analyses showed these factors and Aire to have structure-function relationships in common. In addition, certain PHD1 mutations underlying the polyendocrine disorder autoimmune polyendocrinopathy-candidiases-ectodermaldystrophy compromised Aire recognition of H3. In vitro binding assays demonstrated direct physical interaction between Aire and nucleosomes, which was in part buttressed by its affinity to DNA. In vivo Aire interactions with chromosomal regions depleted of H3K4me3 were dependent on its H3 tail-binding activity, and this binding was necessary but not sufficient for the up-regulation of genes encoding PTAs. Thus, Aire's activity as a histone-binding module mediates the thymic display of PTAs that promotes self-tolerance and prevents organ-specific autoimmunity.


Asunto(s)
Histonas/metabolismo , Tolerancia Inmunológica , Dominios y Motivos de Interacción de Proteínas/inmunología , Factores de Transcripción/metabolismo , Animales , Autoantígenos/genética , Autoinmunidad , Cromatina/metabolismo , Ratones , Especificidad de Órganos/inmunología , Unión Proteica , Factores de Transcripción/inmunología , Regulación hacia Arriba , Proteína AIRE
19.
J Biol Chem ; 284(50): 35165-76, 2009 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-19846556

RESUMEN

Hepatocyte nuclear factor 4alpha (HNF4alpha) is a novel nuclear receptor that participates in a hierarchical network of transcription factors regulating the development and physiology of such vital organs as the liver, pancreas, and kidney. Among the various transcriptional coregulators with which HNF4alpha interacts, peroxisome proliferation-activated receptor gamma (PPARgamma) coactivator 1alpha (PGC-1alpha) represents a novel coactivator whose activation is unusually robust and whose binding mode appears to be distinct from that of canonical coactivators such as NCoA/SRC/p160 family members. To elucidate the potentially unique molecular mechanism of PGC-1alpha recruitment, we have determined the crystal structure of HNF4alpha in complex with a fragment of PGC-1alpha containing all three of its LXXLL motifs. Despite the presence of all three LXXLL motifs available for interactions, only one is bound at the canonical binding site, with no additional contacts observed between the two proteins. However, a close inspection of the electron density map indicates that the bound LXXLL motif is not a selected one but an averaged structure of more than one LXXLL motif. Further biochemical and functional studies show that the individual LXXLL motifs can bind but drive only minimal transactivation. Only when more than one LXXLL motif is involved can significant transcriptional activity be measured, and full activation requires all three LXXLL motifs. These findings led us to propose a model wherein each LXXLL motif has an additive effect, and the multiple binding modes by HNF4alpha toward the LXXLL motifs of PGC-1alpha could account for the apparent robust activation by providing a flexible mechanism for combinatorial recruitment of additional coactivators and mediators.


Asunto(s)
Secuencia de Aminoácidos , Proteínas de Choque Térmico/metabolismo , Factor Nuclear 4 del Hepatocito/metabolismo , Factores de Transcripción/metabolismo , Animales , Activación Enzimática , Células HeLa , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/genética , Factor Nuclear 4 del Hepatocito/química , Factor Nuclear 4 del Hepatocito/genética , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Termodinámica , Factores de Transcripción/química , Factores de Transcripción/genética
20.
J Gerontol A Biol Sci Med Sci ; 75(4): 647-653, 2020 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-30423026

RESUMEN

Nuclear factor (NF)κB is a transcription factor that controls immune and inflammatory signaling pathways. In skeletal muscle, NFκB has been implicated in the regulation of metabolic processes and tissue mass, yet its affects on mitochondrial function in this tissue are unclear. To investigate the role of NFκB on mitochondrial function and its relationship with muscle mass across the life span, we study a mouse model with muscle-specific NFκB suppression (muscle-specific IκBα super-repressor [MISR] mice). In wild-type mice, there was a natural decline in muscle mass with aging that was accompanied by decreased mitochondrial function and mRNA expression of electron transport chain subunits. NFκB inactivation downregulated expression of PPARGC1A, and upregulated TFEB and PPARGC1B. NFκB inactivation also decreased gastrocnemius (but not soleus) muscle mass in early life (1-6 months old). Lower oxygen consumption rates occurred in gastrocnemius and soleus muscles from young MISR mice, whereas soleus (but not gastrocnemius) muscles from old MISR mice displayed increased oxygen consumption compared to age-matched controls. We conclude that the NFκB pathway plays an important role in muscle development and growth. The extent to which NFκB suppression alters mitochondrial function is age dependent and muscle specific. Finally, mitochondrial function and muscle mass are tightly associated in both genotypes and across the life span.


Asunto(s)
Mitocondrias Musculares/fisiología , Desarrollo de Músculos/fisiología , FN-kappa B/fisiología , Envejecimiento/genética , Envejecimiento/patología , Envejecimiento/fisiología , Animales , Citrato (si)-Sintasa/metabolismo , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias Musculares/genética , Desarrollo de Músculos/genética , Músculo Esquelético/fisiología , Músculo Esquelético/ultraestructura , FN-kappa B/antagonistas & inhibidores , Consumo de Oxígeno , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA