Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Genes Dev ; 26(18): 2009-14, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22987635

RESUMEN

Aging is associated with impaired scavenging of reactive oxygen species (ROS). Here, we show that TAp73, a p53 family member, protects against aging by regulating mitochondrial activity and preventing ROS accumulation. TAp73-null mice show more pronounced aging with increased oxidative damage and senescence. TAp73 deletion reduces cellular ATP levels, oxygen consumption, and mitochondrial complex IV activity, with increased ROS production and oxidative stress sensitivity. We show that the mitochondrial complex IV subunit cytochrome C oxidase subunit 4 (Cox4i1) is a direct TAp73 target and that Cox4i1 knockdown phenocopies the cellular senescence of TAp73-null cells. Results indicate that TAp73 affects mitochondrial respiration and ROS homeostasis, thus regulating aging.


Asunto(s)
Envejecimiento/genética , Envejecimiento/metabolismo , Mitocondrias/metabolismo , Consumo de Oxígeno , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Animales , Células Cultivadas , Complejo IV de Transporte de Electrones/metabolismo , Fibroblastos/metabolismo , Técnicas de Silenciamiento del Gen , Células HCT116 , Humanos , Ratones , Ratones Noqueados
2.
Carcinogenesis ; 39(9): 1095-1104, 2018 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-29982315

RESUMEN

New evidences indicate that the metabolic instruction of immunity (immune metabolism) results from the integration of cell metabolism and whole-body metabolism, which are both influenced by nutrition, microbiome metabolites and disease-driven metabolism (e.g. cancer metabolism). Cancer metabolism influences the immunological homeostasis and promotes immune alterations that support disease progression, hence influencing the clinical outcome. Cancer cells display increased glucose uptake and fermentation of glucose to lactate, even in the presence of completely functioning mitochondria. A major side effect of this event is immunosuppression, characterized by limited immunogenicity of cancer cells and restriction of the therapeutic efficacy of anticancer immunotherapy. Here, we discuss how the metabolism of myeloid cells associated with cancer contributes to the differentiation of their suppressive phenotype and therefore to cancer immune evasion.


Asunto(s)
Tolerancia Inmunológica/inmunología , Células Supresoras de Origen Mieloide/inmunología , Neoplasias/inmunología , Neoplasias/metabolismo , Escape del Tumor/inmunología , Glucosa/metabolismo , Humanos , Metabolismo de los Lípidos/fisiología , Microbiota/inmunología , Escape del Tumor/fisiología , Microambiente Tumoral/inmunología
3.
Arterioscler Thromb Vasc Biol ; 32(1): 74-81, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22015660

RESUMEN

OBJECTIVE: Tissue inhibitor of metalloproteinase 3 (TIMP3) is a stromal protein that inhibits the activity of proteases and receptors. TIMP3 is downregulated in metabolic and inflammatory disorders, such as type 2 diabetes mellitus and atherosclerosis, particularly in regions enriched with monocyte/macrophage cells. To investigate the role of TIMP3 in atherosclerosis, we generated a new mouse model in which Timp3 was overexpressed in the atherosclerotic plaque via a macrophage-specific promoter (MacT3). We elucidated any potential antiatherosclerotic effects of TIMP3, including regulation of monocyte/macrophage recruitment within atherosclerotic plaques, in MacT3 mice crossbred with low-density lipoprotein receptor knockout (LDLR(-/-)) mice. METHODS AND RESULTS: MacT3/LDLR(-/-) mice had an improvement of atherosclerosis and metabolic parameters compared with LDLR(-/-). En face aorta and aortic root examination of MacT3/LDLR(-/-) mice revealed smaller atherosclerotic plaques with features of stability, such as increased collagen content and decreased necrotic core formation. Atherosclerotic plaques in MacT3/LDLR(-/-) mice contained fewer T cells and macrophages. Furthermore, TIMP3 overexpression in macrophages resulted in reduced oxidative stress signals, as evidenced by lower lipid peroxidation, protein carbonylation, and nitration in atheromas. CONCLUSIONS: Our study confirmed that macrophage-specific overexpression of TIMP3 decreases the inflammatory content and the amplitude of atherosclerotic plaques in mice.


Asunto(s)
Aterosclerosis/prevención & control , Macrófagos/metabolismo , Receptores de LDL/deficiencia , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Animales , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Dieta Aterogénica/efectos adversos , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Regiones Promotoras Genéticas , Receptores de LDL/genética , Inhibidor Tisular de Metaloproteinasa-3/genética , Regulación hacia Arriba
4.
Mol Metab ; 69: 101686, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36746333

RESUMEN

OBJECTIVE: Obesity is associated with metabolic dysfunction of white adipose tissue (WAT). Activated adipocytes secrete pro-inflammatory cytokines resulting in the recruitment of pro-inflammatory macrophages, which contribute to WAT insulin resistance. The bile acid (BA)-activated nuclear Farnesoid X Receptor (FXR) controls systemic glucose and lipid metabolism. Here, we studied the role of FXR in adipose tissue function. METHODS: We first investigated the immune phenotype of epididymal WAT (eWAT) from high fat diet (HFD)-fed whole-body FXR-deficient (FXR-/-) mice by flow cytometry and gene expression analysis. We then generated adipocyte-specific FXR-deficient (Ad-FXR-/-) mice and analyzed systemic and eWAT metabolism and immune phenotype upon HFD feeding. Transcriptomic analysis was done on mature eWAT adipocytes from HFD-fed Ad-FXR-/- mice. RESULTS: eWAT from HFD-fed whole-body FXR-/- and Ad-FXR-/- mice displayed decreased pro-inflammatory macrophage infiltration and inflammation. Ad-FXR-/- mice showed lower blood glucose concentrations, improved systemic glucose tolerance and WAT insulin sensitivity and oxidative stress. Transcriptomic analysis identified Gsta4, a modulator of oxidative stress in WAT, as the most upregulated gene in Ad-FXR-/- mouse adipocytes. Finally, chromatin immunoprecipitation analysis showed that FXR binds the Gsta4 gene promoter. CONCLUSIONS: These results indicate a role for the adipocyte FXR-GSTA4 axis in controlling HFD-induced inflammation and systemic glucose homeostasis.


Asunto(s)
Resistencia a la Insulina , Animales , Ratones , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Glucosa/metabolismo , Homeostasis , Inflamación/metabolismo , Resistencia a la Insulina/fisiología , Estrés Oxidativo , Receptores Citoplasmáticos y Nucleares/metabolismo
5.
Mol Metab ; 59: 101454, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35150905

RESUMEN

OBJECTIVE: Metabolic syndrome, obesity, and steatosis are characterized by a range of dysregulations including defects in ubiquitin ligase tagging proteins for degradation. The identification of novel hepatic genes associated with fatty liver disease and metabolic dysregulation may be relevant to unravelling new mechanisms involved in liver disease progression METHODS: Through integrative analysis of liver transcriptomic and metabolomic obtained from obese subjects with steatosis, we identified itchy E ubiquitin protein ligase (ITCH) as a gene downregulated in human hepatic tissue in relation to steatosis grade. Wild-type or ITCH knockout mouse models of non-alcoholic fatty liver disease (NAFLD) and obesity-related hepatocellular carcinoma were analyzed to dissect the causal role of ITCH in steatosis RESULTS: We show that ITCH regulation of branched-chain amino acids (BCAAs) degradation enzymes is impaired in obese women with grade 3 compared with grade 0 steatosis, and that ITCH acts as a gatekeeper whose loss results in elevation of circulating BCAAs associated with hepatic steatosis. When ITCH expression was specifically restored in the liver of ITCH knockout mice, ACADSB mRNA and protein are restored, and BCAA levels are normalized both in liver and plasma CONCLUSIONS: Our data support a novel functional role for ITCH in the hepatic regulation of BCAA metabolism and suggest that targeting ITCH in a liver-specific manner might help delay the progression of metabolic hepatic diseases and insulin resistance.


Asunto(s)
Aminoácidos de Cadena Ramificada , Neoplasias Hepáticas , Enfermedad del Hígado Graso no Alcohólico , Obesidad , Ubiquitina-Proteína Ligasas , Aminoácidos de Cadena Ramificada/metabolismo , Animales , Regulación hacia Abajo , Femenino , Humanos , Ratones , Ratones Noqueados , Obesidad/complicaciones , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
6.
Front Immunol ; 10: 949, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31130949

RESUMEN

Myeloid-derived suppressor cells (MDSCs) represent a heterogeneous population of immature myeloid cells with major regulatory functions and rise during pathological conditions, including cancer, infections and autoimmune conditions. MDSC expansion is generally linked to inflammatory processes that emerge in response to stable immunological stress, which alter both magnitude and quality of the myelopoietic output. Inability to reinstate physiological myelopoiesis would fall in an "emergency state" that perpetually reprograms myeloid cells toward suppressive functions. While differentiation and reprogramming of myeloid cells toward an immunosuppressive phenotype can be considered the result of a multistep process that originates in the bone marrow and culminates in the tumor microenvironment, the identification of its driving events may offer potential therapeutic approaches in different pathologies. Indeed, whereas expansion of MDSCs, in both murine and human tumor bearers, results in reduced immune surveillance and antitumor cytotoxicity, placing an obstacle to the effectiveness of anticancer therapies, adoptive transfer of MDSCs has shown therapeutic benefits in autoimmune disorders. Here, we describe relevant mechanisms of myeloid cell reprogramming leading to generation of suppressive MDSCs and discuss their therapeutic ductility in disease.


Asunto(s)
Células Supresoras de Origen Mieloide/inmunología , Animales , Autoinmunidad , Humanos , Tolerancia Inmunológica , Neoplasias/inmunología
8.
Cell Rep ; 16(3): 731-43, 2016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27373162

RESUMEN

The effect of gut microbiota on obesity and insulin resistance is now recognized, but the underlying host-dependent mechanisms remain poorly undefined. We find that tissue inhibitor of metalloproteinase 3 knockout (Timp3(-/-)) mice fed a high-fat diet exhibit gut microbiota dysbiosis, an increase in branched chain and aromatic (BCAA) metabolites, liver steatosis, and an increase in circulating soluble IL-6 receptors (sIL6Rs). sIL6Rs can then activate inflammatory cells, such as CD11c(+) cells, which drive metabolic inflammation. Depleting the microbiota through antibiotic treatment significantly improves glucose tolerance, hepatic steatosis, and systemic inflammation, and neutralizing sIL6R signaling reduces inflammation, but only mildly impacts glucose tolerance. Collectively, our results suggest that gut microbiota is the primary driver of the observed metabolic dysfunction, which is mediated, in part, through IL-6 signaling. Our findings also identify an important role for Timp3 in mediating the effect of the microbiota in metabolic diseases.


Asunto(s)
Hígado Graso/metabolismo , Hígado Graso/patología , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/patología , Microbiota/fisiología , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Disbiosis/metabolismo , Disbiosis/patología , Hígado Graso/microbiología , Microbioma Gastrointestinal/fisiología , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/microbiología , Tracto Gastrointestinal/patología , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa/métodos , Inflamación/metabolismo , Inflamación/microbiología , Inflamación/patología , Resistencia a la Insulina/fisiología , Interleucina-6/metabolismo , Hígado/metabolismo , Hígado/microbiología , Hígado/patología , Enfermedades Metabólicas/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/metabolismo , Obesidad/patología , Receptores de Interleucina-6/metabolismo , Transducción de Señal/fisiología
9.
PLoS One ; 9(6): e99053, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24918924

RESUMEN

OBJECTIVE: To investigate the effects of TLR4 antagonism on human endothelial cells activation and cytokine expression, and whether the Asp299Gly TLR4 polymorphism is associated with better endothelial function in patients with rheumatoid arthritis (RA). METHODS: Human aortic endothelial cells (HAECs) were treated with lipopolysaccharide (LPS), OxPAPC, and free fatty acids (FFA) at baseline and after incubation with the TLR4 antagonist eritoran (E5564). Cytokine expression was assessed by quantitative real-time PCR. In vivo endothelial function was assessed as brachial artery flow-mediated dilation (FMD) in RA patients with the wild type gene (aa) and with the Asp299Gly TLR4 polymorphic variant (ag). RESULTS: In HAEC, TLR4 antagonism with eritoran inhibited LPS-induced mRNA expression of IL-6, IL-8, TNFα, CCL-2, VCAM and ICAM (P<0.05 for all) and inhibited Ox-PAPC-induced mRNA expression of IL-8 (P<0.05) and IL-6, albeit not to a statistically significant level (p = 0.07). In contrast, eritoran did not affect FFA-induced mRNA expression of IL-6 (P>0.05). In 30 patients with RA (15 with the ag allele) undergoing measurement of FMD, no differences in FMD and plasma levels of IL-6, IL-8, VCAM, and ICAM were found between the aa and the ag phenotype (P>0.05 for all). CONCLUSIONS: TLR4 signaling in endothelial cells may be triggered by LPS and oxidized phospholipids, leading to endothelial activation and inflammation, which are inhibited by eritoran. Our in vivo investigation, however, does not support an association between the Asp299Gly TLR4 polymorphism and improved endothelium-dependent vasodilator function in patients with RA. Further study is needed to better understand the potential role of TLR4 on endothelial dysfunction in this and other patient populations.


Asunto(s)
Artritis Reumatoide/fisiopatología , Endotelio Vascular/fisiología , FN-kappa B/metabolismo , Receptor Toll-Like 4/fisiología , Aorta/efectos de los fármacos , Aorta/fisiología , Células Cultivadas , Disacáridos/farmacología , Endotelio Vascular/efectos de los fármacos , Humanos , Lipopolisacáridos/antagonistas & inhibidores , Fosforilación , Fosfatos de Azúcar/farmacología
10.
Diabetes ; 63(2): 550-61, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24170694

RESUMEN

Classically activated macrophages (M1) secrete proinflammatory cytokine and are predominant in obese adipose tissue. M2 macrophages, prevalent in lean adipose tissue, are induced by IL-13 and IL-4, mainly secreted by Th2 lymphocytes, and produce the anti-inflammatory cytokine IL-10. ITCH is a ubiquitously expressed E3 ubiquitin ligase involved in T-cell differentiation and in a wide range of inflammatory pathways. ITCH downregulation in lymphocytes causes aberrant Th2 differentiation. To investigate the role of Th2/M2 polarization in obesity-related inflammation and insulin resistance, we compared wild-type and Itch(-/-) mice in a context of diet-induced obesity (high-fat diet [HFD]). When subjected to HFD, Itch(-/-) mice did not show an increase in body weight or insulin resistance; calorimetric analysis suggested an accelerated metabolism. The molecular analysis of metabolically active tissue revealed increased levels of M2 markers and genes involved in fatty acid oxidation. Histological examination of livers from Itch(-/-) mice suggested that ITCH deficiency protects mice from obesity-related nonalcoholic fatty liver disease. We also found a negative correlation between ITCH and M2 marker expression in human adipose tissues. Taken together, our data indicate that ITCH E3 ubiquitin ligase deficiency protects from the metabolic disorder caused by obesity.


Asunto(s)
Grasas de la Dieta/efectos adversos , Obesidad/etiología , Ubiquitina-Proteína Ligasas/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/fisiología , Animales , Biomarcadores/metabolismo , Regulación de la Expresión Génica/fisiología , Humanos , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Hígado/metabolismo , Macrófagos Peritoneales/fisiología , Receptor de Manosa , Lectinas de Unión a Manosa/genética , Lectinas de Unión a Manosa/metabolismo , Ratones , Ratones Noqueados , Obesidad/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Ubiquitina-Proteína Ligasas/genética
11.
Diabetes ; 63(6): 2086-96, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24430438

RESUMEN

Obesity elicits immune cell infiltration of adipose tissue provoking chronic low-grade inflammation. Regulatory T cells (Tregs) are specifically reduced in adipose tissue of obese animals. Since interleukin (IL)-21 plays an important role in inducing and maintaining immune-mediated chronic inflammatory processes and negatively regulates Treg differentiation/activity, we hypothesized that it could play a role in obesity-induced insulin resistance. We found IL-21 and IL-21R mRNA expression upregulated in adipose tissue of high-fat diet (HFD) wild-type (WT) mice and in stromal vascular fraction from human obese subjects in parallel to macrophage and inflammatory markers. Interestingly, a larger infiltration of Treg cells was seen in the adipose tissue of IL-21 knockout (KO) mice compared with WT animals fed both normal diet and HFD. In a context of diet-induced obesity, IL-21 KO mice, compared with WT animals, exhibited lower body weight, improved insulin sensitivity, and decreased adipose and hepatic inflammation. This metabolic phenotype is accompanied by a higher induction of interferon regulatory factor 4 (IRF4), a transcriptional regulator of fasting lipolysis in adipose tissue. Our data suggest that IL-21 exerts negative regulation on IRF4 and Treg activity, developing and maintaining adipose tissue inflammation in the obesity state.


Asunto(s)
Tejido Adiposo/metabolismo , Inflamación/metabolismo , Resistencia a la Insulina/inmunología , Factores Reguladores del Interferón/metabolismo , Interleucinas/metabolismo , Lipólisis , Obesidad/metabolismo , Células 3T3-L1/metabolismo , Animales , Western Blotting , Células Cultivadas , Citometría de Flujo , Inflamación/inmunología , Interleucinas/genética , Lipólisis/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/inmunología , ARN Mensajero/metabolismo , Receptores de Interleucina-21/metabolismo , Linfocitos T Reguladores , Regulación hacia Arriba
12.
PLoS One ; 8(1): e54458, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23372727

RESUMEN

BACKGROUND: Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide. Prevention and risk reduction are important and the identification of specific biomarkers for early diagnosis of HCC represents an active field of research. Increasing evidence indicates that fat accumulation in the liver, defined as hepatosteatosis, is an independent and strong risk factor for developing an HCC. MacroH2A1, a histone protein generally associated with the repressed regions of chromosomes, is involved in hepatic lipid metabolism and is present in two alternative spliced isoforms, macroH2A1.1 and macroH2A1.2. These isoforms have been shown to predict lung and colon cancer recurrence but to our knowledge, their role in fatty-liver associated HCC has not been investigated previously. METHODS: We examined macroH2A1.1 and macroH2A1.2 protein expression levels in the liver of two murine models of fat-associated HCC, the high fat diet/diethylnistrosamine (DEN) and the phosphatase and tensin homolog (PTEN) liver specific knock-out (KO) mouse, and in human liver samples of subjects with steatosis or HCC, using immunoblotting and immunohistochemistry. RESULTS: Protein levels for both macroH2A1 isoforms were massively upregulated in HCC, whereas macroH2A1.2 was specifically upregulated in steatosis. In addition, examination of human liver samples showed a significant difference (p<0.01) in number of positive nuclei in HCC (100% of tumor cells positive for either macroH2A1.1 or macroH2A1.2), when compared to steatosis (<2% of hepatocytes positive for either isoform). The steatotic areas flanking the tumors were highly immunopositive for macroH2A1.1 and macroH2A1.2. CONCLUSIONS: These data obtained in mice and humans suggest that both macroH2A1 isoforms may play a role in HCC pathogenesis and moreover may be considered as novel diagnostic markers for human HCC.


Asunto(s)
Carcinoma Hepatocelular/genética , Hígado Graso/genética , Histonas/genética , Neoplasias Hepáticas/genética , Animales , Biomarcadores/metabolismo , Carcinoma Hepatocelular/etiología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/patología , Dieta Alta en Grasa , Dietilnitrosamina , Hígado Graso/inducido químicamente , Hígado Graso/complicaciones , Hígado Graso/metabolismo , Regulación de la Expresión Génica , Hepatocitos/metabolismo , Hepatocitos/patología , Histonas/metabolismo , Humanos , Inmunohistoquímica , Neoplasias Hepáticas/etiología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Noqueados , Fosfohidrolasa PTEN/deficiencia , Fosfohidrolasa PTEN/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Microambiente Tumoral/genética
13.
Diabetes ; 61(2): 454-62, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22228717

RESUMEN

The tissue inhibitor of metalloproteinase (TIMP)3, a stromal protein that restrains the activity of proteases and receptors, is reduced in inflammatory metabolic disorders such as type 2 diabetes mellitus (T2DM) and atherosclerosis. We overexpressed Timp3 in mouse macrophages (MacT3) to analyze its potential antidiabetic and antiatherosclerotic effects. Transgenic mice with myeloid cells targeting overexpression of TIMP3 were generated and fed a high-fat diet for 20 weeks. Physical and metabolic phenotypes were determined. Inflammatory markers, lipid accumulation, and insulin sensitivity were measured in white adipose tissue (WAT), liver, and skeletal muscle. In a model of insulin resistance, MacT3 mice were more glucose tolerant and insulin sensitive than wild-type mice in both in vitro and in vivo tests. Molecular and biochemical analyses revealed that increased expression of TIMP3 restrained metabolic inflammation and stress-related pathways, including Jun NH2-terminal kinase and p38 kinase activation, in WAT and liver. TIMP3 overexpression in macrophages resulted in reduced activation of oxidative stress signals related to lipid peroxidation, protein carbonylation, and nitration in WAT and liver. Our data show that macrophage-specific overexpression of TIMP3 protects from metabolic inflammation and related metabolic disorders such as insulin resistance, glucose intolerance, and nonalcoholic steatohepatitis.


Asunto(s)
Tejido Adiposo/patología , Hígado Graso/prevención & control , Resistencia a la Insulina , Macrófagos/fisiología , Inhibidor Tisular de Metaloproteinasa-3/fisiología , Adiposidad , Animales , Antígenos CD/análisis , Antígenos de Diferenciación Mielomonocítica/análisis , Dieta Alta en Grasa , Fluorodesoxiglucosa F18 , Inflamación/prevención & control , Ratones , Enfermedad del Hígado Graso no Alcohólico , Obesidad/metabolismo , ARN Mensajero/análisis , Inhibidor Tisular de Metaloproteinasa-3/genética
15.
Diabetes ; 58(10): 2396-401, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19581416

RESUMEN

OBJECTIVE: Atherosclerosis is accelerated in subjects with type 2 diabetes by unknown mechanisms. We identified tissue inhibitor of metalloproteinase 3 (TIMP3), the endogenous inhibitor of A disintegrin and metalloprotease domain 17 (ADAM17) and other matrix metalloproteinases (MMPs), as a gene modifier for insulin resistance and vascular inflammation in mice. We tested its association with atherosclerosis in subjects with type 2 diabetes and identified Sirtuin 1 (SirT1) as a major regulator of TIMP3 expression. RESEARCH DESIGN AND METHODS: We investigated ADAM10, ADAM17, MMP9, TIMP1, TIMP2, TIMP3, and TIMP4 expression levels in human carotid atherosclerotic plaques (n = 60) from subjects with and without diabetes. Human vascular smooth muscle cells exposed to several metabolic stimuli were used to identify regulators of TIMP3 expression. SirT1 small interference RNA, cDNA, and TIMP3 promoter gene reporter were used to study SirT1-dependent regulation of TIMP3. RESULTS: Here, we show that in human carotid atherosclerotic plaques, TIMP3 was significantly reduced in subjects with type 2 diabetes, leading to ADAM17 and MMP9 overactivity. Reduced expression of TIMP3 was associated in vivo with SirT1 levels. In smooth muscle cells, inhibition of SirT1 activity and levels reduced TIMP3 expression, whereas SirT1 overexpression increased TIMP3 promoter activity. CONCLUSIONS: In atherosclerotic plaques from subjects with type 2 diabetes, the deregulation of ADAM17 and MMP9 activities is related to inadequate expression of TIMP3 via SirT1. Studies in vascular cells confirmed the role of SirT1 in tuning TIMP3 expression.


Asunto(s)
Aterosclerosis/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Sirtuinas/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAM17 , Animales , Aterosclerosis/patología , Arterias Carótidas/metabolismo , Enfermedades de las Arterias Carótidas/metabolismo , Humanos , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Valores de Referencia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sirtuina 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA