Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 36(9): 1854-67, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27386938

RESUMO

OBJECTIVE: S100A6, a member of the S100 protein family, has been described as relevant for cell cycle entry and progression in endothelial cells. The molecular mechanism conferring S100A6's proliferative actions, however, remained elusive. APPROACH AND RESULTS: Originating from the clinically relevant observation of enhanced S100A6 protein expression in proliferating endothelial cells in remodeling coronary and carotid arteries, our study unveiled S100A6 as a suppressor of antiproliferative signal transducers and activators of transcription 1 signaling. Discovery of the molecular liaison was enabled by combining gene expression time series analysis with bioinformatic pathway modeling in S100A6-silenced human endothelial cells stimulated with vascular endothelial growth factor A. This unbiased approach led to successful identification and experimental validation of interferon-inducible transmembrane protein 1 and protein inhibitors of activated signal transducers and activators of transcription as key components of the link between S100A6 and signal transducers and activators of transcription 1. CONCLUSIONS: Given the important role of coordinated endothelial cell cycle activity for integrity and reconstitution of the inner lining of arterial blood vessels in health and disease, signal transducers and activators of transcription 1 suppression by S100A6 may represent a promising therapeutic target to facilitate reendothelialization in damaged vessels.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular , Proliferação de Células , Células Endoteliais/metabolismo , Proteínas S100/metabolismo , Fator de Transcrição STAT1/metabolismo , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/genética , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Biologia Computacional , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Perfilação da Expressão Gênica/métodos , Redes Reguladoras de Genes , Inativação Gênica , Humanos , Masculino , Proteínas Inibidoras de STAT Ativados/genética , Proteínas Inibidoras de STAT Ativados/metabolismo , Interferência de RNA , Ratos Sprague-Dawley , Reepitelização , Proteína A6 Ligante de Cálcio S100 , Proteínas S100/genética , Fator de Transcrição STAT1/genética , Transdução de Sinais , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sus scrofa , Fatores de Tempo , Transcriptoma , Transfecção , Fator A de Crescimento do Endotélio Vascular/farmacologia , Lesões do Sistema Vascular/genética , Lesões do Sistema Vascular/metabolismo , Lesões do Sistema Vascular/patologia
2.
Mol Ther ; 23(8): 1320-1330, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26005840

RESUMO

Restoring expression levels of the EF-hand calcium (Ca(2+)) sensor protein S100A1 has emerged as a key factor in reconstituting normal Ca(2+) handling in failing myocardium. Improved sarcoplasmic reticulum (SR) function with enhanced Ca(2+) resequestration appears critical for S100A1's cyclic adenosine monophosphate-independent inotropic effects but raises concerns about potential diastolic SR Ca(2+) leakage that might trigger fatal arrhythmias. This study shows for the first time a diminished interaction between S100A1 and ryanodine receptors (RyR2s) in experimental HF. Restoring this link in failing cardiomyocytes, engineered heart tissue and mouse hearts, respectively, by means of adenoviral and adeno-associated viral S100A1 cDNA delivery normalizes diastolic RyR2 function and protects against Ca(2+)- and ß-adrenergic receptor-triggered proarrhythmogenic SR Ca(2+) leakage in vitro and in vivo. S100A1 inhibits diastolic SR Ca(2+) leakage despite aberrant RyR2 phosphorylation via protein kinase A and calmodulin-dependent kinase II and stoichiometry with accessory modulators such as calmodulin, FKBP12.6 or sorcin. Our findings demonstrate that S100A1 is a regulator of diastolic RyR2 activity and beneficially modulates diastolic RyR2 dysfunction. S100A1 interaction with the RyR2 is sufficient to protect against basal and catecholamine-triggered arrhythmic SR Ca(2+) leak in HF, combining antiarrhythmic potency with chronic inotropic actions.


Assuntos
Insuficiência Cardíaca/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Proteínas S100/metabolismo , Animais , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Calmodulina/metabolismo , DNA Complementar/metabolismo , Eletrocardiografia , Técnicas de Transferência de Genes , Insuficiência Cardíaca/prevenção & controle , Masculino , Camundongos , Microscopia de Fluorescência , Miocárdio/metabolismo , Miócitos Cardíacos/citologia , Fosforilação , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo , Proteínas de Ligação a Tacrolimo/metabolismo , Engenharia Tecidual/métodos
3.
Circ Res ; 112(1): 66-78, 2013 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-23048072

RESUMO

RATIONALE: Mice lacking the EF-hand Ca2+ sensor S100A1 display endothelial dysfunction because of distorted Ca2+ -activated nitric oxide (NO) generation. OBJECTIVE: To determine the pathophysiological role of S100A1 in endothelial cell (EC) function in experimental ischemic revascularization. METHODS AND RESULTS: Patients with chronic critical limb ischemia showed almost complete loss of S100A1 expression in hypoxic tissue. Ensuing studies in S100A1 knockout (SKO) mice subjected to femoral artery resection unveiled insufficient perfusion recovery and high rates of autoamputation. Defective in vivo angiogenesis prompted cellular studies in SKO ECs and human ECs, with small interfering RNA-mediated S100A1 knockdown demonstrating impaired in vitro and in vivo proangiogenic properties (proliferation, migration, tube formation) and attenuated vascular endothelial growth factor (VEGF)-stimulated and hypoxia-stimulated endothelial NO synthase (eNOS) activity. Mechanistically, S100A1 deficiency compromised eNOS activity in ECs by interrupted stimulatory S100A1/eNOS interaction and protein kinase C hyperactivation that resulted in inhibitory eNOS phosphorylation and enhanced VEGF receptor-2 degradation with attenuated VEGF signaling. Ischemic SKO tissue recapitulated the same molecular abnormalities with insufficient in vivo NO generation. Unresolved ischemia entailed excessive VEGF accumulation in SKO mice with aggravated VEGF receptor-2 degradation and blunted in vivo signaling through the proangiogenic phosphoinositide-3-kinase/Akt/eNOS cascade. The NO supplementation strategies rescued defective angiogenesis and salvaged limbs in SKO mice after femoral artery resection. CONCLUSIONS: Our study shows for the first time downregulation of S100A1 expression in patients with critical limb ischemia and identifies S100A1 as critical for EC function in postnatal ischemic angiogenesis. These findings link its pathological plasticity in critical limb ischemia to impaired neovascularization, prompting further studies to probe the microvascular therapeutic potential of S100A1.


Assuntos
Células Endoteliais/enzimologia , Isquemia/enzimologia , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Óxido Nítrico Sintase Tipo III/metabolismo , Proteínas S100/deficiência , Idoso , Idoso de 80 Anos ou mais , Animais , Cálcio/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Feminino , Membro Posterior , Células Endoteliais da Veia Umbilical Humana/enzimologia , Humanos , Isquemia/tratamento farmacológico , Isquemia/genética , Isquemia/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Músculo Esquelético/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/farmacologia , Fosfatidilinositol 3-Quinase/metabolismo , Fosforilação , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Fluxo Sanguíneo Regional , Proteínas S100/genética , Transdução de Sinais , Fatores de Tempo , Transfecção , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
4.
Circ Res ; 108(1): 27-39, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-21106943

RESUMO

RATIONALE: The G(ßγ)-sequestering peptide ß-adrenergic receptor kinase (ßARK)ct derived from the G-protein-coupled receptor kinase (GRK)2 carboxyl terminus has emerged as a promising target for gene-based heart failure therapy. Enhanced downstream cAMP signaling has been proposed as the underlying mechanism for increased ß-adrenergic receptor (ßAR) responsiveness. However, molecular targets mediating improved cardiac contractile performance by ßARKct and its impact on G(ßγ)-mediated signaling have yet to be fully elucidated. OBJECTIVE: We sought to identify G(ßγ)-regulated targets and signaling mechanisms conveying ßARKct-mediated enhanced ßAR responsiveness in normal (NC) and failing (FC) adult rat ventricular cardiomyocytes. METHODS AND RESULTS: Assessing viral-based ßARKct gene delivery with electrophysiological techniques, analysis of contractile performance, subcellular Ca²(+) handling, and site-specific protein phosphorylation, we demonstrate that ßARKct enhances the cardiac L-type Ca²(+) channel (LCC) current (I(Ca)) both in NCs and FCs on ßAR stimulation. Mechanistically, ßARKct augments I(Ca) by preventing enhanced inhibitory interaction between the α1-LCC subunit (Cav1.2α) and liberated G(ßγ) subunits downstream of activated ßARs. Despite improved ßAR contractile responsiveness, ßARKct neither increased nor restored cAMP-dependent protein kinase (PKA) and calmodulin-dependent kinase II signaling including unchanged protein kinase (PK)Cε, extracellular signal-regulated kinase (ERK)1/2, Akt, ERK5, and p38 activation both in NCs and FCs. Accordingly, although ßARKct significantly increases I(Ca) and Ca²(+) transients, being susceptible to suppression by recombinant G(ßγ) protein and use-dependent LCC blocker, ßARKct-expressing cardiomyocytes exhibit equal basal and ßAR-stimulated sarcoplasmic reticulum Ca²(+) load, spontaneous diastolic Ca²(+) leakage, and survival rates and were less susceptible to field-stimulated Ca²(+) waves compared with controls. CONCLUSION: Our study identifies a G(ßγ)-dependent signaling pathway attenuating cardiomyocyte I(Ca) on ßAR as molecular target for the G(ßγ)-sequestering peptide ßARKct. Targeted interruption of this inhibitory signaling pathway by ßARKct confers improved ßAR contractile responsiveness through increased I(Ca) without enhancing regular or restoring abnormal cAMP-signaling. ßARKct-mediated improvement of I(Ca) rendered cardiomyocytes neither susceptible to ßAR-induced damage nor arrhythmogenic sarcoplasmic reticulum Ca²(+) leakage.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Cálcio/metabolismo , Cardiotônicos/metabolismo , Quinase 2 de Receptor Acoplado a Proteína G , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Terapia Genética/métodos , Insuficiência Cardíaca , Contração Miocárdica/genética , Miócitos Cardíacos/metabolismo , Peptídeos/metabolismo , Animais , Canais de Cálcio Tipo L/genética , Sobrevivência Celular/genética , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades gama da Proteína de Ligação ao GTP/genética , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/terapia , Ventrículos do Coração/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Peptídeos/genética , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Ratos , Retículo Sarcoplasmático/genética , Retículo Sarcoplasmático/metabolismo
5.
Arterioscler Thromb Vasc Biol ; 32(2): 308-16, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22095977

RESUMO

OBJECTIVE: G protein-coupled receptor kinase-5 (GRK5) is a widely expressed Ser/Thr kinase that regulates several atherogenic receptors and may activate or inhibit nuclear factor-κB (NF-κB). This study sought to determine whether and by what mechanisms GRK5 affects atherosclerosis. METHODS AND RESULTS: Grk5(-/-)/Apoe(-/-) mice developed 50% greater aortic atherosclerosis than Apoe(-/-) mice and demonstrated greater proliferation of macrophages and smooth muscle cells (SMCs) in atherosclerotic lesions. In Apoe(-/-) mice, carotid interposition grafts from Grk5(-/-) mice demonstrated greater upregulation of cell adhesion molecules than grafts from wild-type mice and, subsequently, more atherosclerosis. By comparing Grk5(-/-) with wild-type cells, we found that GRK5 desensitized 2 key atherogenic receptor tyrosine kinases: the platelet-derived growth factor receptor-ß in SMCs, by augmenting ubiquitination/degradation; and the colony-stimulating factor-1 receptor (CSF-1R) in macrophages, by reducing CSF-1-induced tyrosyl phosphorylation. GRK5 activity in monocytes also reduced migration promoted by the 7-transmembrane receptor for monocyte chemoattractant protein-1 CC chemokine receptor-2. Whereas GRK5 diminished NF-κB-dependent gene expression in SMCs and endothelial cells, it had no effect on NF-κB activity in macrophages. CONCLUSIONS: GRK5 attenuates atherosclerosis through multiple cell type-specific mechanisms, including reduction of SMC and endothelial cell NF-κB activity and desensitization of receptor-specific signaling through the monocyte CC chemokine receptor-2, macrophage CSF-1R, and the SMC platelet-derived growth factor receptor-ß.


Assuntos
Aterosclerose/metabolismo , Aterosclerose/prevenção & controle , Quinase 5 de Receptor Acoplado a Proteína G/metabolismo , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores CCR2/metabolismo , Transdução de Sinais/fisiologia , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/fisiopatologia , Movimento Celular/fisiologia , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Quinase 5 de Receptor Acoplado a Proteína G/deficiência , Quinase 5 de Receptor Acoplado a Proteína G/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , NF-kappa B/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptor 4 Toll-Like/metabolismo
6.
Hum Mol Genet ; 19(14): 2754-66, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20421368

RESUMO

Aging is believed to be among the most important contributors to atherosclerosis, through mechanisms that remain largely obscure. Serum levels of tumor necrosis factor (TNF) rise with aging and have been correlated with the incidence of myocardial infarction. We therefore sought to determine whether genetic variation in the TNF receptor-1 gene (TNFR1) contributes to aging-related atherosclerosis in humans and whether Tnfr1 expression aggravates aging-related atherosclerosis in mice. With 1330 subjects from a coronary angiography database, we performed a case-control association study of coronary artery disease (CAD) with 16 TNFR1 single-nucleotide polymorphisms (SNPs). Two TNFR1 SNPs significantly associated with CAD in subjects >55 years old, and this association was supported by analysis of a set of 759 independent CAD cases. In multiple linear regression analysis, accounting for TNFR1 SNP rs4149573 significantly altered the relationship between aging and CAD index among 1811 subjects from the coronary angiography database. To confirm that TNFR1 contributes to aging-dependent atherosclerosis, we grafted carotid arteries from 18- and 2-month-old wild-type (WT) and Tnfr1(-/-) mice into congenic apolipoprotein E-deficient (Apoe(-/-)) mice and harvested grafts from 1 to 7 weeks post-operatively. Aged WT arteries developed accelerated atherosclerosis associated with enhanced TNFR1 expression, enhanced macrophage recruitment, reduced smooth muscle cell proliferation and collagen content, augmented apoptosis and plaque hemorrhage. In contrast, aged Tnfr1(-/-) arteries developed atherosclerosis that was indistinguishable from that in young Tnfr1(-/-) arteries and significantly less than that observed in aged WT arteries. We conclude that TNFR1 polymorphisms associate with aging-related CAD in humans, and TNFR1 contributes to aging-dependent atherosclerosis in mice.


Assuntos
Envelhecimento/fisiologia , Artérias/metabolismo , Aterosclerose/genética , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/genética , Animais , Aterosclerose/etiologia , Aterosclerose/metabolismo , Modelos Animais de Doenças , Feminino , Expressão Gênica/fisiologia , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Humanos , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/fisiologia , Regulação para Cima/genética
7.
Curr Med Chem ; 29(3): 453-466, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34047257

RESUMO

Sickle Cell Disease (SCD) is one of the most common monogenic disorders caused by a point mutation in the ß-globin gene. This mutation results in polymerization of hemoglobin (Hb) under reduced oxygenation conditions, causing rigid sickle-shaped RBCs and hemolytic anemia. This clearly defined fundamental molecular mechanism makes SCD a prototypical target for precision therapy. Both the mutant ß-globin protein and its downstream pathophysiology are pharmacological targets of intensive research. SCD also is a disease well-suited for biological interventions like gene therapy. Recent advances in hematopoietic stem cell (HSC) transplantation and gene therapy platforms, like Lentiviral vectors and gene editing strategies, expand the potentially curative options for patients with SCD. This review discusses the recent advances in precision therapy for SCD and the preclinical and clinical advances in autologous HSC gene therapy for SCD.


Assuntos
Anemia Falciforme , Transplante de Células-Tronco Hematopoéticas , Anemia Falciforme/genética , Anemia Falciforme/terapia , Edição de Genes , Terapia Genética , Humanos , Globinas beta/genética
8.
Am J Physiol Heart Circ Physiol ; 301(5): H1932-40, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21890689

RESUMO

Akt2 protein kinase has been shown to promote cell migration and actin polymerization in several cell types, including macrophages. Because migrating macrophages constitute an important inflammatory response after myocardial ischemia, we determined cardiac macrophage expression after ischemia-reperfusion (I/R) injury and cryo-injury in mice lacking Akt2 (Akt2-KO). At 7 days post-I/R, Akt2-KO cardiac tissues showed an increase in immunohistochemical staining for macrophage markers (Galectin 3 and F4/80) compared with wild-type (WT) mice, indicating macrophage density was increased in the injured Akt2-KO myocardium. This change was time dependent because macrophage density was similar between WT and Akt2-KO myocardium at 3 days post-I/R, but by 7 and 14 days post-I/R, macrophage density was significantly increased in Akt2-KO myocardium. Concomitantly, infarct size was larger and cardiac function was reduced in Akt2-KO mice subjected to I/R. However, when cryo-infarction produced similar infarct sizes in the anterior wall in both WT and Akt2-KO mice, macrophage density remained higher in Akt2-KO mouse myocardium, suggesting Akt2 regulates myocardial macrophage density independent of infarct size. Consistently, bone marrow from Akt2-KO mice enhanced myocardial macrophage density in both C57/B6 WT and Akt2-KO recipient mice. Finally, reciprocal ex-vivo coculturing of macrophages and cardiac myocytes showed that activated Akt2-KO peritoneal macrophages had reduced mobility and adhesion when compared with WT littermate controls. Thus, although Akt-2 KO mice did not affect the initial inflammation response after injury and Akt2 deficiency has been shown to impair cell migration or motility in macrophages, our data suggested a novel mechanism in which increasing retention of Akt2-KO macrophages resulted in increasing cardiac Akt2-KO macrophage density in the myocardial space.


Assuntos
Movimento Celular , Macrófagos/enzimologia , Infarto do Miocárdio/enzimologia , Traumatismo por Reperfusão Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio/enzimologia , Proteínas Proto-Oncogênicas c-akt/deficiência , Animais , Antígenos de Diferenciação/metabolismo , Apoptose , Transplante de Medula Óssea , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Galectina 3/metabolismo , Regulação da Expressão Gênica , Hemodinâmica , Imuno-Histoquímica , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miocárdio/patologia , Proteínas Proto-Oncogênicas c-akt/genética , Fatores de Tempo , Função Ventricular Esquerda , Cicatrização
9.
CPT Pharmacometrics Syst Pharmacol ; 10(7): 696-708, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34139105

RESUMO

We developed a mathematical model for autologous stem cell therapy to cure sickle cell disease (SCD). Experimental therapies using this approach seek to engraft stem cells containing a curative gene. These stem cells are expected to produce a lifelong supply of red blood cells (RBCs) containing an anti-sickling hemoglobin. This complex, multistep treatment is expensive, and there is limited patient data available from early clinical trials. Our objective was to quantify the impact of treatment parameters, such as initial stem cell dose, efficiency of lentiviral transduction, and degree of bone marrow preconditioning on engraftment efficiency, peripheral RBC numbers, and anti-sickling hemoglobin levels over time. We used ordinary differential equations to model RBC production from progenitor cells in the bone marrow, and hemoglobin assembly from its constituent globin monomers. The model recapitulates observed RBC and hemoglobin levels in healthy and SCD phenotypes. Treatment simulations predict dynamics of stem cell engraftment and RBC containing the therapeutic gene product. Post-treatment dynamics show an early phase of reconstitution due to short lived stem cells, followed by a sustained RBC production from stable engraftment of long-term stem cells. This biphasic behavior was previously reported in the literature. Sensitivity analysis of the model quantified relationships between treatment parameters and efficacy. The initial dose of transduced stem cells, and the intensity of myeloablative bone marrow preconditioning are predicted to most positively impact long-term outcomes. The quantitative systems pharmacology approach used here demonstrates the value of model-assisted therapeutic design for gene therapies in SCD.


Assuntos
Anemia Falciforme/terapia , Terapia Genética/métodos , Modelos Teóricos , Transplante de Células-Tronco/métodos , Anemia Falciforme/genética , Células da Medula Óssea/citologia , Eritrócitos/citologia , Hemoglobinas/metabolismo , Humanos , Farmacologia em Rede
10.
Circ Res ; 103(1): 70-9, 2008 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-18519945

RESUMO

Atherosclerosis and arterial injury-induced neointimal hyperplasia involve medial smooth muscle cell (SMC) proliferation and migration into the arterial intima. Because many 7-transmembrane and growth factor receptors promote atherosclerosis, we hypothesized that the multifunctional adaptor proteins beta-arrestin1 and -2 might regulate this pathological process. Deficiency of beta-arrestin2 in ldlr(-/-) mice reduced aortic atherosclerosis by 40% and decreased the prevalence of atheroma SMCs by 35%, suggesting that beta-arrestin2 promotes atherosclerosis through effects on SMCs. To test this potential atherogenic mechanism more specifically, we performed carotid endothelial denudation in congenic wild-type, beta-arrestin1(-/-), and beta-arrestin2(-/-) mice. Neointimal hyperplasia was enhanced in beta-arrestin1(-/-) mice, and diminished in beta-arrestin2(-/-) mice. Neointimal cells expressed SMC markers and did not derive from bone marrow progenitors, as demonstrated by bone marrow transplantation with green fluorescent protein-transgenic cells. Moreover, the reduction in neointimal hyperplasia seen in beta-arrestin2(-/-) mice was not altered by transplantation with either wild-type or beta-arrestin2(-/-) bone marrow cells. After carotid injury, medial SMC extracellular signal-regulated kinase activation and proliferation were increased in beta-arrestin1(-/-) and decreased in beta-arrestin2(-/-) mice. Concordantly, thymidine incorporation and extracellular signal-regulated kinase activation and migration evoked by 7-transmembrane receptors were greater than wild type in beta-arrestin1(-/-) SMCs and less in beta-arrestin2(-/-) SMCs. Proliferation was less than wild type in beta-arrestin2(-/-) SMCs but not in beta-arrestin2(-/-) endothelial cells. We conclude that beta-arrestin2 aggravates atherosclerosis through mechanisms involving SMC proliferation and migration and that these SMC activities are regulated reciprocally by beta-arrestin2 and beta-arrestin1. These findings identify inhibition of beta-arrestin2 as a novel therapeutic strategy for combating atherosclerosis and arterial restenosis after angioplasty.


Assuntos
Aorta/metabolismo , Arrestinas/metabolismo , Aterosclerose/metabolismo , Movimento Celular , Proliferação de Células , Miócitos de Músculo Liso/metabolismo , Animais , Aorta/patologia , Arrestinas/genética , Aterosclerose/genética , Aterosclerose/patologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Oclusão de Enxerto Vascular/genética , Oclusão de Enxerto Vascular/metabolismo , Oclusão de Enxerto Vascular/patologia , Hiperplasia/genética , Hiperplasia/metabolismo , Hiperplasia/patologia , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Camundongos Knockout , Miócitos de Músculo Liso/patologia , Receptores de LDL/genética , Receptores de LDL/metabolismo , beta-Arrestinas
11.
Arterioscler Thromb Vasc Biol ; 28(2): 284-9, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18006858

RESUMO

OBJECTIVE: Inflammation appears intricately linked to vein graft arterialization. We have previously shown that tumor necrosis factor (TNF) receptor-1 (TNFR1, p55) signaling augments vein graft neointimal hyperplasia (NH) and remodeling through its effects on vascular smooth muscle cells (SMCs). In this study we examined the role of TNFR2 (p75) signaling in vein graft arterialization. METHODS AND RESULTS: Inferior vena cava-to-carotid artery interposition grafting was performed between p75-/- and congenic (C57B1/6J) wild-type (WT) mice. Six weeks postoperatively, neointimal and medial dimensions were greater in p75-/- grafts placed into p75-/- recipients (by 42% or 60%, respectively; P<0.05), when compared with WT veins grafted into WT recipients. Relative to WT vein grafts, p75 deficiency augmented early (2-week-old) graft vascular cell adhesion molecule (VCAM)-1 expression (by 2.4-fold, P<0.05), increased endothelial cell apoptosis (2-fold), and delayed graft re-endothelialization. Both cellular proliferation in early, and collagen I content of mature (6-week-old) vein grafts were increased (by 70% and 50%, respectively) in p75-/- grafts. P75 deficiency augmented TNF-induced apoptosis of cultured endothelial cells, but did not affect TNF-stimulated SMC proliferation or migration induced by co-cultured macrophages. CONCLUSIONS: TNF signaling via p75 reduces vein graft neointimal hyperplasia through mechanisms involving reduction of adhesion molecule expression and endothelial cell apoptosis.


Assuntos
Endotélio Vascular/fisiopatologia , Hiperplasia/fisiopatologia , Receptores Tipo II do Fator de Necrose Tumoral/fisiologia , Túnica Íntima/fisiopatologia , Veia Cava Inferior/transplante , Animais , Apolipoproteínas E/genética , Apoptose , Proliferação de Células , Modelos Animais de Doenças , Masculino , Camundongos , Receptores de Fator de Crescimento Neural/genética , Transdução de Sinais/fisiologia
12.
Arterioscler Thromb Vasc Biol ; 27(5): 1087-94, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17442899

RESUMO

OBJECTIVE: Mechanisms by which tumor necrosis factor-alpha (TNF) contributes to atherosclerosis remain largely obscure. We therefore sought to determine the role of the arterial wall TNF receptor-1 (TNFR1) in atherogenesis. METHODS AND RESULTS: Carotid artery-to-carotid artery interposition grafting was performed with tnfr1-/- and congenic (C57Bl/6) wild-type (WT) mice as graft donors, and congenic chow-fed apolipoprotein E-deficient mice as recipients. Advanced atherosclerotic graft lesions developed within 8 weeks, and had 2-fold greater area in WT than in tnfr1-/- grafts. While the prevalence of specific atheroma cells was equivalent in WT and tnfr1-/- grafts, the overall abundance of cells was substantially greater in WT grafts. WT grafts demonstrated greater MCP-1, vascular cell adhesion molecule-1, and intercellular adhesion molecule-1 expression at both early and late time points, and proliferating cell nuclear antigen expression at early time points. Aortic atherosclerosis was also reduced in 14-month-old apoe(-/-)/tnfr1(-/-) mice, as compared with cognate apoe-/- mice. In coculture with activated macrophages, smooth muscle cells expressing the TNFR1 demonstrated enhanced migration and reduced scavenger receptor activity. CONCLUSIONS: TNFR1 signaling, just in arterial wall cells, contributes to the pathogenesis of atherosclerosis by enhancing arterial wall chemokine and adhesion molecule expression, as well as by augmenting medial smooth muscle cell proliferation and migration.


Assuntos
Aterosclerose/metabolismo , Doenças das Artérias Carótidas/metabolismo , Artéria Carótida Primitiva/patologia , Receptores Tipo I de Fatores de Necrose Tumoral/biossíntese , Animais , Aterosclerose/patologia , Bioensaio , Biomarcadores/metabolismo , Doenças das Artérias Carótidas/patologia , Artéria Carótida Primitiva/metabolismo , Artéria Carótida Primitiva/cirurgia , Modelos Animais de Doenças , Progressão da Doença , Seguimentos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
13.
Cardiovasc Res ; 65(3): 674-82, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15664394

RESUMO

OBJECTIVE: Because tumor necrosis factor-alpha (TNF) has been implicated in the pathogenesis of vein graft neointimal hyperplasia, we sought to determine mechanisms by which TNF could induce proliferative and migratory responses in smooth muscle cells (SMCs). METHODS AND RESULTS: In rabbit jugulocarotid interposition vein grafts, SMCs expressed TNF as early as four days postoperatively. In rabbit aortic SMCs, TNF and platelet-derived growth factor (PDGF) elicited comparable migration (1.7-fold/basal), and their effects were partially additive. In contrast, while TNF failed to promote SMC [(3)H]thymidine incorporation alone, it doubled the [(3)H]thymidine incorporation observed with PDGF alone. To gain mechanistic insight into these phenomena, we found that TNF and PDGF each activated p38(mapk) equivalently in SMCs, but that PDGF was two to three times more efficacious than TNF in activating SMC extracellular signal-regulated kinases (ERK) 1 and 2 and phosphoinositide 3-kinase. However, only TNF activated NF kappa B. SMC [(3)H]thymidine incorporation that depended on TNF, but not PDGF, was abolished by overexpression of a dominant-negative I kappa B alpha mutant. Inhibition of ERK activation by U0126 reduced SMC migration stimulated only by PDGF (by 35%, P<0.05), but not by TNF. Inhibition of phosphoinositide 3-kinase by LY294002, however, significantly reduced both TNF- and PDGF-stimulated chemotaxis (by 38-54%, P<0.05). In contrast, both U0126 and LY294002 abolished SMC [(3)H]thymidine incorporation induced by either TNF, PDGF, or both agonists. CONCLUSIONS: In primary rabbit SMCs, TNF promotes migration and mitogenesis through signaling mechanisms that are both distinct from and overlapping with those employed by PDGF. TNF-induced SMC mitogenesis requires complementary co-stimulation with other growth factors.


Assuntos
Músculo Liso Vascular/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Animais , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Sinergismo Farmacológico , Oclusão de Enxerto Vascular/metabolismo , Oclusão de Enxerto Vascular/patologia , Hiperplasia , Masculino , Músculo Liso Vascular/citologia , NF-kappa B/fisiologia , Coelhos , Ratos , Fator de Necrose Tumoral alfa/biossíntese , Túnica Íntima/patologia , Veias/transplante
14.
FASEB J ; 17(3): 523-5, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12631586

RESUMO

The role of Galpha(i)-2 overexpression in desensitization of beta-adrenergic signaling in heart failure is controversial. An adenovirus-based approach was used to investigate whether overexpression of Galpha(i)-2 impairs beta-adrenergic stimulation of adenylyl cyclase (AC) activity and cAMP levels in neonatal rat cardiac myocytes (NRCM) and cell shortening of adult rat ventricular myocytes (ARVM). Infection of NRCM with Ad5Galpha(i)-2 increased Galpha(i)-2 by 50-600% in a virus dose-dependent manner. Overexpression was paralleled by suppression of GTP- and isoprenaline-stimulated AC by 10-72% (P<0.001) in a PTX-sensitive manner. Isoprenaline-stimulated shortening of Ad5Galpha(i)-2-infected ARVM was attenuated by 34% (P<0.01). Ad5Galpha(i)-2/GFP (Galpha(i)-2, green fluorescent protein; bicistronic) was constructed to monitor transfection homogeneity and target Galpha(i)-2 overexpression to levels found in heart failure. At Galpha(i)-2 levels of 93% above control, isoprenaline-stimulated AC activity and cAMP levels were reduced by 17% and 40% (P<0.02), respectively. Beta1- and beta2-adrenergic stimulation was reduced similarly. Our results suggest that (a) the Galpha(i)-2 system exhibits tonic inhibition of stimulated AC in cardiac myocytes, (b) Galpha(i)-2-mediated inhibition is concentration-dependent and occurs at Galpha(i)-2 levels seen in heart failure, and (c) Galpha(i)-2-mediated inhibition affects both beta1- and beta2-adrenergic stimulation of AC. The data argue for an important, independent role of the Galpha(i)-2 increase in heart failure.


Assuntos
Agonistas Adrenérgicos beta/farmacologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Isoproterenol/antagonistas & inibidores , Miócitos Cardíacos/metabolismo , Proteínas Proto-Oncogênicas/genética , Transdução de Sinais , Adenoviridae/genética , Adenilil Ciclases/metabolismo , Animais , Células Cultivadas , AMP Cíclico/metabolismo , Subunidade alfa Gi2 de Proteína de Ligação ao GTP , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/antagonistas & inibidores , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Expressão Gênica , Vetores Genéticos , Modelos Biológicos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Toxina Pertussis/farmacologia , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/metabolismo , Ratos
15.
Arterioscler Thromb Vasc Biol ; 24(3): 470-6, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14726410

RESUMO

OBJECTIVE: Vein graft disease involves neointimal smooth muscle cells, the origins of which are unclear. This study sought to characterize and quantitate vein graft infiltration by cells extrinsic to the graft in a mouse model of vein graft disease. METHODS AND RESULTS: Inferior vena cava-to-carotid artery interposition grafting between C57Bl/6 and congenic beta-galactosidase-expressing ROSA26 mice was performed. Vein grafts were harvested 6 weeks postoperatively and stained with X-gal. More than 60% of neointimal cells derived from the recipient, and 50% of these cells expressed smooth muscle alpha-actin. The distribution of donor and recipient-derived cells within this vein graft wall layer was distinctly focal, consistent with focal infiltration and expansion of progenitor cells. When bone marrow transplantation with congenic green fluorescent protein (GFP)-expressing cells was used in vein graft recipients 1 month before surgery, abundant GFP-expressing cells appeared in the media, but not the neointima, of mature grafts. Endothelial cells in mature grafts derived from graft-intrinsic and graft-extrinsic sources and were, in part, of bone marrow origin. CONCLUSIONS: Cells extrinsic to the graft, including bone marrow-derived cells, predominate during vein graft remodeling.


Assuntos
Artéria Carótida Primitiva/cirurgia , Miócitos de Músculo Liso/citologia , Túnica Íntima/citologia , Veia Cava Inferior/transplante , Anastomose Cirúrgica , Animais , Animais Congênicos , Transplante de Medula Óssea , Artéria Carótida Primitiva/citologia , Contagem de Células , Diferenciação Celular , Linhagem da Célula , Movimento Celular , Sobrevivência Celular , Células Endoteliais/citologia , Genes Reporter , Sobrevivência de Enxerto , Proteínas de Fluorescência Verde , Técnicas Imunoenzimáticas , Óperon Lac , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Quimera por Radiação , Células-Tronco/citologia , Subpopulações de Linfócitos T/citologia , Veia Cava Inferior/citologia
16.
Arterioscler Thromb Vasc Biol ; 24(12): 2277-83, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15486311

RESUMO

OBJECTIVE: Vein graft remodeling and neointimal hyperplasia involve inflammation, graft-intrinsic cells, and recruitment of vascular progenitor cells. We sought to examine if the inflammatory cytokine tumor necrosis factor (TNF) affects vein graft remodeling via its p55 TNF receptor-1 (p55). METHODS AND RESULTS: Inferior vena cava-to-carotid artery interposition grafting was performed between p55-/- and congenic (C57Bl/6) wild-type (WT) mice. Immunofluorescence revealed TNF in early (2-week) vein grafts. Six weeks postoperatively, luminal and medial areas were indistinguishable among all vein graft groups. However, neointimal area was reduced in p55-/- grafts: by 40% in p55-/- grafts placed in p55-/- recipients, and by 21% in p55-/- grafts placed in WT recipients, compared with WT grafts in WT recipients (P<0.05). In 2-week-old vein grafts, p55 deficiency reduced intercellular adhesion molecule-1, vascular cell adhesion molecule-1, and monocyte chemoattractant protein-1 expression by 50% to 60%, and increased the extent of graft endothelialization. In vitro, TNF promoted chemokine expression and [3H]thymidine incorporation in vascular smooth muscle cells (SMCs) from WT, but not from p55-/- mice. However, responses of WT and p55-/- SMCs to other growth factors were equivalent. CONCLUSIONS: Signaling via p55, in vein graft-intrinsic cells, contributes to the pathogenesis of vein graft neointimal hyperplasia.


Assuntos
Hiperplasia/genética , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Transdução de Sinais/fisiologia , Túnica Íntima/patologia , Veia Cava Inferior/transplante , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Artéria Carótida Primitiva/cirurgia , Moléculas de Adesão Celular/biossíntese , Linhagem Celular , Quimiocina CCL2/biossíntese , Quimiocinas/biossíntese , Ativação Enzimática/fisiologia , Hiperplasia/enzimologia , Hiperplasia/patologia , Masculino , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/enzimologia , Neovascularização Patológica/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/deficiência , Receptores Tipo I de Fatores de Necrose Tumoral/fisiologia , Fator de Necrose Tumoral alfa/biossíntese , Túnica Íntima/química , Túnica Íntima/enzimologia , Túnica Íntima/metabolismo , Veias/enzimologia , Veias/metabolismo , Veias/transplante , Veia Cava Inferior/citologia
17.
Future Cardiol ; 11(3): 309-21, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-26021637

RESUMO

Over the past decade, basic and translational research delivered comprehensive evidence for the relevance of the Ca(2+)-binding protein S100A1 in cardiovascular diseases. Aberrant expression levels of S100A1 surfaced as molecular key defects, driving the pathogenesis of chronic heart failure, arterial and pulmonary hypertension, peripheral artery disease and disturbed myocardial infarction healing. Loss of intracellular S100A1 renders entire Ca(2+)-controlled networks dysfunctional, thereby leading to cardiomyocyte failure and endothelial dysfunction. Lack of S100A1 release in ischemic myocardium compromises cardiac fibroblast function, entailing impaired damage healing. This review focuses on molecular pathways and signaling cascades regulated by S100A1 in cardiomyocytes, endothelial cells and cardiac fibroblasts in order to provide an overview of our current mechanistic understanding of S100A1's action in cardiovascular pathophysiology.


Assuntos
Doenças Cardiovasculares/metabolismo , Células Endoteliais/patologia , Miócitos Cardíacos/patologia , Proteínas S100/metabolismo , Doenças Cardiovasculares/patologia , Células Endoteliais/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Miócitos Cardíacos/metabolismo
18.
FEBS Lett ; 588(6): 906-14, 2014 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-24486907

RESUMO

Exposure to pro-inflammatory cytokines, such as Angiotensin II, endothelin-1 or TNF leads to endothelial dysfunction, characterized by the reduced production of nitric oxide via endothelial nitric oxide synthase (eNOS). We recently identified the Ca(2+) binding protein S100A1 as an essential factor required for eNOS activity. Here we report that pro-inflammatory cytokines down-regulate expression of S100A1 in primary human microvascular endothelial cells (HMVECs) via induction of microRNA-138 (miR-138), in a manner that depends on the stabilization of HIF1-α. We show that loss of S100A1 in ECs reduces stimulus-induced NO production, which can be prevented by inhibition of miR-138. Our study suggests that targeting miR-138 might be beneficial for the treatment of cardiovascular disease.


Assuntos
Células Endoteliais/metabolismo , MicroRNAs/genética , Ativação Transcricional , Fator de Necrose Tumoral alfa/fisiologia , Angiotensina II/fisiologia , Doenças Cardiovasculares/imunologia , Doenças Cardiovasculares/metabolismo , Linhagem Celular , Endotelina-1/fisiologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Mediadores da Inflamação/fisiologia , Sistema de Sinalização das MAP Quinases , MicroRNAs/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas S100/genética , Proteínas S100/metabolismo
19.
EMBO Mol Med ; 6(6): 778-94, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24833748

RESUMO

Members of the S100 protein family have been reported to function as endogenous danger signals (alarmins) playing an active role in tissue inflammation and repair when released from necrotic cells. Here, we investigated the role of S100A1, the S100 isoform with highest abundance in cardiomyocytes, when released from damaged cardiomyocytes during myocardial infarction (MI). Patients with acute MI showed significantly increased S100A1 serum levels. Experimental MI in mice induced comparable S100A1 release. S100A1 internalization was observed in cardiac fibroblasts (CFs) adjacent to damaged cardiomyocytes. In vitro analyses revealed exclusive S100A1 endocytosis by CFs, followed by Toll-like receptor 4 (TLR4)-dependent activation of MAP kinases and NF-κB. CFs exposed to S100A1 assumed an immunomodulatory and anti-fibrotic phenotype characterized i.e. by enhanced intercellular adhesion molecule-1 (ICAM1) and decreased collagen levels. In mice, intracardiac S100A1 injection recapitulated these transcriptional changes. Moreover, antibody-mediated neutralization of S100A1 enlarged infarct size and worsened left ventricular functional performance post-MI. Our study demonstrates alarmin properties for S100A1 from necrotic cardiomyocytes. However, the potentially beneficial role of extracellular S100A1 in MI-related inflammation and repair warrants further investigation.


Assuntos
Infarto do Miocárdio/patologia , Miocárdio/patologia , Miócitos Cardíacos/patologia , Proteínas S100/sangue , Receptor 4 Toll-Like/imunologia , Animais , Endocitose , Fibroblastos/imunologia , Fibroblastos/patologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Quinases Ativadas por Mitógeno/imunologia , Infarto do Miocárdio/sangue , Infarto do Miocárdio/imunologia , Miocárdio/citologia , Miocárdio/imunologia , Miócitos Cardíacos/imunologia , NF-kappa B/imunologia , Proteínas S100/imunologia , Transdução de Sinais
20.
PLoS One ; 8(11): e78684, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24244340

RESUMO

The Ca(2+) sensor S100A1 is essential for proper endothelial cell (EC) nitric oxide (NO) synthase (eNOS) activation. S100A1 levels are greatly reduced in primary human microvascular ECs subjected to hypoxia, rendering them dysfunctional. However mechanisms that regulate S100A1 levels in ECs are unknown. Here we show that ECs transfected with a S100A1-3' untranslated region (UTR) luciferase reporter construct display significantly reduced gene expression when subjected to low oxygen levels or chemical hypoxia. Bioinformatic analysis suggested that microRNA -138 (MiR-138) could target the 3'UTR of S100A1. Patients with critical limb ischemia (CLI) or mice subjected to femoral artery resection (FAR) displayed increased MiR-138 levels and decreased S100A1 protein expression. Consistent with this finding, hypoxia greatly increased MiR-138 levels in ECs, but not in skeletal muscle C2C12 myoblasts or differentiated myotubes or primary human vascular smooth muscle cells. Transfection of a MiR-138 mimic into ECs reduced S100A1-3 'UTR reporter gene expression, while transfection of an anti MiR-138 prevented the hypoxia-induced downregulation of the reporter gene. Deletion of the 22 nucleotide putative MiR-138 target site abolished the hypoxia-induced loss of reporter gene expression. Knockdown of Hif1-α mediated by siRNA prevented loss of hypoxia-induced reporter gene expression. Conversely, specific activation of Hif1-α by a selective prolyl-hydroxylase inhibitor (IOX2) reduced reporter gene expression even in the absence of hypoxia. Finally, primary ECs transfected with a MiR-138 mimic displayed reduced tube formation when plated onto Matrigel matrix and expressed less NO when stimulated with VEGF. These effects were reversed by gene transfer of S100A1 using recombinant adenovirus. We conclude that hypoxia-induced MiR-138 is an essential mediator of EC dysfunction via its ability to target the 3'UTR of S100A1.


Assuntos
Células Endoteliais/metabolismo , Regulação da Expressão Gênica , Isquemia/metabolismo , MicroRNAs/metabolismo , Proteínas S100/biossíntese , Regiões 3' não Traduzidas , Animais , Hipóxia Celular , Linhagem Celular , Células Endoteliais/patologia , Feminino , Humanos , Isquemia/genética , Isquemia/patologia , Masculino , Camundongos , MicroRNAs/genética , Proteínas S100/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa