Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros

Bases de datos
Tipo del documento
Asunto de la revista
Intervalo de año de publicación
1.
Nat Genet ; 36(9): 969-77, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15322543

RESUMEN

Tumor-necrosis factor (TNF), a pleiotropic cytokine, triggers physiological and pathological responses in several organs. Here we show that deletion of the mouse gene Timp3 resulted in an increase in TNF-alpha converting enzyme activity, constitutive release of TNF and activation of TNF signaling in the liver. The increase in TNF in Timp3(-/-) mice culminated in hepatic lymphocyte infiltration and necrosis, features that are also seen in chronic active hepatitis in humans. This pathology was prevented when deletion of Timp3 was combined with Tnfrsf1a deficiency. In a liver regeneration model that requires TNF signaling, Timp3(-/-) mice succumbed to liver failure. Hepatocytes from Timp3(-/-) mice completed the cell cycle but then underwent cell death owing to sustained activation of TNF. This hepatocyte cell death was completely rescued by a neutralizing antibody to TNF. Dysregulation of TNF occurred specifically in Timp3(-/-), and not Timp1(-/-) mice. These data indicate that TIMP3 is a crucial innate negative regulator of TNF in both tissue homeostasis and tissue response to injury.


Asunto(s)
Hepatitis Crónica/genética , Regeneración Hepática/genética , Proteínas/genética , Factor de Necrosis Tumoral alfa/genética , Proteínas ADAM , Proteína ADAM17 , Envejecimiento , Animales , Apoptosis , Ciclo Celular/genética , Hepatectomía , Hígado/enzimología , Hígado/patología , Metaloendopeptidasas/metabolismo , Ratones , Ratones Mutantes , Transducción de Señal , Inhibidores Tisulares de Metaloproteinasas , Factor de Necrosis Tumoral alfa/inmunología , Inhibidor Tisular de Metaloproteinasa-4
2.
Blood ; 116(22): 4474-82, 2010 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-20798233

RESUMEN

Regulating transition of hematopoietic stem cells (HSCs) between quiescent and cycling states is critical for maintaining homeostasis of blood cell production. The cycling states of HSCs are regulated by the extracellular factors such as cytokines and extracellular matrix; however, the molecular circuitry for such regulation remains elusive. Here we show that tissue inhibitor of metalloproteinase-3 (TIMP-3), an endogenous regulator of metalloproteinases, stimulates HSC proliferation by recruiting quiescent HSCs into the cell cycle. Myelosuppression induced TIMP-3 in the bone marrow before hematopoietic recovery. Interestingly, TIMP-3 enhanced proliferation of HSCs and promoted expansion of multipotent progenitors, which was achieved by stimulating cell-cycle entry of quiescent HSCs without compensating their long-term repopulating activity. Surprisingly, this effect did not require metalloproteinase inhibitory activity of TIMP-3 and was possibly mediated through a direct inhibition of angiopoietin-1 signaling, a critical mediator for HSC quiescence. Furthermore, bone marrow recovery from myelosuppression was accelerated by over-expression of TIMP-3, and in turn, impaired in TIMP-3-deficient animals. These results suggest that TIMP-3 may act as a molecular cue in response to myelosuppression for recruiting dormant HSCs into active cell cycle and may be clinically useful for facilitating hematopoietic recovery after chemotherapy or ex vivo expansion of HSCs.


Asunto(s)
Ciclo Celular , Células Madre Hematopoyéticas/citología , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Angiopoyetina 1/metabolismo , Animales , Médula Ósea/metabolismo , Médula Ósea/patología , Línea Celular , Proliferación Celular , Eliminación de Gen , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Leucopenia/inducido químicamente , Ratones , Ratones Endogámicos C57BL , Inhibidor Tisular de Metaloproteinasa-3/genética , Regulación hacia Arriba
3.
J Clin Invest ; 115(12): 3494-505, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16294222

RESUMEN

Activation of inflammatory pathways may contribute to the beginning and the progression of both atherosclerosis and type 2 diabetes. Here we report a novel interaction between insulin action and control of inflammation, resulting in glucose intolerance and vascular inflammation and amenable to therapeutic modulation. In insulin receptor heterozygous (Insr+/-) mice, we identified the deficiency of tissue inhibitor of metalloproteinase 3 (Timp3, an inhibitor of both TNF-alpha-converting enzyme [TACE] and MMPs) as a common bond between glucose intolerance and vascular inflammation. Among Insr+/- mice, those that develop diabetes have reduced Timp3 and increased TACE activity. Unchecked TACE activity causes an increase in levels of soluble TNF-alpha, which subsequently promotes diabetes and vascular inflammation. Double heterozygous Insr+/-Timp3+/- mice develop mild hyperglycemia and hyperinsulinemia at 3 months and overt glucose intolerance and hyperinsulinemia at 6 months. A therapeutic role for Timp3/TACE modulation is supported by the observation that pharmacological inhibition of TACE led to marked reduction of hyperglycemia and vascular inflammation in Insr+/- diabetic mice, as well as by the observation of increased insulin sensitivity in Tace+/- mice compared with WT mice. Our results suggest that an interplay between reduced insulin action and unchecked TACE activity promotes diabetes and vascular inflammation.


Asunto(s)
Inflamación/genética , Receptor de Insulina/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/genética , Inhibidor Tisular de Metaloproteinasa-3/fisiología , Factor de Necrosis Tumoral alfa/genética , Análisis de Varianza , Animales , Desoxiglucosa/metabolismo , Diabetes Mellitus/patología , Electroforesis en Gel de Poliacrilamida , Perfilación de la Expresión Génica , Vectores Genéticos , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Glucógeno/química , Heterocigoto , Homeostasis , Hiperglucemia/patología , Hiperinsulinismo/patología , Insulina/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Esquelético/patología , Músculos/patología , Fosforilación , Regiones Promotoras Genéticas , Unión Proteica , ARN/química , ARN/metabolismo , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Factores de Tiempo , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
4.
Circulation ; 110(16): 2401-9, 2004 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-15262835

RESUMEN

BACKGROUND: Despite the mounting clinical burden of heart failure, the biomolecules that control myocardial tissue remodeling are poorly understood. TIMP-3 is an endogenous inhibitor of matrix metalloproteinases (MMPs) that has been found to be deficient in failing human myocardium. We hypothesized that TIMP-3 expression prevents maladaptive tissue remodeling in the heart, and accordingly, its deficiency in mice would alone be sufficient to trigger progressive cardiac remodeling and dysfunction similar to human heart failure. METHODS AND RESULTS: Mice with a targeted timp-3 deficiency were evaluated with aging and compared with age-matched wild-type littermates. Loss of timp-3 function triggered spontaneous LV dilatation, cardiomyocyte hypertrophy, and contractile dysfunction at 21 months of age consistent with human dilated cardiomyopathy. Its absence also resulted in interstitial matrix disruption with elevated MMP-9 activity, and activation of the proinflammatory tumor necrosis factor-alpha cytokine system, molecular hallmarks of human myocardial remodeling. CONCLUSIONS: TIMP-3 deficiency disrupts matrix homeostasis and the balance of inflammatory mediators, eliciting the transition to cardiac dilation and dysfunction. Therapeutic restoration of myocardial TIMP-3 may provide a novel approach to limit cardiac remodeling and the progression to failure in patients with dilated cardiomyopathy.


Asunto(s)
Cardiomiopatía Dilatada/enzimología , Inhibidor Tisular de Metaloproteinasa-3/deficiencia , Proteínas ADAM , Proteína ADAM17 , Animales , Cardiomiopatía Dilatada/diagnóstico por imagen , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/patología , Colágeno/análisis , Progresión de la Enfermedad , Matriz Extracelular/química , Homeostasis , Hipertrofia , Macrófagos/patología , Metaloproteinasa 9 de la Matriz/análisis , Metaloendopeptidasas/análisis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Cardiovasculares , Contracción Miocárdica , Miocitos Cardíacos/patología , Nitritos/análisis , Receptores Tipo II del Factor de Necrosis Tumoral/análisis , Inhibidor Tisular de Metaloproteinasa-3/genética , Inhibidor Tisular de Metaloproteinasa-3/fisiología , Factor de Necrosis Tumoral alfa/análisis , Ultrasonografía
5.
J Clin Invest ; 120(8): 2731-44, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20628198

RESUMEN

The cell death receptor Fas plays a role in the establishment of fulminant hepatitis, a major cause of drug-induced liver failure. Fas activation elicits extrinsic apoptotic and hepatoprotective signals; however, the mechanisms by which these signals are integrated during disease are unknown. Tissue inhibitor of metalloproteinases 3 (TIMP3) controls the critical sheddase a disintegrin and metalloproteinase 17 (ADAM17) and may dictate stress signaling. Using mice and cells lacking TIMP3, ADAM17, and ADAM17-regulated cell surface molecules, we have found that ADAM17-mediated ectodomain shedding of TNF receptors and EGF family ligands controls activation of multiple signaling cascades in Fas-induced hepatitis. We demonstrated that TNF signaling promoted hepatotoxicity, while excessive TNF receptor 1 (TNFR1) shedding in Timp3-/- mice was protective. Compound Timp3-/-Tnf-/- and Timp3-/-Tnfr1-/- knockout conferred complete resistance to Fas-induced toxicity. Loss of Timp3 enhanced metalloproteinase-dependent EGFR signaling due to increased release of the EGFR ligands TGF-alpha, amphiregulin, and HB-EGF, while depletion of shed amphiregulin resensitized Timp3-/- hepatocytes to apoptosis. Finally, adenoviral delivery of Adam17 prevented acetaminophen-induced liver failure in a clinically relevant model of Fas-dependent fulminant hepatitis. These findings demonstrate that TIMP3 and ADAM17 cooperatively dictate cytokine signaling during death receptor activation and indicate that regulated metalloproteinase activity integrates survival and death signals during acute hepatotoxic stress.


Asunto(s)
Proteínas ADAM/fisiología , Apoptosis , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Receptores ErbB/fisiología , Hepatocitos/patología , Fallo Hepático Agudo/patología , Receptores Tipo I de Factores de Necrosis Tumoral/fisiología , Inhibidor Tisular de Metaloproteinasa-3/fisiología , Proteína ADAM17 , Animales , Células Cultivadas , Citoprotección , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Fosforilación , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-akt/fisiología , Transducción de Señal , Receptor fas/fisiología
6.
J Immunol ; 176(2): 721-5, 2006 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-16393953

RESUMEN

Host response to infectious agents must be rapid and powerful. One mechanism is the release of presynthesized membrane-bound TNF. TNF shedding is mediated by TNF-alpha converting enzyme, which is selectively inhibited by the tissue inhibitor of metalloproteinase 3 (TIMP3). We show that loss of TIMP3 impacts innate immunity by dysregulating cleavage of TNF and its receptors. Cultured timp3-/- macrophages release more TNF in response to LPS than wild-type macrophages. In timp3-/- mice, LPS causes serum levels of TNF and its receptors to rise more rapidly and remain higher compared with wild-type mice. The altered kinetics of ligand and receptor shedding enhances TNF signaling in timp3-/- mice, indicated by elevated serum IL-6. Physiologically, timp3-/- mice are more susceptible to LPS-induced mortality. Ablation of the TNF receptor gene p55 (Tnfrsf1a) or treatment with a synthetic metalloproteinase inhibitor rescues timp3-/- mice. Thus, TIMP3 is essential for normal innate immune function.


Asunto(s)
Inflamación/inmunología , Inhibidor Tisular de Metaloproteinasa-3/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Células Cultivadas , Inmunidad Innata , Cinética , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores del Factor de Necrosis Tumoral/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/deficiencia , Inhibidor Tisular de Metaloproteinasa-3/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA