Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 37(6): 1157-1167, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28408371

RESUMEN

OBJECTIVE: Extracellular nicotinamide phosphoribosyltransferase (eNAMPT) mediates inflammatory and potentially proatherogenic effects, whereas the role of intracellular NAMPT (iNAMPT), the rate limiting enzyme in the salvage pathway of nicotinamide adenine dinucleotide (NAD)+ generation, in atherogenesis is largely unknown. Here we investigated the effects of iNAMPT overexpression in leukocytes on inflammation and atherosclerosis. APPROACH AND RESULTS: Low-density lipoprotein receptor-deficient mice with hematopoietic overexpression of human iNAMPT (iNAMPThi), on a western type diet, showed attenuated plaque burden with features of lesion stabilization. This anti-atherogenic effect was caused by improved resistance of macrophages to apoptosis by attenuated chemokine (C-C motif) receptor 2-dependent monocyte chemotaxis and by skewing macrophage polarization toward an anti-inflammatory M2 phenotype. The iNAMPThi phenotype was almost fully reversed by treatment with the NAMPT inhibitor FK866, indicating that iNAMPT catalytic activity is instrumental in the atheroprotection. Importantly, iNAMPT overexpression did not induce any increase in eNAMPT, and eNAMPT had no effect on chemokine (C-C motif) receptor 2 expression and promoted an inflammatory M1 phenotype in macrophages. The iNAMPT-mediated effects at least partly involved sirtuin 1-dependent molecular crosstalk of NAMPT and peroxisome proliferator-activated receptor γ. Finally, iNAMPT and peroxisome proliferator-activated receptor γ showed a strong correlation in human atherosclerotic, but not healthy arteries, hinting to a relevance of iNAMPT/peroxisome proliferator-activated receptor γ pathway also in human carotid atherosclerosis. CONCLUSIONS: This study highlights the functional dichotomy of intracellular versus extracellular NAMPT, and unveils a critical role for the iNAMPT-peroxisome proliferator-activated receptor γ axis in atherosclerosis.


Asunto(s)
Aterosclerosis/prevención & control , Diferenciación Celular , Citocinas/metabolismo , Leucocitos/enzimología , Macrófagos/metabolismo , Monocitos/metabolismo , Nicotinamida Fosforribosiltransferasa/metabolismo , PPAR gamma/metabolismo , Anciano , Animales , Apoptosis , Aterosclerosis/enzimología , Aterosclerosis/genética , Aterosclerosis/patología , Células Cultivadas , Citocinas/antagonistas & inhibidores , Citocinas/genética , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Predisposición Genética a la Enfermedad , Humanos , Leucocitos/efectos de los fármacos , Leucocitos/patología , Activación de Macrófagos , Macrófagos/efectos de los fármacos , Macrófagos/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Monocitos/efectos de los fármacos , Monocitos/patología , Nicotinamida Fosforribosiltransferasa/antagonistas & inhibidores , Nicotinamida Fosforribosiltransferasa/genética , Fenotipo , Receptores de LDL/deficiencia , Receptores de LDL/genética , Transducción de Señal , Sirtuina 1/metabolismo , Factores de Tiempo , Regulación hacia Arriba
2.
Circulation ; 132(6): 490-501, 2015 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-26068045

RESUMEN

BACKGROUND: Leukocyte migration is critical for the infiltration of monocytes and accumulation of monocyte-derived macrophages in inflammation. Considering that Hck and Fgr are instrumental in this process, their impact on atherosclerosis and on lesion inflammation and stability was evaluated. METHODS AND RESULTS: Hematopoietic Hck/Fgr-deficient, LDLr(-/-) chimeras, obtained by bone marrow transplantation, had smaller but, paradoxically, less stable lesions with reduced macrophage content, overt cap thinning, and necrotic core expansion as the most prominent features. Despite a Ly6C(high)-skewed proinflammatory monocyte phenotype, Hck/Fgr deficiency led to disrupted adhesion of myeloid cells to and transmigration across endothelial monolayers in vitro and atherosclerotic plaques in vivo, as assessed by intravital microscopy, flow cytometry, and histological examination of atherosclerotic arteries. Moreover, Hck/Fgr-deficient macrophages showed blunted podosome formation and mesenchymal migration capacity. In consequence, transmigrated double-knockout macrophages were seen to accumulate in the fibrous cap, potentially promoting its focal erosion, as observed for double-knockout chimeras. CONCLUSIONS: The hematopoietic deficiency of Hck and Fgr led to attenuated atherosclerotic plaque formation by abrogating endothelial adhesion and transmigration; paradoxically, it also promoted plaque instability by causing monocyte subset imbalance and subendothelial accumulation, raising a note of caution regarding src kinase-targeted intervention in plaque inflammation.


Asunto(s)
Quimiotaxis de Leucocito/fisiología , Macrófagos Peritoneales/patología , Monocitos/patología , Placa Aterosclerótica/patología , Proteínas Proto-Oncogénicas c-hck/deficiencia , Proteínas Proto-Oncogénicas/deficiencia , Familia-src Quinasas/deficiencia , Animales , Apoptosis , Adhesión Celular , Extensiones de la Superficie Celular/ultraestructura , Células Cultivadas , Células Endoteliales , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Rodamiento de Leucocito , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitosis , Placa Aterosclerótica/enzimología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-hck/genética , Proteínas Proto-Oncogénicas c-hck/fisiología , Quimera por Radiación , Receptores de LDL/deficiencia , Receptores de LDL/genética , Receptores de LDL/fisiología , Migración Transendotelial y Transepitelial , Familia-src Quinasas/genética , Familia-src Quinasas/fisiología
3.
Arterioscler Thromb Vasc Biol ; 35(1): 79-86, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25395616

RESUMEN

OBJECTIVE: The protein degrading activity of cathepsin C (CatC), combined with its role in leukocyte granule activation, suggests a contribution of this cystein protease in atherosclerosis. However, no experimental data are available to validate this concept. APPROACH AND RESULTS: CatC gene and protein expression were increased in ruptured versus advanced stable human carotid artery lesions. To assess causal involvement of CatC in plaque progression and stability, we generated LDLr(-/-)//CatC(-/-) chimeras by bone marrow transplantation. CatC(-/-) chimeras presented attenuated plaque burden in carotids, descending aorta, aortic arch and root, at both the early and advanced plaque stage. CatC was abundantly expressed by plaque macrophages and foam cells. CatC expression and activity were dramatically downregulated in plaques of CatC(-/-) chimeras, supporting a hematopoietic origin of plaque CatC. Our studies unveiled an unexpected feedback of CatC deficiency on macrophage activation programs and T helper cell differentiation in as much as that CatC expression was upregulated in M1 macrophages, whereas its deficiency led to combined M2 (in vitro) and Th2 polarization (in vivo). CONCLUSIONS: Our data implicate CatC has a role in the selective tuning of innate and adaptive immune responses, relevant to a chronic immune disease, such as atherosclerosis.


Asunto(s)
Inmunidad Adaptativa , Aorta/enzimología , Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , Arterias Carótidas/enzimología , Enfermedades de las Arterias Carótidas/prevención & control , Catepsina C/metabolismo , Inmunidad Innata , Leucocitos/enzimología , Animales , Aorta/inmunología , Aorta/patología , Enfermedades de la Aorta/enzimología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/inmunología , Enfermedades de la Aorta/patología , Aterosclerosis/enzimología , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/patología , Arterias Carótidas/inmunología , Arterias Carótidas/patología , Enfermedades de las Arterias Carótidas/enzimología , Enfermedades de las Arterias Carótidas/genética , Enfermedades de las Arterias Carótidas/inmunología , Enfermedades de las Arterias Carótidas/patología , Catepsina C/genética , Diferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación hacia Abajo , Femenino , Células Espumosas/enzimología , Células Espumosas/inmunología , Humanos , Leucocitos/inmunología , Activación de Macrófagos , Ratones Endogámicos C57BL , Ratones Noqueados , Placa Aterosclerótica , Receptores de LDL/deficiencia , Receptores de LDL/genética , Linfocitos T Colaboradores-Inductores/enzimología , Linfocitos T Colaboradores-Inductores/inmunología , Factores de Tiempo
4.
J Mol Cell Cardiol ; 74: 44-52, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24816217

RESUMEN

AIMS: The SDF-1α/CXCR4 dyad was previously shown by us and others to be instrumental in intimal hyperplasia as well as early stage atherosclerosis. We here sought to investigate its impact on clinically relevant stages of atherosclerosis in mouse and man. METHODS AND RESULTS: Immunohistochemical analysis of CXCR4 expression in human atherosclerotic lesions revealed a progressive accumulation of CXCR4(+) cells during plaque progression. To address causal involvement of CXCR4 in advanced stages of atherosclerosis we reconstituted LDLr(-/-) mice with autologous bone marrow infected with lentivirus encoding SDF-1α antagonist or CXCR4 degrakine, which effects proteasomal degradation of CXCR4. Functional CXCR4 blockade led to progressive plaque expansion with disease progression, while also promoting intraplaque haemorrhage. Moreover, CXCR4 knockdown was seen to augment endothelial adhesion of neutrophils. Concordant with this finding, inhibition of CXCR4 function increased adhesive capacity and reduced apoptosis of neutrophils and resulted in hyperactivation of circulating neutrophils. Compatible with a role of the neutrophil CXCR4 in end-stage atherosclerosis, CXCR4 expression by circulating neutrophils was lowered in patients with acute cardiovascular syndromes. CONCLUSION: In conclusion, CXCR4 contributes to later stages of plaque progression by perturbing neutrophil function.


Asunto(s)
Aterosclerosis/genética , Hemorragia/genética , Neutrófilos/metabolismo , Placa Aterosclerótica/genética , Receptores CXCR4/genética , Animales , Aterosclerosis/metabolismo , Aterosclerosis/patología , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Adhesión Celular , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Progresión de la Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Regulación de la Expresión Génica , Vectores Genéticos , Hemorragia/metabolismo , Hemorragia/patología , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Ratones , Ratones Noqueados , Neutrófilos/patología , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/metabolismo , Receptores de LDL/deficiencia , Receptores de LDL/genética , Transducción de Señal
5.
J Biol Chem ; 285(35): 27449-27456, 2010 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-20566642

RESUMEN

Pathological forms of left ventricular hypertrophy (LVH) often progress to heart failure. Specific transcription factors have been identified that activate the gene program to induce pathological forms of LVH. It is likely that apart from activating transcriptional inducers of LVH, constitutive transcriptional repressors need to be removed during the development of cardiac hypertrophy. Here, we report that the constitutive presence of Krüppel-like factor 15 (KLF15) is lost in pathological hypertrophy and that this loss precedes progression toward heart failure. We show that transforming growth factor-beta-mediated activation of p38 MAPK is necessary and sufficient to decrease KLF15 expression. We further show that KLF15 robustly inhibits myocardin, a potent transcriptional activator. Loss of KLF15 during pathological LVH relieves the inhibitory effects on myocardin and stimulates the expression of serum response factor target genes, such as atrial natriuretic factor. This uncovers a novel mechanism where activated p38 MAPK decreases KLF15, an important constitutive transcriptional repressor whose removal seems a vital step to allow the induction of pathological LVH.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Hipertrofia Ventricular Izquierda/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Miocardio/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Animales , Factor Natriurético Atrial/metabolismo , Células COS , Chlorocebus aethiops , Activación Enzimática , Ratones , Ratas , Ratas Endogámicas Lew , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
Circ Res ; 104(2): 170-8, 6p following 178, 2009 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-19096030

RESUMEN

The myocardium of the failing heart undergoes a number of structural alterations, most notably hypertrophy of cardiac myocytes and an increase in extracellular matrix proteins, often seen as primary fibrosis. Connective tissue growth factor (CTGF) is a key molecule in the process of fibrosis and therefore seems an attractive therapeutic target. Regulation of CTGF expression at the promoter level has been studied extensively, but it is unknown how CTGF transcripts are regulated at the posttranscriptional level. Here we provide several lines of evidence to show that CTGF is importantly regulated by 2 major cardiac microRNAs (miRNAs), miR-133 and miR-30. First, the expression of both miRNAs was inversely related to the amount of CTGF in 2 rodent models of heart disease and in human pathological left ventricular hypertrophy. Second, in cultured cardiomyocytes and fibroblasts, knockdown of these miRNAs increased CTGF levels. Third, overexpression of miR-133 or miR-30c decreased CTGF levels, which was accompanied by decreased production of collagens. Fourth, we show that CTGF is a direct target of these miRNAs, because they directly interact with the 3' untranslated region of CTGF. Taken together, our results indicate that miR-133 and miR-30 importantly limit the production of CTGF. We also provide evidence that the decrease of these 2 miRNAs in pathological left ventricular hypertrophy allows CTGF levels to increase, which contributes to collagen synthesis. In conclusion, our results show that both miR-133 and miR-30 directly downregulate CTGF, a key profibrotic protein, and thereby establish an important role for these miRNAs in the control of structural changes in the extracellular matrix of the myocardium.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Matriz Extracelular/metabolismo , Insuficiencia Cardíaca/metabolismo , Hipertrofia Ventricular Izquierda/metabolismo , MicroARNs/metabolismo , Miocardio/metabolismo , Procesamiento Postranscripcional del ARN , Remodelación Ventricular , Regiones no Traducidas 3' , Animales , Animales Recién Nacidos , Secuencia de Bases , Células Cultivadas , Biología Computacional , Factor de Crecimiento del Tejido Conjuntivo/genética , Modelos Animales de Enfermedad , Femenino , Fibrosis , Técnicas de Silenciamiento del Gen , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Humanos , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Miocardio/patología , Filogenia , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Renina/genética , Renina/metabolismo , Regulación hacia Arriba , Remodelación Ventricular/genética
7.
Circulation ; 115(4): 475-82, 2007 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-17242279

RESUMEN

BACKGROUND: The cell-associated proteoglycan syndecan-1 (Synd1) closely regulates inflammation and cell-matrix interactions during wound healing and tumorigenesis. The present study investigated whether Synd1 may also regulate cardiac inflammation, matrix remodeling, and function after myocardial infarction (MI). METHODS AND RESULTS: First, we showed increased protein and mRNA expression of Synd1 from 24 hours on, reaching its maximum at 7 days after MI and declining thereafter. Targeted deletion of Synd1 resulted in increased inflammation and accelerated, yet functionally adverse, infarct healing after MI. In concordance, adenoviral gene expression of Synd1 protected against exaggerated inflammation after MI, mainly by reducing transendothelial adhesion and migration of leukocytes, as shown in vitro. Increased inflammation in the absence of Synd1 resulted in increased monocyte chemoattractant protein-1 expression, increased activity of matrix metalloproteinase-2 and -9, and decreased activity of tissue transglutaminase, associated with increased collagen fragmentation and disorganization. Exaggerated inflammation and adverse matrix remodeling in the absence of Synd1 increased cardiac dilatation and impaired systolic function, whereas gene overexpression of Synd1 reduced inflammation and protected against cardiac dilatation and failure. CONCLUSIONS: Increased expression of Synd1 in the infarct protects against exaggerated inflammation and adverse infarct healing, thereby reducing cardiac dilatation and dysfunction after MI in mice.


Asunto(s)
Cardiomiopatía Dilatada/genética , Infarto del Miocardio/genética , Sindecano-1/genética , Sindecano-1/fisiología , Remodelación Ventricular/genética , Animales , Cardiomiopatía Dilatada/inmunología , Cardiomiopatía Dilatada/fisiopatología , Colágeno/genética , Citocinas/genética , Citocinas/inmunología , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Corazón/fisiología , Leucocitos/inmunología , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Mutagénesis , Infarto del Miocardio/inmunología , Infarto del Miocardio/fisiopatología , Miocardio/enzimología , Miocardio/patología , ARN Mensajero/metabolismo , Sístole/genética , Remodelación Ventricular/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...