Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Biochim Biophys Acta ; 1853(8): 1879-88, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25960397

RESUMEN

Phosphatidylinositol 3-kinaseß (PI3Kß) plays a predominant role in integrin outside-in signaling and in platelet activation by GPVI engagement. We have shown that the tyrosine kinase Pyk2 mediates PI3Kß activation downstream of integrin αIIbß3, and promotes the phosphorylation of the PI3K-associated adaptor protein c-Cbl. In this study, we compared the functional correlation between Pyk2 and PI3Kß upon recruitment of the two main platelet collagen receptors, integrin α2ß1 and GPVI. PI3Kß-mediated phosphorylation of Akt was inhibited in Pyk2-deficient platelets adherent to monomeric collagen through integrin α2ß1, but occurred normally upon GPVI ligation. Integrin α2ß1 engagement led to Pyk2-independent association of c-Cbl with PI3K. However, c-Cbl was not phosphorylated in adherent platelets, and phosphorylation of Akt occurred normally in c-Cbl-deficient platelets, indicating that the c-Cbl is dispensable for Pyk2-mediated PI3Kß activation. Stimulation of platelets with CRP, a selective GPVI ligand, induced c-Cbl phosphorylation in the absence of Pyk2, but failed to promote its association with PI3K. Pyk2 activation was completely abrogated in PI3KßKD, but not in PI3KγKD platelets, and was strongly inhibited by Src kinases and phospholipase C inhibitors, and by BAPTA-AM. The absence of PI3Kß activity also hampered GPVI-induced tyrosine-phosphorylation and activation of PLCγ2, preventing intracellular Ca2+ increase and phosphorylation of pleckstrin. Moreover, GPVI-induced intracellular Ca2+ increase and pleckstrin phosphorylation were also strongly inhibited in human platelets treated with the PI3Kß inhibitor TGX-221. These results outline important differences in the regulation of PI3Kß by GPVI and integrin α2ß1 and suggest that inhibition of Pyk2 may target PI3Kß activation in a selective context of platelet stimulation.


Asunto(s)
Quinasa 2 de Adhesión Focal/fisiología , Integrina alfa2beta1/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Glicoproteínas de Membrana Plaquetaria/fisiología , Proteínas Proto-Oncogénicas c-cbl/fisiología , Animales , Células Cultivadas , Activación Enzimática , Humanos , Ratones , Ratones Noqueados , Transducción de Señal
2.
Biochem J ; 469(2): 199-210, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25967238

RESUMEN

In blood platelets, stimulation of G protein-coupled receptors (GPCRs) by thrombin triggers the activation of Src family kinases (SFKs), resulting in the tyrosine-phosphorylation of multiple substrates, but the mechanism underlying this process is still poorly understood. In the present study, we show that the time-dependent protein-tyrosine phosphorylation triggered by thrombin in human or murine platelets was totally suppressed only upon concomitant chelation of intracellular Ca(2+) and inhibition of SFKs. Thrombin-induced activation of SFKs was regulated by intracellular Ca(2+) and accordingly the Ca(2+) ionophore A23187 was sufficient to stimulate SFKs. A23187 also triggered the phosphorylation and activation of the Ca(2+)-dependent focal adhesion kinase Pyk2 and Pyk2 activation by thrombin was Ca(2+)-dependent. Stimulation of SFKs by thrombin or A23187 was strongly reduced in platelets from Pyk2 knockout (KO) mice, as was the overall pattern of protein-tyrosine phosphorylation. By immunoprecipitation experiments, we demonstrate that Lyn and Fyn, but not Src, were activated by Pyk2. Inhibition of SFKs by PP2 also reduced the phosphorylation of Pyk2 in thrombin or A23187-stimulated platelets. Analysis of KO mice demonstrated that Fyn, but not Lyn, was required for complete Pyk2 phosphorylation by thrombin. Finally, PP2 reduced aggregation of murine platelets to a level comparable to that of Pyk2-deficient platelets, but did not have further effects in the absence of Pyk2. These results indicate that in thrombin-stimulated platelets, stimulation of Pyk2 by intracellular Ca(2+) initiates SFK activation, establishing a positive loop that reinforces the Pyk2/SFK axis and allows the subsequent massive tyrosine phosphorylation of multiple substrates required for platelet aggregation.


Asunto(s)
Plaquetas/enzimología , Señalización del Calcio/efectos de los fármacos , Quinasa 2 de Adhesión Focal/metabolismo , Hemostáticos/farmacología , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Trombina/farmacología , Familia-src Quinasas/metabolismo , Animales , Plaquetas/citología , Señalización del Calcio/fisiología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Quinasa 2 de Adhesión Focal/genética , Humanos , Ratones , Ratones Noqueados , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Activación Plaquetaria/efectos de los fármacos , Activación Plaquetaria/genética , Proteínas Proto-Oncogénicas c-fyn/genética , Familia-src Quinasas/genética
3.
Blood ; 121(4): 648-57, 2013 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-23175689

RESUMEN

In the present study, we used a knockout murine model to analyze the contribution of the Ca(2+)-dependent focal adhesion kinase Pyk2 in platelet activation and thrombus formation in vivo. We found that Pyk2-knockout mice had a tail bleeding time that was slightly increased compared with their wild-type littermates. Moreover, in an in vivo model of femoral artery thrombosis, the time to arterial occlusion was significantly prolonged in mice lacking Pyk2. Pyk2-deficient mice were also significantly protected from collagen plus epinephrine-induced pulmonary thromboembolism. Ex vivo aggregation of Pyk2-deficient platelets was normal on stimulation of glycoprotein VI, but was significantly reduced in response to PAR4-activating peptide, low doses of thrombin, or U46619. Defective platelet aggregation was accompanied by impaired inside-out activation of integrin α(IIb)ß(3) and fibrinogen binding. Granule secretion was only slightly reduced in the absence of Pyk2, whereas a marked inhibition of thrombin-induced thromboxane A(2) production was observed, which was found to be responsible for the defective aggregation. Moreover, we have demonstrated that Pyk2 is implicated in the signaling pathway for cPLA(2) phosphorylation through p38 MAPK. The results of the present study show the importance of the focal adhesion kinase Pyk2 downstream of G-protein-coupled receptors in supporting platelet aggregation and thrombus formation.


Asunto(s)
Quinasa 2 de Adhesión Focal/genética , Activación Plaquetaria/genética , Trombina/metabolismo , Trombosis/genética , Trombosis/metabolismo , Animales , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Calcio/metabolismo , Fosfolipasas A2 Grupo II/metabolismo , Ratones , Ratones Noqueados , Fosforilación , Activación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/genética , Transducción de Señal , Trombina/farmacología , Tromboxano A2/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
4.
J Biol Chem ; 288(25): 18194-203, 2013 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-23640884

RESUMEN

Proline-rich tyrosine kinase 2 (Pyk2) is activated by various agonists in platelets. We evaluated the signaling mechanism and the functional role of Pyk2 in platelets by using pharmacological inhibitors and Pyk2-deficient platelets. We found that platelet aggregation and secretion in response to 2-methylthio-ADP (2-MeSADP) and AYPGKF were diminished in the presence of Pyk2 inhibitors or in Pyk2-deficient platelets, suggesting that Pyk2 plays a positive regulatory role in platelet functional responses. It has been shown that ADP-, but not thrombin-induced thromboxane (TxA2) generation depends on integrin signaling. Unlike ADP, thrombin activates G12/13 pathways, and G12/13 pathways can substitute for integrin signaling for TxA2 generation. We found that Pyk2 was activated downstream of both G12/13 and integrin-mediated pathways, and both 2-MeSADP- and AYPGKF-induced TxA2 generation was significantly diminished in Pyk2-deficient platelets. In addition, TxA2 generation induced by co-stimulation of Gi and Gz pathways, which is dependent on integrin signaling, was inhibited by blocking Pyk2. Furthermore, inhibition of 2-MeSADP-induced TxA2 generation by fibrinogen receptor antagonist was not rescued by co-stimulation of G12/13 pathways in the presence of Pyk2 inhibitor. We conclude that Pyk2 is a common signaling effector downstream of both G12/13 and integrin αIIbß3 signaling, which contributes to thromboxane generation.


Asunto(s)
Plaquetas/metabolismo , Quinasa 2 de Adhesión Focal/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Tromboxano A2/biosíntesis , Adenosina Difosfato/análogos & derivados , Adenosina Difosfato/farmacología , Animales , Plaquetas/efectos de los fármacos , Western Blotting , Células Cultivadas , Relación Dosis-Respuesta a Droga , Quinasa 2 de Adhesión Focal/antagonistas & inhibidores , Quinasa 2 de Adhesión Focal/genética , Humanos , Ratones , Ratones Noqueados , Oligopéptidos/farmacología , Fosforilación/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Tionucleótidos/farmacología , Factores de Tiempo , Tirfostinos/farmacología
5.
Clin Immunol ; 153(1): 64-72, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24721319

RESUMEN

Kawasaki disease (KD) is a paediatric idiopathic vasculitis. In this study, on the basis of studies using an established animal model for KD, we report that mannose-binding lectin (MBL) is involved in the pathogenesis of the disease. KD-like experimental murine vasculitis was induced by intraperitoneally administering a Candida albicans water-soluble extract (CAWS). MBL-A gradually increased in the serum of the model mice treated with CAWS. Deposition of MBL-A and MBL-C was observed in the aortic root, including the coronary arteries, which is a predilection site in experimental vasculitis. Corresponding to the distribution patterns of MBLs, marked deposition of C3/C3-derived peptides was also observed. Regarding the self-reactivity of MBLs, we observed that MBLs interacted with core histones to activate the lectin pathway. These results suggest that some types of pathogens provoke the MBL-dependent complement pathway (lectin pathway) to cause and/or exacerbate KD-like vasculitis.


Asunto(s)
Lectina de Unión a Manosa/metabolismo , Síndrome Mucocutáneo Linfonodular/inmunología , Síndrome Mucocutáneo Linfonodular/metabolismo , Animales , Antígenos Fúngicos/inmunología , Aorta/metabolismo , Aorta/patología , Candida albicans/inmunología , Activación de Complemento/inmunología , Lectina de Unión a Manosa de la Vía del Complemento/inmunología , Modelos Animales de Enfermedad , Histonas/metabolismo , Masculino , Ratones , Transducción de Señal
6.
Blood ; 119(3): 847-56, 2012 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-22106345

RESUMEN

Integrin α2ß1-mediated adhesion of human platelets to monomeric type I collagen or to the GFOGER peptide caused a time-dependent activation of PI3K and Akt phosphorylation. This process was abrogated by pharmacologic inhibition of PI3Kß, but not of PI3Kγ or PI3Kα. Moreover, Akt phosphorylation was undetectable in murine platelets expressing a kinase-dead mutant of PI3Kß (PI3Kß(KD)), but occurred normally in PI3Kγ(KD) platelets. Integrin α2ß1 failed to stimulate PI3Kß in platelets from phospholipase Cγ2 (PLCγ2)-knockout mice, and we found that intracellular Ca(2+) linked PLCγ2 to PI3Kß activation. Integrin α2ß1 also caused a time-dependent stimulation of the focal kinase Pyk2 downstream of PLCγ2 and intracellular Ca(2+). Whereas activation of Pyk2 occurred normally in PI3Kß(KD) platelets, stimulation of PI3Kß was strongly reduced in Pyk2-knockout mice. Neither Pyk2 nor PI3Kß was required for α2ß1-mediated adhesion and spreading. However, activation of Rap1b and inside-out stimulation of integrin αIIbß3 were reduced after inhibition of PI3Kß and were significantly impaired in Pyk2-deficient platelets. Finally, both PI3Kß and Pyk2 significantly contributed to thrombus formation under flow. These results demonstrate that Pyk2 regulates PI3Kß downstream of integrin α2ß1, and document a novel role for Pyk2 and PI3Kß in integrin α2ß1 promoted inside-out activation of integrin αIIbß3 and thrombus formation.


Asunto(s)
Plaquetas/metabolismo , Quinasa 2 de Adhesión Focal/fisiología , Integrina alfa2beta1/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Adhesividad Plaquetaria , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Calcio/metabolismo , Colágeno/metabolismo , Fibrinógeno/metabolismo , Humanos , Immunoblotting , Ratones , Ratones Noqueados , Fosforilación , Agregación Plaquetaria , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Transducción de Señal
7.
Proc Natl Acad Sci U S A ; 108(23): 9472-7, 2011 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-21593423

RESUMEN

Endothelial and endothelial progenitor cells (ECs and EPCs) play a fundamental role in angiogenesis that is essential for numerous physiological and pathological processes. The phosphatase and tensin homolog (PTEN)/ phosphoinositide 3-kinase (PI3K) pathway has been implicated in angiogenesis, but the mechanism in the regulation of this pathway in ECs and EPCs is poorly understood. Here we show that ARIA (apoptosis regulator through modulating IAP expression), a transmembrane protein that we recently identified, regulates the PTEN/PI3K pathway in ECs and EPCs and controls developmental and postnatal angiogenesis in vivo. We found that ARIA is abundantly expressed in EPCs and regulates their angiogenic functions by modulating PI3K/Akt/endothelial nitric oxide synthase (eNOS) signaling. Genetic deletion of ARIA caused nonfatal bleeding during embryogenesis, in association with increased small vessel density and altered expression of various vascular growth factors including angiopoietins and VEGF receptors. Postnatal neovascularization induced by critical limb ischemia was substantially enhanced in ARIA-null mice, in conjunction with more bone marrow (BM)-derived ECs detected in ischemic muscles. Administration of PI3K or NO synthase inhibitor completely abolished the enhanced neovascularization in ARIA(-/-) mice. Mechanistically, we identified that ARIA interacts with PTEN at the intracellular domain independently of the PTEN phosphorylation in its C-terminal tail. Overexpressed ARIA increased PTEN in the membrane fraction, whereas ARIA-silencing reduced the membrane-associated PTEN, resulting in modified PI3K/Akt signaling. Taken together, our findings establish a previously undescribed mode of regulation of the PTEN/PI3K/Akt pathway by ARIA, and reveal a unique mechanism in the control of angiogenesis. These functions of ARIA might offer a unique therapeutic potential.


Asunto(s)
Células Endoteliales/metabolismo , Neurregulina-1/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células Madre/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Vasos Sanguíneos/embriología , Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/metabolismo , Células CHO , Línea Celular , Membrana Celular/metabolismo , Células Cultivadas , Cricetinae , Cricetulus , Células Endoteliales/citología , Humanos , Immunoblotting , Inmunoprecipitación , Ratones , Ratones Noqueados , Mutación , Neurregulina-1/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Unión Proteica , Interferencia de ARN , Transducción de Señal , Células Madre/citología
8.
Am J Nephrol ; 37(4): 302-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23548793

RESUMEN

BACKGROUND/AIMS: Minimal-change nephrotic syndrome (MCNS) is a kidney disease defined by selective proteinuria and hypoalbuminemia occurring in the absence of cellular glomerular infiltrates or immunoglobulin deposits. Recent observations suggest that nuclear factor κB (NF-κB) of podocyte is strongly associated with the development of proteinuria in MCNS. Dehydroxymethylepoxyquinomicin (DHMEQ) is a novel NF-κB inhibitor that potently inhibits DNA-binding activity of NF-κB, resulting in several therapeutic effects in various pathological conditions. We conducted this study to ask whether DHMEQ may ameliorate the nephrosis in mice induced by puromycin aminonucleoside (PAN), which is considered to be an animal model for MCNS. METHODS/RESULTS: Pretreatment with DHMEQ alleviated the proteinuria and reversed the serum abnormalities in mice nephrosis induced by 450 mg/kg of PAN. Increased serum interleukin-6 level in PAN-induced nephrosis was also completely suppressed by DHMEQ. Electron microscopic analyses of glo-meruli indicated that DHMEQ can inhibit the podocyte foot process effacement via blocking the translocation of podocyte NF-κB from cytoplasm to nucleus. CONCLUSIONS: These results suggest that DHMEQ can be a potential therapeutic agent for MCNS.


Asunto(s)
Benzamidas/administración & dosificación , Ciclohexanonas/administración & dosificación , FN-kappa B/antagonistas & inhibidores , Nefrosis/prevención & control , Puromicina Aminonucleósido/toxicidad , Adenosina Desaminasa/metabolismo , Albuminuria/orina , Animales , Proteínas Sanguíneas/análisis , Colesterol/sangre , Glicerolfosfato Deshidrogenasa/metabolismo , Interleucina-6/sangre , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Riñón/patología , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Nefrosis/inducido químicamente , Nefrosis/metabolismo , Nefrosis/patología , Proteinuria/orina , Ratas , Albúmina Sérica/análisis
9.
Proc Natl Acad Sci U S A ; 107(45): 19308-13, 2010 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-20966350

RESUMEN

Klotho is a circulating protein, and Klotho deficiency disturbs endothelial integrity, but the molecular mechanism is not fully clarified. We report that vascular endothelium in Klotho-deficient mice showed hyperpermeability with increased apoptosis and down-regulation of vascular endothelial (VE)-cadherin because of an increase in VEGF-mediated internal calcium concentration ([Ca(2+)]i) influx and hyperactivation of Ca(2+)-dependent proteases. Immunohistochemical analysis, the pull-down assay using Klotho-fixed agarose, and FRET confocal imaging confirmed that Klotho protein binds directly to VEGF receptor 2 (VEGFR-2) and endothelial, transient-receptor potential canonical Ca(2+) channel 1 (TRPC-1) and strengthens the association to promote their cointernalization. An in vitro mutagenesis study revealed that the second hydrolase domain of Klotho interacts with sixth and seventh Ig domains of VEGFR-2 and the third extracellular loop of TRPC-1. In Klotho-deficient endothelial cells, VEGF-mediated internalization of the VEGFR-2/TRPC-1 complex was impaired, and surface TRPC-1 expression increased 2.2-fold; these effects were reversed by supplementation of Klotho protein. VEGF-mediated elevation of [Ca(2+)]i was sustained at higher levels in an extracellular Ca(2+)-dependent manner, and normalization of TRCP-1 expression restored the abnormal [Ca(2+)]i handling. These findings provide evidence that Klotho protein is associated with VEGFR-2/TRPC-1 in causing cointernalization, thus regulating TRPC-1-mediated Ca(2+) entry to maintain endothelial integrity.


Asunto(s)
Glucuronidasa/metabolismo , Canales Catiónicos TRPC/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Sitios de Unión , Calcio/metabolismo , Canales de Calcio , Glucuronidasa/deficiencia , Proteínas Klotho , Ratones , Unión Proteica
10.
J Mol Cell Cardiol ; 52(1): 175-84, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22044588

RESUMEN

Inhibition of tumor suppressor p53 is cardioprotective against ischemic injury and provides resistance to subsequent cardiac remodeling. We investigated p53-mediated expansion of ischemic damage with a focus on mitochondrial integrity in association with autophagy and apoptosis. p53(-/-) heart showed that autophagic flux was promoted under ischemia without a change in cardiac tissue ATP content. Electron micrographs revealed that ischemic border zone in p53(-/-) mice had 5-fold greater numbers of autophagic vacuoles containing mitochondria, indicating the occurrence of mitophagy, with an apparent reduction of abnormal mitochondria compared with those in WT mice. Analysis of autophagic mediators acting downstream of p53 revealed that TIGAR (TP53-induced glycolysis and apoptosis regulator) was exclusively up-regulated in ischemic myocardium. TIGAR(-/-) mice exhibited the promotion of mitophagy followed by decrease of abnormal mitochondria and resistance to ischemic injury, consistent with the phenotype of p53(-/-) mice. In p53(-/-) and TIGAR(-/-) ischemic myocardium, ROS production was elevated and followed by Bnip3 activation which is an initiator of mitophagy. Furthermore, the activation of Bnip3 and mitophagy due to p53/TIGAR inhibition were reversed with antioxidant N-acetyl-cysteine, indicating that this adaptive response requires ROS signal. Inhibition of mitophagy using chloroquine in p53(-/-) or TIGAR(-/-) mice exacerbated accumulation of damaged mitochondria to the level of wild-type mice and attenuated cardioprotective action. These findings indicate that p53/TIGAR-mediated inhibition of myocyte mitophagy is responsible for impairment of mitochondrial integrity and subsequent apoptosis, the process of which is closely involved in p53-mediated ventricular remodeling after myocardial infarction.


Asunto(s)
Isquemia Miocárdica/metabolismo , Proteínas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis , Autofagia/genética , Regulación de la Expresión Génica , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/patología , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Isquemia Miocárdica/genética , Estrés Oxidativo , Monoéster Fosfórico Hidrolasas , Proteínas/genética , Especies Reactivas de Oxígeno/metabolismo , Proteína p53 Supresora de Tumor/genética , Remodelación Ventricular/genética
11.
Kidney Int ; 81(5): 449-57, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22157654

RESUMEN

Unilateral ureteral obstruction is a well-established experimental model of progressive renal fibrosis. We tested whether mechanical stretch and subsequent renal tubular distension might lead to renal fibrosis by first studying renal tubular epithelial cells in culture. We found that mechanical stretch induced reactive oxygen species that in turn activated the cytoplasmic proline-rich tyrosine kinase-2 (Pyk2). This kinase is abundantly expressed in tubular epithelial cells where it is activated by several stimuli. Using mice with deletion of Pyk2 we found that the expression of transforming growth factor-ß1 induced by mechanical stretch in renal tubular epithelial cells was significantly reduced. The expression of connective tissue growth factor was also reduced in the Pyk2(-/-) mice. We also found that expression of connective tissue growth factor was independent of transforming growth factor-ß1, but dependent on the Rho-associated coiled-coil forming protein kinase pathway. Thus, Pyk2 may be an important initiating factor in renal fibrosis and might be a new therapeutic target for ameliorating renal fibrosis.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Quinasa 2 de Adhesión Focal/metabolismo , Túbulos Renales/metabolismo , Riñón/metabolismo , Riñón/patología , Estrés Mecánico , Animales , Células Cultivadas , Células Epiteliales/metabolismo , Células Epiteliales/patología , Fibrosis , Quinasa 2 de Adhesión Focal/deficiencia , Quinasa 2 de Adhesión Focal/genética , Túbulos Renales/patología , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Fosforilación , Especies Reactivas de Oxígeno/metabolismo , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Obstrucción Ureteral/complicaciones
12.
Biochem Biophys Res Commun ; 425(1): 39-44, 2012 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-22820183

RESUMEN

Vascular calcification is a major risk factor for the cardiovascular disease, yet its underlying molecular mechanisms remain to be elucidated. Recently, we identified that osteogenic signals via bone morphogenetic protein (BMP)-2 exerted by vascular smooth muscle cells (VSMCs) play a crucial role in the formation of atherosclerotic plaque calcification. Here we report a synergistic interaction between macrophages and VSMCs with respect to plaque calcification. Treatment with conditioned medium (CM) of macrophages dramatically enhanced BMP-2 expression in VSMCs, while it substantially reduced the expression of matrix Gla-protein (MGP) that inhibits the BMP-2 osteogenic signaling. As a result, macrophages significantly accelerated the osteoblastic differentiation of C2C12 cells induced by VSMC-CM. In contrast, macrophage-CM did not enhance the osteoblastic gene expressions in VSMCs, indicating that macrophages unlikely induced the osteoblastic trans-differentiation of VSMCs. We then examined the effect of recombinant TNF-α and IL-1ß on the VSMC-derived osteogenic signals. Similar to the macrophage-CM, both cytokines enhanced BMP-2 expression and reduced MGP expression in VSMCs. Nevertheless, only the neutralization of TNF-α but not IL-1ß attenuated the effect of macrophage-CM on the expression of these genes in VSMCs, due to the very low concentration of IL-1ß in the macrophage-CM. On the other hand, VSMCs significantly enhanced IL-1ß expression in macrophages, which might in turn accelerate the VSMC-mediated osteogenic signals. Together, we identified a unique role of macrophages in the formation of plaque calcification in coordination with VSMCs. This interaction between macrophages and VSMCs is a potential therapeutic target to treat and prevent the atherosclerotic plaque calcification.


Asunto(s)
Macrófagos/inmunología , Músculo Liso Vascular/inmunología , Miocitos del Músculo Liso/inmunología , Osteogénesis/inmunología , Placa Aterosclerótica/inmunología , Calcificación Vascular/inmunología , Proteína Morfogenética Ósea 2/biosíntesis , Proteínas de Unión al Calcio/biosíntesis , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Proteínas de la Matriz Extracelular/biosíntesis , Humanos , Interleucina-1beta/farmacología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Osteoblastos/inmunología , Proteínas Recombinantes/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Calcificación Vascular/tratamiento farmacológico , Proteína Gla de la Matriz
13.
Arterioscler Thromb Vasc Biol ; 31(5): 1084-92, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21372295

RESUMEN

OBJECTIVE: Reactive oxygen species (ROS) are involved in the initial process of atherosclerosis, whereas it remains to be determined how atherogenic stimulus causes ROS-mediated proinflammatory reactions. Here, we focused on proline-rich tyrosine kinase (PYK2)-mediated ROS generation and examined how atherogenic stimulus causes early proinflammatory reactions. METHODS AND RESULTS: PYK2-deficient (knockout [KO]) (PYK2-KO) mice were crossbred with apolipoprotein E (ApoE)-deficient (PYK2-KO/ApoE-KO) mice. PYK2-KO/ApoE-KO mice and endothelial cells (EC) were used for the study. Aortic atherogenic lesions in PYK2-KO/ApoE-KO mice were markedly decreased (55% versus ApoE-KO) after 8 weeks of a Western diet. Aortic PYK2 was activated as early as 7 days after the Western diet, when inflammatory cells were not yet activated. Addition of the proatherogenic oxidized phospholipid lysophosphatidylcholine caused activation of endothelial PYK2. Lysophosphatidylcholine-activated PYK2 induced NADPH oxidase-mediated ROS generation and ROS-mediated synthesis of tumor necrosis factor-α (TNFα), vascular cell adhesion molecule-1 (VCAM-1), monocyte chemotactic protein-1 (MCP-1), and p21Cip1/Ets-1. Neutralizing anti-TNFα antibody or knockdown of p21Cip1/Ets-1 system blocked the induction of VCAM-1 and MCP-1. PYK2 deficiency abolished these ROS-mediated proinflammatory reactions. Further analysis revealed that PYK2/ROS-mediated p21Cip1/Ets-1 activation upregulated the transcription of the MCP-1 gene in collaboration with p300 transcription coactivator. CONCLUSIONS: PYK2 is a key tyrosine kinase activated by high cholesterol exposure, which causes ROS-mediated TNFα release and induces TNFα-dependent expression of proinflammatory molecules via the p21Cip1/Ets-1/p300 transcription system.


Asunto(s)
Enfermedades de la Aorta/enzimología , Aterosclerosis/enzimología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Endoteliales/enzimología , Quinasa 2 de Adhesión Focal/metabolismo , Mediadores de Inflamación/metabolismo , Proteína Proto-Oncogénica c-ets-1/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Animales , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Enfermedades de la Aorta/prevención & control , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/prevención & control , Trasplante de Médula Ósea , Células Cultivadas , Quimiocina CCL2/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Modelos Animales de Enfermedad , Células Endoteliales/patología , Quinasa 2 de Adhesión Focal/deficiencia , Quinasa 2 de Adhesión Focal/genética , Hipercolesterolemia/enzimología , Hipercolesterolemia/genética , Lipoproteínas LDL/metabolismo , Lisofosfatidilcolinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NADPH Oxidasas/metabolismo , Proteína Proto-Oncogénica c-ets-1/genética , Interferencia de ARN , Transducción de Señal , Factores de Tiempo , Activación Transcripcional , Transfección , Molécula 1 de Adhesión Celular Vascular/metabolismo , Factores de Transcripción p300-CBP/genética
14.
Ophthalmic Res ; 48(4): 199-207, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22868688

RESUMEN

PURPOSE: It has been reported that granulocyte colony-stimulating factor (G-CSF) provides neuroprotection in models in which neuronal cell death is induced. This research was designed to investigate the effects of G-CSF on neurodegeneration of the inner retinal layer in a rat model of ischemic reperfusion (I/R) injury. MATERIALS AND METHODS: Retinal ischemia was induced by increasing the intraocular pressure to 110 mm Hg for 45 min in the left eyes of the rats. A sham operation was carried out on the right eyes. G-CSF (100 µg/kg/day in 0.3 ml saline) or the same volume of saline was intraperitoneally injected just before the operation and continued for 4 consecutive days (a total of 5 consecutive days). Morphological examinations, including the terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay, were performed 7 days after I/R induction. The expression of phosphorylated AKT in the retina was examined by Western blot analysis and immunohistochemistry. RESULTS: Cell loss in the ganglion cell layer was more significantly reduced in the I/R-induced eyes of the G-CSF-injected rats than in the I/R-induced eyes of the saline-injected rats (20.3 vs. 6.6%). The inner retinal thickness ratios, such as the inner plexiform layer to the inner limiting membrane/outer nuclear layer and the inner nuclear layer/outer nuclear layer, were significantly better preserved in the I/R-induced eyes of the G-CSF-injected rats than in the I/R-induced eyes of the saline-injected rats. TUNEL assays showed fewer apoptotic cells in the retinal sections of the I/R-induced eyes of the G-CSF-injected rats. The phosphorylation of AKT (p-AKT/AKT) was upregulated in the retinas of the I/R-induced eyes of the G-CSF-injected rats. CONCLUSION: Our results demonstrated that systemic injection of G-CSF can protect retinal ganglion cells and inner retinal layers from I/R injury. The effects could be associated with the activation of AKT.


Asunto(s)
Modelos Animales de Enfermedad , Factor Estimulante de Colonias de Granulocitos/farmacología , Fármacos Neuroprotectores/farmacología , Daño por Reperfusión/prevención & control , Enfermedades de la Retina/prevención & control , Células Ganglionares de la Retina/efectos de los fármacos , Animales , Apoptosis , Western Blotting , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Inyecciones Intraperitoneales , Recuento de Leucocitos , Masculino , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Enfermedades de la Retina/metabolismo , Enfermedades de la Retina/patología , Células Ganglionares de la Retina/patología
15.
Am J Physiol Heart Circ Physiol ; 300(1): H154-61, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20952669

RESUMEN

Members of the fibroblast growth factor (FGF) family have been clinically applied to the treatment of ischemic diseases because of their strong angiogenic actions. Although tissue ischemia is predominantly caused by atherosclerosis, the roles of endothelial FGF receptors (FGF-Rs) in atherosclerosis remain obscure. We generated endothelial cell (EC)-targeted constitutively active FGF-R2-overexpressing mice, using the Tie2 promoter (Tie2-FGF-R2-Tg), and crossed them with apolipoprotein E (ApoE)-deficient mice (ApoE-KO) to generate Tie2-FGF-R2-Tg/ApoE-deficient mice (Tie2-FGF-R2-Tg/ApoE-KO). After being fed a Western diet for 8 wk, the Tie2-FGF-R2-Tg/ApoE-KO demonstrated 2.0-fold greater atherosclerotic lesion area on the luminal surfaces of the aortas than the ApoE-KO (P < 0.01). The level of p21(Cip1) protein, a cell cycle inhibitor, in the FGF-R2-overexpressing EC was 2.5-fold greater than that in the wild-type (WT) EC at the baseline (P < 0.01). FGF-R2 overexpression in the EC resulted in increased expression of VCAM-1 and ICAM-1, acceleration of apoptosis, and decreased proliferative activity, all of which were normalized by small interfering RNA (siRNA)-mediated knockdown of p21(Cip1) (75% reduction in protein level, P < 0.01). Furthermore, the expression of PDGF-B and Egr-1, a PDGF/p21(Cip1)-inducible transcription factor, in the aortic endothelium of Tie2-FGF-R2-Tg/ApoE-KO was significantly greater than that in ApoE-KO. The proliferation of vascular smooth muscle cells in the aortic media of Tie2-FGF-R2-Tg/ApoE-KO was 2.0-fold higher than that in ApoE-KO (P < 0.01). Thus our study reveals that endothelial FGF-R2 signaling aggravates atherosclerosis by promoting p21(Cip1)-mediated EC dysfunction and cautions against the use of FGF for therapeutic angiogenesis in the setting of atherosclerosis.


Asunto(s)
Aterosclerosis/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal/fisiología , Análisis de Varianza , Animales , Aorta/metabolismo , Aorta/fisiopatología , Apoptosis , Aterosclerosis/fisiopatología , Proliferación Celular , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Dieta , Endotelio Vascular/fisiopatología , Inmunohistoquímica , Masculino , Ratones , Ratones Transgénicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Circ Res ; 105(11): 1149-58, 2009 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-19834007

RESUMEN

RATIONALE: It has been reported that interleukin (IL)-1 is associated with pathological cardiac remodeling and LV dilatation, whereas IL-1beta has also been shown to induce cardiomyocyte hypertrophy. Thus, the role of IL-1 in the heart remains to be determined. OBJECTIVE: We studied the role of hypertrophy signal-mediated IL-1beta/insulin-like growth factor (IGF)-1 production in regulating the progression from compensative pressure-mediated hypertrophy to heart failure. METHODS AND RESULTS: Pressure overload was performed by aortic banding in IL-1beta-deficient mice. Primarily cultured cardiac fibroblasts (CFs) and cardiac myocytes (CMs) were exposed to cyclic stretch. Heart weight, myocyte size, and left ventricular ejection fraction were significantly lower in IL-1beta-deficient mice (20%, 23% and 27%, respectively) than in the wild type 30 days after aortic banding, whereas interstitial fibrosis was markedly augmented. DNA microarray analysis revealed that IGF-1 mRNA level was markedly (approximately 50%) decreased in the IL-1beta-deficient hypertrophied heart. Stretch of CFs, rather than CMs, abundantly induced the generation of IL-1beta and IGF-1, whereas such IGF-1 induction was markedly decreased in IL-1beta-deficient CFs. IL-1beta released by stretch is at a low level unable to induce IL-6 but sufficient to stimulate IGF-1 production. Promoter analysis showed that stretch-mediated IL-1beta activates JAK/STAT to transcriptionally regulate the IGF-1 gene. IL-1beta deficiency markedly increased c-Jun N-terminal kinase (JNK) and caspase-3 activities and enhanced myocyte apoptosis and fibrosis, whereas replacement of IGF-1 or JNK inhibitor restored them. CONCLUSIONS: We demonstrate for the first time that pressure-mediated hypertrophy and mechanical stretch generates a subinflammatory low level of IL-1beta, which constitutively causes IGF-1 production to maintain adaptable compensation hypertrophy and inhibit interstitial fibrosis.


Asunto(s)
Hipertrofia Ventricular Izquierda/metabolismo , Hipertrofia Ventricular Izquierda/fisiopatología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Interleucina-1beta/metabolismo , Animales , Apoptosis/fisiología , Fibrosis Endomiocárdica/metabolismo , Fibrosis Endomiocárdica/patología , Fibrosis Endomiocárdica/fisiopatología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hipertrofia Ventricular Izquierda/patología , Interleucina-1beta/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Janus Quinasa 2/metabolismo , Ratones , Ratones Mutantes , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Interleucina-1/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal/fisiología , Estrés Mecánico , Presión Ventricular/fisiología
17.
Nephrol Dial Transplant ; 26(4): 1252-7, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20817670

RESUMEN

BACKGROUND: A decreased plasma level of vitamin C has been reported to be associated with an increased risk of cardiovascular morbidity and mortality. Here, we sought to determine the vitamin C status of patients with chronic kidney disease and the pathophysiological role of vitamin C in these patients. METHODS: We studied 58 patients and evaluated the relationship between renal function and plasma vitamin C concentration, as well as the effect of diabetes on this relationship. Endothelium-dependent flow-mediated dilation of brachial artery was measured to assess the endothelial function. Serum malondialdehyde low-density lipoprotein was measured as a marker for oxidative stress. RESULTS: Plasma vitamin C concentration had a positive linear relationship with eGFR in both diabetic and non-diabetic patients (P = 0.006 and P = 0.004, respectively). When vitamin C concentration and eGFR relationships were compared in the two groups, vitamin C concentration was significantly lower in diabetic patients at every eGFR (P = 0.006). Flow-mediated vasodilatation of the brachial artery was positively correlated with vitamin C concentration in non-diabetic patients (P = 0.047) but not in diabetic patients. There was a negative correlation between serum malondialdehyde low-density lipoprotein and vitamin C concentration in non-diabetic patients (P = 0.044) but not in diabetic patients. CONCLUSIONS: Renal dysfunction was associated with a decrease in plasma vitamin C level. Moreover, decreased vitamin C may cause endothelial dysfunction via an increase in oxidative stress in non-diabetic chronic kidney disease patients.


Asunto(s)
Antioxidantes/metabolismo , Deficiencia de Ácido Ascórbico/complicaciones , Ácido Ascórbico/sangre , Diabetes Mellitus Tipo 2/fisiopatología , Insuficiencia Renal Crónica/etiología , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Pruebas de Función Renal , Masculino , Persona de Mediana Edad , Pronóstico , Insuficiencia Renal Crónica/sangre
18.
Arterioscler Thromb Vasc Biol ; 30(10): 1908-15, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20651281

RESUMEN

OBJECTIVE: Vascular calcification is an important risk factor for cardiovascular diseases. Here, we investigated a role of dedifferentiated vascular smooth muscle cells (VSMCs) in the atherosclerotic intimal calcification. METHODS AND RESULTS: We prepared human cultured VSMCs in either redifferentiatiated or dedifferentiated state and analyzed the gene expressions of bone-calcification regulatory factors. Expression of bone morphogenetic protein-2 (BMP-2), a potent initiator for osteoblast differentiation, was significantly enhanced in dedifferentiated VSMCs. Furthermore, endogenous BMP-2 antagonists, such as noggin, chordin, and matrix gamma-carboxyglutamic acid protein, were all downregulated in the dedifferentiated VSMCs. Conditioned medium from dedifferentiated VSMCs, but not from redifferentiated VSMCs, stimulated the osteoblastic differentiation of the mesenchymal progenitor C2C12 cells, which was abolished by BMP-2 knockdown. In atherosclerotic intima from apolipoprotein (apo)E-deficient mice, αSM-actin-positive cells, presumably dedifferentiated VSMCs, expressed BMP-2. We generated BMP-2-transgenic mice using αSM-actin promoter and crossed them with apoE-deficient mice (BMP-2-transgenic/apoE-knockout). Significantly accelerated atherosclerotic intimal calcification was detected in BMP-2-transgenic/apoE-knockout mice, although serum lipid concentration and atherosclerotic plaque size were not different from those in apoE-knockout mice. Enhanced calcification appeared to be associated with the frequent emergence of osteoblast-like cells in atherosclerotic intima in BMP-2-transgenic/apoE-knockout mice. CONCLUSIONS: Our findings collectively demonstrate an important role of dedifferentiated VSMCs in the pathophysiology of atherosclerotic calcification through activating paracrine BMP-2 osteogenic signals.


Asunto(s)
Aterosclerosis/etiología , Proteína Morfogenética Ósea 2/fisiología , Calcinosis/etiología , Osteogénesis/fisiología , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/fisiopatología , Proteína Morfogenética Ósea 2/genética , Calcinosis/genética , Calcinosis/patología , Calcinosis/fisiopatología , Desdiferenciación Celular , Diferenciación Celular , Células Cultivadas , Medios de Cultivo Condicionados , Femenino , Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modelos Biológicos , Miocitos del Músculo Liso/patología , Miocitos del Músculo Liso/fisiología , Osteoblastos/patología , Osteoblastos/fisiología , Osteogénesis/genética , Comunicación Paracrina , Túnica Íntima/patología , Túnica Íntima/fisiopatología
19.
Arterioscler Thromb Vasc Biol ; 30(1): 60-7, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19834109

RESUMEN

OBJECTIVE: Bone marrow (BM)-derived endothelial progenitor cells (EPCs) and vascular smooth muscle progenitor cells (VPCs) contribute to neointima formation, whereas the angiotensin II (Ang II) type 1 receptor (AT(1))-mediated action on BM-derived progenitors remains undefined. METHODS AND RESULTS: A wire-induced vascular injury was performed in the femoral artery of BM-chimeric mice whose BM was repopulated with AT(1)-deficient (BM-Agtr1(-/-)) or wild-type (BM-Agtr1(+/+)) cells. Neointima formation was profoundly reduced by 38% in BM-Agtr1(-/-) mice. Although the number of circulating EPCs (Sca-1(+)Flk-1(+)) and extent of reendothelialization did not differ between the 2 groups, the numbers of both circulating VPCs (c-Kit(-)Sca-1(+)Lin(-)) and tissue VPCs (Sca-1(+)CD31(-)) incorporated into neointima were markedly decreased in BM-Agtr1(-/-) mice. The accumulation of aggregated platelets and their content of stromal cell-derived factor-1alpha (SDF-1alpha) were significantly reduced in BM-Agtr1(-/-) mice, accompanied by a decrease in the serum level of SDF-1alpha. Thrombin-induced platelets aggregation was dose-dependently inhibited (45% at 0.1 IU/mL, P<0.05) in Agtr1(-/-) platelets compared with Agtr1(+/+) platelets, accompanied by the reduced expression and release of SDF-1alpha. CONCLUSIONS: The BM-AT(1) receptor promotes neointima formation by regulating the mobilization and homing of BM-derived VPCs in a platelet-derived SDF-1alpha-dependent manner without affecting EPC-mediated reendothelialization.


Asunto(s)
Médula Ósea/fisiología , Quimiocina CXCL12/metabolismo , Células Madre Hematopoyéticas/citología , Músculo Liso Vascular/citología , Receptor de Angiotensina Tipo 1/metabolismo , Animales , Anticuerpos/farmacología , Plaquetas/metabolismo , Linaje de la Célula/fisiología , Quimiocina CXCL12/inmunología , Quimiocina CXCL12/farmacología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Músculo Liso Vascular/metabolismo , Agregación Plaquetaria/fisiología , Receptor de Angiotensina Tipo 1/genética , Túnica Íntima/citología , Túnica Íntima/metabolismo
20.
Am J Physiol Heart Circ Physiol ; 299(6): H1908-16, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20935145

RESUMEN

Bioenergetic homeostasis is altered in heart failure and may play an important role in pathogenesis. p53 has been implicated in heart failure, and although its role in regulating tumorigenesis is well characterized, its activities on cellular metabolism are just beginning to be understood. We investigated the role of p53 and its transcriptional target gene TP53-induced glycolysis and apoptosis regulator (TIGAR) in myocardial energy metabolism under conditions simulating ischemia that can lead to heart failure. Expression of p53 and TIGAR was markedly upregulated after myocardial infarction, and apoptotic myocytes were decreased by 42% in p53-deficient mouse hearts compared with those in wild-type mice. To examine the effect of p53 on energy metabolism, cardiac myocytes were exposed to hypoxia. Hypoxia induced p53 and TIGAR expression in a p53-dependent manner. Knockdown of p53 or TIGAR increased glycolysis with elevated fructose-2,6-bisphosphate levels and reduced myocyte apoptosis. Hypoxic stress decreased phosphocreatine content and the mitochondrial membrane potential of myocytes without changes in ATP content, the effects of which were prevented by the knockdown of TIGAR. Inhibition of glycolysis by 2-deoxyglucose blocked these bioenergetic effects and TIGAR siRNA-mediated prevention of apoptosis, and, in contrast, overexpression of TIGAR reduced glucose utilization and increased apoptosis. Our data demonstrate that p53 and TIGAR inhibit glycolysis in hypoxic myocytes and that inhibition of glycolysis is closely involved in apoptosis, suggesting that p53 and TIGAR are significant mediators of cellular energy homeostasis and cell death under ischemic stress.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Metabolismo Energético , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas/metabolismo , Estrés Fisiológico , Proteína p53 Supresora de Tumor/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/genética , Hipoxia de la Célula , Células Cultivadas , Desoxiglucosa/metabolismo , Modelos Animales de Enfermedad , Glucólisis , Homeostasis , Potencial de la Membrana Mitocondrial , Ratones , Ratones Noqueados , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Miocitos Cardíacos/patología , Fosfocreatina/metabolismo , Fosfofructoquinasa-2/metabolismo , Monoéster Fosfórico Hidrolasas , Proteínas/genética , Interferencia de ARN , Ratas , Factores de Tiempo , Transfección , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA