Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biomed Pharmacother ; 143: 112194, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34563949

RESUMO

Cardiac pressure overload is a crucial risk factor for cardiac hypertrophy and heart failure. Our previous study showed that depletion of the ß3-adrenergic receptor (ADRB3) induced left ventricular diastolic dysfunction via potential regulation of energy metabolism and cardiac contraction. However, the effects of ADRB3 on pressure overload-induced heart failure remain unclear. In the present study, systemic ADRB3-knockout mice suffering from transverse aortic constriction (TAC) surgery were used to identify the effects of ADRB3 on pressure overload-induced heart failure. Compared to wild-type mice, ADRB3 depletion significantly improved the left ventricular ejection fraction, reduced left ventricular posterior wall thickness and interventricular septum thickness, and decreased the area of cardiomyocytes after TAC. RNA sequencing and bioinformatics analysis showed that ADRB3 depletion up-regulated 275 mRNAs and down-regulated 105 mRNAs in mice suffering TAC surgery. GO analysis, GO-tree analysis, and GSEA showed that ADRB3 depletion mainly enhanced the innate immune response of hearts in cardiac pressure overload mice. In addition, pathway analysis and Pathway-Act analysis presented that innate immune response-related pathways, including RIG-I-like receptor signaling pathway, antigen processing and presentation, Toll-like receptor signaling pathway, and cell adhesion molecules, were significantly enriched in ADRB3-KO-TAC mice. Ten hub genes were identified using protein-protein interaction network, MCODE, and cytoHubba analysis. Furthermore, the depletion and activation of ADRB3 validated the effects of ADRB3 on the innate immune response of hearts after TAC. In conclusion, ADRB3 depletion relieves pressure overload-induced cardiac hypertrophy and heart failure, and these effects could be explained by the enhancement of innate immune response.


Assuntos
Cardiomegalia/prevenção & controle , Insuficiência Cardíaca/prevenção & controle , Imunidade Inata , Miocárdio/metabolismo , Receptores Adrenérgicos beta 3/deficiência , Animais , Cardiomegalia/imunologia , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatologia , Modelos Animais de Doenças , Redes Reguladoras de Genes , Insuficiência Cardíaca/imunologia , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/fisiopatologia , Imunidade Inata/genética , Masculino , Camundongos Knockout , Miocárdio/imunologia , Mapas de Interação de Proteínas , Receptores Adrenérgicos beta 3/genética , Transdução de Sinais , Volume Sistólico , Transcriptoma , Função Ventricular Esquerda , Remodelação Ventricular
2.
Front Immunol ; 12: 790511, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34992607

RESUMO

Cardiac fibrosis, a pathological condition due to excessive extracellular matrix (ECM) deposition in the myocardium, is associated with nearly all forms of heart disease. The processes and mechanisms that regulate cardiac fibrosis are not fully understood. In response to cardiac injury, macrophages undergo marked phenotypic and functional changes and act as crucial regulators of myocardial fibrotic remodeling. Here we show that the mitogen-activated protein kinase (MAPK) phosphatase-5 (MKP-5) in macrophages is involved in pressure overload-induced cardiac fibrosis. Cardiac pressure overload resulting from transverse aortic constriction (TAC) leads to the upregulation of Mkp-5 gene expression in the heart. In mice lacking MKP-5, p38 MAPK and JNK were hyperactivated in the heart, and TAC-induced cardiac hypertrophy and myocardial fibrosis were attenuated. MKP-5 deficiency upregulated the expression of the ECM-degrading matrix metalloproteinase-9 (Mmp-9) in the Ly6Clow (M2-type) cardiac macrophage subset. Consistent with in vivo findings, MKP-5 deficiency promoted MMP-9 expression and activity of pro-fibrotic macrophages in response to IL-4 stimulation. Furthermore, using pharmacological inhibitors against p38 MAPK, JNK, and ERK, we demonstrated that MKP-5 suppresses MMP-9 expression through a combined effect of p38 MAPK/JNK/ERK, which subsequently contributes to the inhibition of ECM-degrading activity. Taken together, our study indicates that pressure overload induces MKP-5 expression and facilitates cardiac hypertrophy and fibrosis. MKP-5 deficiency attenuates cardiac fibrosis through MAPK-mediated regulation of MMP-9 expression in Ly6Clow cardiac macrophages.


Assuntos
Cardiomegalia/imunologia , Fosfatases de Especificidade Dupla/deficiência , Insuficiência Cardíaca/imunologia , Sistema de Sinalização das MAP Quinases/imunologia , Miocárdio/patologia , Animais , Pressão Sanguínea , Cardiomegalia/diagnóstico , Cardiomegalia/patologia , Células Cultivadas , Modelos Animais de Doenças , Fosfatases de Especificidade Dupla/genética , Ecocardiografia , Fibrose , Coração/diagnóstico por imagem , Insuficiência Cardíaca/patologia , Humanos , Interleucina-4/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Knockout , Fosforilação/imunologia , Cultura Primária de Células , Remodelação Ventricular/imunologia
3.
Physiol Rep ; 8(15): e14532, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32786064

RESUMO

Pulmonary arterial hypertension (PAH) is a syndrome diagnosed by increased mean pulmonary artery (PA) pressure and resistance and normal pulmonary capillary wedge pressure. PAH is characterized pathologically by distal pulmonary artery remodeling, increased pulmonary vascular resistance, and plexiform lesions (PLs). Right ventricular fibrosis and hypertrophy, leading to right ventricular failure, are the main determinants of mortality in PAH. Recent work suggests that right ventricular fibrosis results from resident cardiac fibroblast activation and conversion to myofibroblasts, leading to replacement of contractile cardiomyocytes with nondistensible tissue incapable of conductivity or contractility. However, the origins, triggers, and consequences of myofibroblast expansion and its pathophysiological relationship with PAH are unclear. Recent advances indicate that signals generated by adaptive and innate immune cells may play a role in right ventricular fibrosis and remodeling. This review summarizes recent insights into the mechanisms by which adaptive and innate immune signals participate in the transition of cardiac fibroblasts to activated myofibroblasts and highlights the existing gaps of knowledge as relates to the development of right ventricular fibrosis.


Assuntos
Imunidade Adaptativa , Cardiomegalia/imunologia , Hipertensão Pulmonar/complicações , Imunidade Inata , Animais , Cardiomegalia/etiologia , Cardiomegalia/patologia , Transdiferenciação Celular , Fibrose , Humanos , Macrófagos/imunologia , Macrófagos/patologia , Miofibroblastos/imunologia , Miofibroblastos/patologia
4.
Exp Physiol ; 105(3): 489-501, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31957919

RESUMO

NEW FINDINGS: What is the central question of this study? We questioned whether the disruption of invariant natural killer T (iNKT) cells exacerbates left ventricular (LV) remodelling and heart failure after transverse aortic constriction in mice. What are the main findings and their importance? Pressure overload induced by transverse aortic constriction increased the infiltration of iNKT cells in mouse hearts. The disruption of iNKT cells exacerbated LV remodelling and hastened the transition from hypertrophy to heart failure, in association with the activation of mitogen-activated protein kinase signalling. Activation of iNKT cells modulated the immunological balance in this process and played a protective role against LV remodelling and failure. ABSTRACT: Chronic inflammation is involved in the development of cardiac remodelling and heart failure (HF). Invariant natural killer T (iNKT) cells, a subset of T lymphocytes, have been shown to produce various cytokines and orchestrate tissue inflammation. The pathophysiological role of iNKT cells in HF caused by pressure overload has not been studied. In the present study, we investigated whether the disruption of iNKT cells affected this process in mice. Transverse aortic constriction (TAC) and a sham operation were performed in male C57BL/6J wild-type (WT) and iNKT cell-deficient Jα18 knockout (KO) mice. The infiltration of iNKT cells was increased after TAC. The disruption of iNKT cells exacerbated left ventricular (LV) remodelling and hastened the transition to HF after TAC. Histological examinations also revealed that the disruption of iNKT cells induced greater myocyte hypertrophy and a greater increase in interstitial fibrosis after TAC. The expressions of interleukin-10 and tumour necrosis factor-α mRNA and their ratio in the LV after TAC were decreased in the KO compared with WT mice, which might indicate that the disruption of iNKT cells leads to an imbalance between T-helper type 1 and type 2 cytokines. The phosphorylation of extracellular signal-regulated kinase was significantly increased in the KO mice. The disruption of iNKT cells exacerbated the development of cardiac remodelling and HF after TAC. The activation of iNKT cells might play a protective role against HF caused by pressure overload. Targeting the activation of iNKT cells might thus be a promising candidate as a new therapeutic strategy for HF.


Assuntos
Cardiomegalia/imunologia , Insuficiência Cardíaca/imunologia , Células T Matadoras Naturais/imunologia , Animais , Fibrose/imunologia , Ventrículos do Coração/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/imunologia , Miócitos Cardíacos/imunologia , Fosforilação/imunologia , Transdução de Sinais/imunologia , Remodelação Ventricular/imunologia
5.
Sci Rep ; 9(1): 6055, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30988323

RESUMO

Heart failure (HF) is the leading cause of death in the Western world. Pathophysiological processes underlying HF development, including cardiac hypertrophy, fibrosis and inflammation, are controlled by specific microRNAs (miRNAs). Whereas most studies investigate miRNA function in one particular cardiac cell type, their multicellular function is poorly investigated. The present study probed 194 miRNAs -differentially expressed in cardiac inflammatory disease - for regulating cardiomyocyte size, cardiac fibroblasts collagen content, and macrophage polarization. Of the tested miRNAs, 13%, 26%, and 41% modulated cardiomyocyte size, fibroblast collagen production, and macrophage polarization, respectively. Seventeen miRNAs affected all three cellular processes, including miRNAs with established (miR-210) and unknown roles in cardiac pathophysiology (miR-145-3p). These miRNAs with a multi-cellular function commonly target various genes. In-depth analysis in vitro of previously unstudied miRNAs revealed that the observed phenotypical alterations concurred with changes in transcript and protein levels of hypertrophy-, fibrosis- and inflammation-related genes. MiR-145-3p and miR-891a-3p were identified to regulate the fibrotic response, whereas miR-223-3p, miR-486-3p, and miR-488-5p modulated macrophage activation and polarisation. In conclusion, miRNAs are multi-cellular regulators of different cellular processes underlying cardiac disease. We identified previously undescribed roles of miRNAs in hypertrophy, fibrosis, and inflammation, and attribute new cellular effects to various well-known miRNAs.


Assuntos
Cardiomegalia/patologia , Insuficiência Cardíaca/genética , MicroRNAs/metabolismo , Miocardite/imunologia , Miocárdio/patologia , Animais , Animais Recém-Nascidos , Cardiomegalia/genética , Cardiomegalia/imunologia , Células Cultivadas , Fibroblastos , Fibrose , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Insuficiência Cardíaca/imunologia , Insuficiência Cardíaca/patologia , Humanos , Ativação de Macrófagos/genética , Ativação de Macrófagos/imunologia , Macrófagos , Camundongos , Miocardite/genética , Miocardite/patologia , Miocárdio/citologia , Miocárdio/imunologia , Miócitos Cardíacos , Cultura Primária de Células , Ratos
6.
Sci Rep ; 9(1): 6047, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30988334

RESUMO

Early adaptive cardiac hypertrophy (EACH) is initially a compensatory process to optimize pump function. We reported the emergence of Orai3 activity during EACH. This study aimed to characterize how inflammation regulates store-independent activation of Orai3-calcium influx and to evaluate the functional role of this influx. Isoproterenol infusion or abdominal aortic banding triggered EACH. TNFα or conditioned medium from cardiac CD11b/c cells activated either in vivo [isolated from rats displaying EACH], or in vitro [isolated from normal rats and activated with lipopolysaccharide], were added to adult cardiomyocytes before measuring calcium entry, cell hypertrophy and cell injury. Using intramyocardial injection of siRNA, Orai3 was in vivo knockdown during EACH to evaluate its protective activity in heart failure. Inflammatory CD11b/c cells trigger a store-independent calcium influx in hypertrophied cardiomyocytes, that is mimicked by TNFα. Pharmacological or molecular (siRNA) approaches demonstrate that this calcium influx, depends on TNFR2, is Orai3-driven, and elicits cardiomyocyte hypertrophy and resistance to oxidative stress. Neutralization of Orai3 inhibits protective GSK3ß phosphorylation, impairs EACH and accelerates heart failure. Orai3 exerts a pathophysiological protective impact in EACH promoting hypertrophy and resistance to oxidative stress. We highlight inflammation arising from CD11b/c cells as a potential trigger of TNFR2- and Orai3-dependent signaling pathways.


Assuntos
Canais de Cálcio/metabolismo , Cardiomegalia/imunologia , Insuficiência Cardíaca/imunologia , Miócitos Cardíacos/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Animais , Antígeno CD11b/metabolismo , Antígeno CD11c/metabolismo , Cálcio/metabolismo , Cardiomegalia/induzido quimicamente , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Meios de Cultivo Condicionados/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Técnicas de Silenciamento de Genes , Glicogênio Sintase Quinase 3 beta/metabolismo , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Humanos , Isoproterenol/toxicidade , Masculino , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/imunologia , Fosforilação/imunologia , RNA Interferente Pequeno/metabolismo , Ratos , Transdução de Sinais/imunologia , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo
7.
Biochem Biophys Res Commun ; 509(1): 16-23, 2019 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-30581005

RESUMO

Pathological cardiac hypertrophy is a leading cause of morbidity and mortality in the world. However, it is still unclear the molecular mechanism revealing the progression of the disease. In the study, we illustrated that the expression of leukocyte immunoglobulin-like receptor B4 (LILRB4), associated with the pathological development of various inflammatory diseases, was down-regulated in pressure overload-induced hearts of patients and mice. LILRB4-knockout mice developed cardiac hypertrophy and heart failure by promoting cardiac dysfunction, fibrosis, inflammation and apoptosis. Mechanistically, transforming growth factor ß1 (TGF-ß1) expression was significantly promoted by LILRB4 deficiency in hearts of mice after aortic banding (AB) surgery. AB-induced inflammation in cardiac tissues was accelerated by LILRB4 deletion through elevating nuclear factor κB (NF-κB) signaling pathway. Furthermore, apoptosis triggered by AB operation in heart tissues was markedly enhanced in LILRB4-KO mice through promoting Caspase-3 activation. Importantly, the in vitro study indicated that LILRB4 knockdown-promoted fibrosis; inflammation and apoptosis were largely via the NF-κB signaling. Therefore, the findings above identified LILRB4 might be a negative regulator of cardiac remodeling, illustrating that LILRB4 represented as a therapeutic target for the prevention of cardiac hypertrophy and heart failure.


Assuntos
Cardiomegalia/patologia , Inflamação/patologia , Glicoproteínas de Membrana/imunologia , Miocárdio/patologia , NF-kappa B/imunologia , Receptores de Superfície Celular/imunologia , Receptores Imunológicos/imunologia , Animais , Apoptose , Cardiomegalia/genética , Cardiomegalia/imunologia , Células Cultivadas , Regulação para Baixo , Fibrose , Humanos , Inflamação/genética , Inflamação/imunologia , Masculino , Glicoproteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/metabolismo , Receptores de Superfície Celular/genética , Receptores Imunológicos/genética
8.
Proc Natl Acad Sci U S A ; 115(20): E4661-E4669, 2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29712858

RESUMO

Nonischemic cardiomyopathy (NICM) resulting from long-standing hypertension, valvular disease, and genetic mutations is a major cause of heart failure worldwide. Recent observations suggest that myeloid cells can impact cardiac function, but the role of tissue-intrinsic vs. tissue-extrinsic myeloid cells in NICM remains poorly understood. Here, we show that cardiac resident macrophage proliferation occurs within the first week following pressure overload hypertrophy (POH; a model of heart failure) and is requisite for the heart's adaptive response. Mechanistically, we identify Kruppel-like factor 4 (KLF4) as a key transcription factor that regulates cardiac resident macrophage proliferation and angiogenic activities. Finally, we show that blood-borne macrophages recruited in late-phase POH are detrimental, and that blockade of their infiltration improves myocardial angiogenesis and preserves cardiac function. These observations demonstrate previously unappreciated temporal and spatial roles for resident and nonresident macrophages in the development of heart failure.


Assuntos
Cardiomegalia/patologia , Cardiomiopatias/patologia , Insuficiência Cardíaca/patologia , Fatores de Transcrição Kruppel-Like/metabolismo , Macrófagos/patologia , Miocárdio/patologia , Animais , Cardiomegalia/imunologia , Cardiomegalia/metabolismo , Cardiomiopatias/imunologia , Cardiomiopatias/metabolismo , Células Cultivadas , Insuficiência Cardíaca/imunologia , Insuficiência Cardíaca/metabolismo , Fator 4 Semelhante a Kruppel , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Miocárdio/imunologia , Miocárdio/metabolismo , Pressão
9.
Front Immunol ; 9: 3029, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30619368

RESUMO

The transcription factor STAT3 has a protective function in the heart. Until recently, the role of STAT3 in hypertension-induced cardiac hypertrophy was unsettled. Earlier studies revealed that global reduction of STAT3 activity reduced cardiac hypertrophy with hypertension, but caused a disruption of myofilaments and increased contractile dysfunction. However, newer studies with cardiomyocyte-specific deletion of STAT3 indicate that STAT3 does not cause cardiac hypertrophy with increased blood pressure. Rather, cardiac STAT3 is important for maintaining metabolic homeostasis, and loss of STAT3 in cardiomyocytes makes the heart more susceptible to chronic pathological insult, for example by disrupting glucose metabolism and protective signaling networks via the upregulation of certain microRNAs. This scenario has implications for understanding peripartum cardiomyopathy as well. In viral myocarditis, STAT3 opposes the initiation of the dilated phenotype by maintaining membrane integrity via the expression of dystrophin. STAT3 signaling was also found to attenuate myocarditis by polarizing macrophages to a less inflammatory phenotype. On the other hand, STAT3 contributes to immune-mediated myocarditis due to IL-6-induced complement component C3 production in the liver, as well as the differentiation of Th17 cells, which play a role in initiation and development of myocarditis. Besides canonical signaling pathways, unphosphorylated STAT3 (U-STAT3) and redox-activated STAT3 have been shown to couple to transcription in the heart. In addition, tissue signaling cytokines such as IL-22 and IL-17 have been proposed to have actions on the heart that involve STAT3, but are not fully defined. Understanding the novel and often protective aspects of STAT3 in the myocardium could lead to new therapeutic approaches to treat heart disease.


Assuntos
Cardiomegalia/imunologia , Coração/fisiologia , Miocardite/imunologia , Miocárdio/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Cardiomegalia/fisiopatologia , Complemento C3/imunologia , Modelos Animais de Doenças , Humanos , Interleucina-17/imunologia , Interleucina-17/metabolismo , Interleucina-6/imunologia , Interleucina-6/metabolismo , Interleucinas/imunologia , Interleucinas/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Miocardite/fisiopatologia , Miocárdio/citologia , Miocárdio/imunologia , Miócitos Cardíacos/imunologia , Miócitos Cardíacos/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/imunologia , Transdução de Sinais/imunologia , Células Th17/imunologia , Células Th17/metabolismo , Interleucina 22
10.
Basic Res Cardiol ; 112(3): 25, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28349258

RESUMO

Inflammatory responses play an important role in the development of left ventricular (LV) hypertrophy and dysfunction. Recent studies demonstrated that increased T-cell infiltration and T-cell activation contribute to LV hypertrophy and dysfunction. Dendritic cells (DCs) are professional antigen-presenting cells that orchestrate immune responses, especially by modulating T-cell function. In this study, we investigated the role of bone marrow-derived CD11c+ DCs in transverse aortic constriction (TAC)-induced LV fibrosis and hypertrophy in mice. We observed that TAC increased the number of CD11c+ cells and the percentage of CD11c+ MHCII+ (major histocompatibility complex class II molecule positive) DCs in the LV, spleen and peripheral blood in mice. Using bone marrow chimeras and an inducible CD11c+ DC ablation model, we found that depletion of bone marrow-derived CD11c+ DCs significantly attenuated LV fibrosis and hypertrophy in mice exposed to 24 weeks of moderate TAC. CD11c+ DC ablation significantly reduced TAC-induced myocardial inflammation as indicated by reduced myocardial CD45+ cells, CD11b+ cells, CD8+ T cells and activated effector CD8+CD44+ T cells in LV tissues. Moreover, pulsing of autologous DCs with LV homogenates from TAC mice promoted T-cell proliferation. These data indicate that bone marrow-derived CD11c+ DCs play a maladaptive role in hemodynamic overload-induced cardiac inflammation, hypertrophy and fibrosis through the presentation of cardiac self-antigens to T cells.


Assuntos
Células Dendríticas/imunologia , Hipertrofia Ventricular Esquerda/imunologia , Ativação Linfocitária/imunologia , Remodelação Ventricular/imunologia , Animais , Apresentação de Antígeno/imunologia , Células da Medula Óssea/imunologia , Antígeno CD11c/imunologia , Linfócitos T CD8-Positivos/imunologia , Cardiomegalia/imunologia , Modelos Animais de Doenças , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos C57BL , Miocardite/imunologia
11.
Endocrinology ; 157(1): 336-45, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26469138

RESUMO

IGF-1 receptor (IGF-1R) signaling is implicated in cardiac hypertrophy and longevity. However, the role of IGF-1R in age-related cardiac remodeling is only partially understood. We therefore sought to determine whether the deletion of the IGF-1R in cardiomyocytes might delay the development of aging-associated myocardial pathologies by examining 2-year-old male cardiomyocyte-specific IGF-1R knockout (CIGF1RKO) mice. Aging was associated with the induction of IGF-1R expression in hearts. Cardiomyocytes hypertrophied with age in wild-type (WT) mice. In contrast, the cardiac hypertrophic response associated with aging was blunted in CIGF1RKO mice. Concomitantly, fibrosis was reduced in aged CIGF1RKO compared with aged WT hearts. Expression of proinflammatory cytokines such as IL-1α, IL-1ß, IL-6, and receptor activator of nuclear factor-κB ligand was increased in aged WT hearts, but this increase was attenuated in aged CIGF1RKO hearts. Phosphorylation of Akt was increased in aged WT, but not in aged CIGF1RKO, hearts. In cultured cardiomyocytes, IGF-1 induced senescence as demonstrated by increased senescence-associated ß-galactosidase staining, and a phosphoinositide 3-kinase inhibitor inhibited this effect. Furthermore, inhibition of phosphoinositide 3-kinase significantly prevented the increase in IL-1α, IL-1ß, receptor activator of nuclear factor-κB ligand, and p21 protein expression by IGF-1. These data reveal an essential role for the IGF-1-IGF-1R-Akt pathway in mediating cardiomyocyte senescence.


Assuntos
Envelhecimento , Cardiomegalia/metabolismo , Ventrículos do Coração/metabolismo , Miócitos Cardíacos/metabolismo , Receptor IGF Tipo 1/metabolismo , Remodelação Ventricular , Animais , Biomarcadores/metabolismo , Cardiomegalia/imunologia , Cardiomegalia/patologia , Cardiomegalia/prevenção & controle , Células Cultivadas , Senescência Celular/efeitos dos fármacos , Citocinas/antagonistas & inibidores , Citocinas/genética , Citocinas/metabolismo , Inibidores Enzimáticos/farmacologia , Fibrose , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/imunologia , Ventrículos do Coração/patologia , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/imunologia , Miócitos Cardíacos/patologia , Fosfatidilinositol 3-Quinase/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor IGF Tipo 1/agonistas , Receptor IGF Tipo 1/genética , Transdução de Sinais/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos
12.
Toxicol Appl Pharmacol ; 284(3): 315-22, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25759242

RESUMO

We hypothesized that chronic inhibition of NF-κB activity in the hypothalamic paraventricular nucleus (PVN) delays the progression of hypertension and attenuates cardiac hypertrophy by up-regulating anti-inflammatory cytokines, reducing pro-inflammatory cytokines (PICs), attenuating nuclear factor-κB (NF-κB) p65 and NAD(P)H oxidase in the PVN of young spontaneously hypertensive rats (SHR). Young normotensive Wistar-Kyoto (WKY) and SHR rats received bilateral PVN infusions with NF-κB inhibitor pyrrolidine dithiocarbamate (PDTC) or vehicle for 4 weeks. SHR rats had higher mean arterial pressure and cardiac hypertrophy as indicated by increased whole heart weight/body weight ratio, whole heart weight/tibia length ratio, left ventricular weight/tibia length ratio, cardiomyocyte diameters of the left cardiac ventricle, and mRNA expressions of cardiac atrial natriuretic peptide (ANP) and beta-myosin heavy chain (ß-MHC). These SHR rats had higher PVN levels of proinflammatory cytokines (PICs), reactive oxygen species (ROS), the chemokine monocyte chemoattractant protein-1 (MCP-1), NAD(P)H oxidase activity, mRNA expression of NOX-2 and NOX-4, and lower PVN IL-10, and higher plasma levels of PICs and NE, and lower plasma IL-10. PVN infusion of NF-κB inhibitor PDTC attenuated all these changes. These findings suggest that NF-κB activation in the PVN increases sympathoexcitation and hypertensive response, which are associated with the increases of PICs and oxidative stress in the PVN; PVN inhibition of NF-κB activity attenuates PICs and oxidative stress in the PVN, thereby attenuates hypertension and cardiac hypertrophy.


Assuntos
Cardiomegalia/prevenção & controle , Citocinas/metabolismo , Hipertensão/tratamento farmacológico , Mediadores da Inflamação/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Pirrolidinas/farmacologia , Tiocarbamatos/farmacologia , Fator de Transcrição RelA/antagonistas & inibidores , Animais , Pressão Arterial/efeitos dos fármacos , Biomarcadores/metabolismo , Cardiomegalia/etiologia , Cardiomegalia/imunologia , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatologia , Modelos Animais de Doenças , Hipertensão/complicações , Hipertensão/imunologia , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Núcleo Hipotalâmico Paraventricular/imunologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Transdução de Sinais/efeitos dos fármacos , Sistema Nervoso Simpático/efeitos dos fármacos , Sistema Nervoso Simpático/metabolismo , Sistema Nervoso Simpático/fisiopatologia , Fatores de Tempo , Fator de Transcrição RelA/metabolismo
13.
Endocrinology ; 156(4): 1577-89, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25562617

RESUMO

A transient model for human Graves' disease was successfully established in mice using up to 3 immunizations with recombinant adenovirus expressing the extracellular A-subunit of the human TSH receptor (TSHR) (Ad-TSHR). We studied extension of adenovirally induced TSHR A-subunit immunization in mice by using a novel protocol of long-term 3- and 4-weekly injections. Generation of TSHR binding stimulatory antibodies (capacity to stimulate cAMP activity in TSHR-expressing test cells), goiter, and histological thyroid alterations were maintained for at least 9 months in all Ad-TSHR-immunized mice. In response to injection of 10(10) plaque-forming units of Ad-TSHR, also elevated mean serum T4 levels were observed throughout the study. Moreover, cardiac organ involvement (tachycardia and hypertrophy) were consistently observed in these mice. Higher doses of Ad-TSHR (10(11) plaque-forming units) did not produce consistent elevation of T4 and were not associated with a clear increase in heart rate vs controls, probably because these high doses provoked an immune response-induced tachycardia on their own. In summary, a long-term model of Graves' disease induced by a relatively simple protocol of continuing monthly immunizations should allow to investigate long-term disease mechanisms and may possibly obviate the need for more complicated disease models. Moreover, the clinical outcome predictor of tachycardia and cardiac involvement was reliably detected in the model.


Assuntos
Cardiomegalia/etiologia , Doença de Graves/etiologia , Receptores da Tireotropina/imunologia , Taquicardia/etiologia , Adenoviridae , Animais , Autoanticorpos , Cardiomegalia/imunologia , Modelos Animais de Doenças , Feminino , Doença de Graves/imunologia , Imunização , Camundongos , Taquicardia/imunologia , Tiroxina/sangue
14.
PLoS One ; 9(11): e112710, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25415601

RESUMO

Cardiac tissue remodeling in the course of chronic left ventricular hypertrophy requires phagocytes which degrade cellular debris, initiate and maintain tissue inflammation and reorganization. The dynamics of phagocytes in left ventricular hypertrophy have not been systematically studied. Here, we characterized the temporal accumulation of leukocytes in the cardiac immune response by flow cytometry and fluorescence microscopy at day 3, 6 and 21 following transverse aortic constriction (TAC). Cardiac hypertrophy due to chronic pressure overload causes cardiac immune response and inflammation represented by an increase of immune cells at all three time points among which neutrophils reached their maximum at day 3 and macrophages at day 6. The cardiac macrophage population consisted of both Ly6C(low) and Ly6C(high) macrophages. Ly6C(low) macrophages were more abundant peaking at day 6 in response to pressure overload. During the development of cardiac hypertrophy the expression pattern of adhesion molecules was investigated by qRT-PCR and flow cytometry. CD11b, CX3CR1 and ICAM-1 determined by qRT-PCR in whole cardiac tissue were up-regulated in response to pressure overload at day 3 and 6. CD11b and CX3CR1 were significantly increased by TAC on the surface of Ly6C(low) but not on Ly6C(high) macrophages. Furthermore, ICAM-1 was up-regulated on cardiac endothelial cells. In fluorescence microscopy Ly6C(low) macrophages could be observed attached to the intra- and extra-vascular vessel-wall. Taken together, TAC induced the expression of adhesion molecules, which may explain the accumulation of Ly6C(low) macrophages in the cardiac tissue, where these cells might contribute to cardiac inflammation and remodeling in response to pressure overload.


Assuntos
Cardiomegalia/imunologia , Macrófagos/imunologia , Animais , Cardiomegalia/fisiopatologia , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Pressão , Reação em Cadeia da Polimerase em Tempo Real
15.
Cardiovasc Res ; 96(3): 422-32, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22977006

RESUMO

AIMS: Inflammation and Toll-like receptor (TLR) signalling have been linked to the development of cardiac hypertrophy following transverse aortic constriction (TAC). In the present study, we investigated whether pre-treatment with the synthetic TLR9 ligands 1668-thioate or 1612-thioate modulates the progression of TAC-induced cardiac inflammation and hypertrophy. METHODS AND RESULTS: C57BL/6N-mice were pre-treated with 1668-thioate, 1612-thioate (0.25 nmol/g, i.p.), or phosphate-buffered saline 16 h prior to TAC or sham surgery. Heart-weight/body-weight ratio (HW/BW), cardiomyocyte cell size, cellular macrophage accumulation, myofibroblast differentiation, and collagen deposition were investigated for up to 28 days. Cardiac function was monitored using a pressure-volume catheter and M-mode echocardiography. Inflammatory gene expression in the heart was analysed via gene array, while the time course of mRNA expression of key inflammatory mediators was assessed via RT-qPCR. TAC increased the HW/BW ratio and cardiomyocyte cell size and induced macrophage accumulation, myofibroblast differentiation, and collagen deposition. These changes were accompanied by cardiac inflammation and a significant loss of left ventricular function. Pre-treatment with cytosine-phosphate-guanine (CpG)-containing 1668-thioate attenuated the inflammatory response, the progression of cardiac hypertrophy, and cardiac remodelling, which resulted in a prolonged preservation of left ventricular function. These changes were induced to a smaller extent by the use of the non-CG-containing oligodeoxynucleotide 1612-thioate. CONCLUSION: Pre-treatment with 1668-thioate attenuated cardiac hypertrophy following pressure overload, possibly by modifying the hypertrophy-induced inflammatory response, thereby reducing cardiac growth and fibrosis as well as delaying loss of cardiac function.


Assuntos
Cardiomegalia/prevenção & controle , Cardiotônicos/farmacologia , Miocardite/prevenção & controle , Miocárdio/imunologia , Oligodesoxirribonucleotídeos/farmacologia , Receptor Toll-Like 9/agonistas , Animais , Cateterismo Cardíaco , Cardiomegalia/diagnóstico por imagem , Cardiomegalia/genética , Cardiomegalia/imunologia , Cardiomegalia/metabolismo , Cardiotônicos/síntese química , Quimiocina CCL2/metabolismo , Quimiocina CCL4/metabolismo , Colágeno/metabolismo , Modelos Animais de Doenças , Fibrose , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica , Insuficiência Cardíaca/imunologia , Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/prevenção & controle , Mediadores da Inflamação/metabolismo , Ligantes , Ativação de Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miocardite/diagnóstico por imagem , Miocardite/genética , Miocardite/imunologia , Miocardite/metabolismo , Miocárdio/metabolismo , Miocárdio/patologia , Oligodesoxirribonucleotídeos/síntese química , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Tempo , Receptor Toll-Like 9/metabolismo , Ultrassonografia , Função Ventricular Esquerda/efeitos dos fármacos , Pressão Ventricular/efeitos dos fármacos
16.
J Mol Cell Cardiol ; 52(5): 1135-44, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22326848

RESUMO

The toll-like receptors (TLR) and myocardial infarction (MI) promote NF-κB-dependent inflammatory transcription and oxidative injury in myocardium. The multifunctional Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) is activated by oxidation and contributes to NF-κB-dependent transcription, myocardial hypertrophy and post-MI death. The myeloid differentiation protein 88 (MyD88) is an adapter protein critical for many TLR functions, but downstream targets for TLR/MyD88 signaling in MI are not well understood. We asked if CaMKII and TLR/MyD88 pathways are interconnected and if TLR/MyD88 contributes to adverse outcomes after MI. Here we show that TLR-4 activation by lipopolysaccharide (LPS) induces CaMKII oxidation (ox-CaMKII) in cardiomyocytes. MI enhances ox-CaMKII in wild type (WT) hearts but not in MyD88(-/-) hearts that are defective in MyD88-dependent TLR signaling. In post-MI WT hearts expression of pro-inflammatory genes TNF-α (Tnfa), complement factor B (Cfb), myocyte death and fibrosis were significantly increased, but increases were significantly less in MyD88(-/-) hearts after MI. MyD88(-/-) cardiomyocytes were defective in NF-κB activation by LPS but not by the MyD88-independent TLR agonist poly(I:C). In contrast, TNF-α induced Cfb gene expression was not deficient in MyD88(-/-) cardiomyocytes. Several hypertrophy marker genes were upregulated in both WT and MyD88(-/-) hearts after MI, but Acta1 was significantly attenuated in MyD88(-/-) hearts, suggesting that MyD88 selectively affects expression of hypertrophic genes. Post-MI cardiac hypertrophy, inflammation, apoptosis, ox-CaMKII expression and mortality were significantly reduced in MyD88(-/-) compared to WT littermates. These data suggest that MyD88 contributes to CaMKII oxidation and is important for adverse hypertrophic and inflammatory responses to LPS and MI.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Cardiomegalia/metabolismo , Fator 88 de Diferenciação Mieloide/fisiologia , Infarto do Miocárdio/metabolismo , Transdução de Sinais , Animais , Apoptose , Cardiomegalia/imunologia , Cardiomegalia/patologia , Células Cultivadas , Fator B do Complemento/genética , Fator B do Complemento/metabolismo , Ativação Enzimática , Feminino , Fibrose , Regulação da Expressão Gênica , Inflamação/genética , Inflamação/metabolismo , Estimativa de Kaplan-Meier , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/genética , Infarto do Miocárdio/imunologia , Infarto do Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , NF-kappa B/metabolismo , Infiltração de Neutrófilos , Oxirredução , Receptores Toll-Like/metabolismo , Transcrição Gênica , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
17.
Cardiovasc Res ; 94(1): 105-14, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22298642

RESUMO

AIMS: Although increased levels of myocardial receptor activator of nuclear factor (NF)-κB ligand (RANKL) have been reported in heart failure, the role of this pathway in mediating activation of inflammatory pathways during myocardial remodelling is less well understood. This study sought to determine the role of myocardial RANKL in regulating cytokine expression. METHODS AND RESULTS: A marked increase in RANKL expression occurred as early as 6h following transverse aortic constriction (TAC) in mouse hearts and persisted at 3 and 17 days. An increase in tumour necrosis factor-α (TNF-α), interleukin (IL)-1α, and IL-1ß was observed in the hypertrophied hearts only at 3 or 17 days after TAC. Treatment with losartan significantly attenuated TAC-induced cardiac hypertrophy, in parallel with decreased expression of RANKL, TNF-α, IL-1α, and IL-1ß. Furthermore, injection of a RANKL-neutralizing monoclonal antibody attenuated RANKL-induced cytokine expression. RANKL stimulated expression of TNF-α, IL-1α, and IL-1ß in neonatal rat cardiomyocytes via activation of NF-κB. RANKL-induced NF-κB activation and expression of these cytokines were both attenuated when RANK, receptor for RANKL, or TRAF2 or TRAF6, adaptors for RANK, was silenced by siRNA. Furthermore, inhibitors of phospholipase C (PLC), protein kinase C (PKC), and inhibitor of κB kinase also significantly inhibited RANKL-induced cellular activities, but inhibitors of phosphatidylinositol 3-kinase, extracellular signal-regulated kinase, or p38 mitogen-activated protein kinase were without effect. CONCLUSION: Our data demonstrate for the first time that the pressure-overloaded myocardium generates RANKL, which induces TNF-α, IL-1α, and IL-1ß production via a RANK-TRAF2/TRAF6-PLC-PKC-NF-κB-mediated autocrine mechanism.


Assuntos
Cardiomegalia/imunologia , Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Miocardite/imunologia , Miócitos Cardíacos/imunologia , Ligante RANK/metabolismo , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Animais Recém-Nascidos , Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/farmacologia , Comunicação Autócrina , Cardiomegalia/genética , Cardiomegalia/prevenção & controle , Células Cultivadas , Citocinas/genética , Modelos Animais de Doenças , Interleucina-1alfa/genética , Interleucina-1alfa/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Losartan/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miocardite/genética , Miocardite/prevenção & controle , Ligante RANK/antagonistas & inibidores , Interferência de RNA , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor Ativador de Fator Nuclear kappa-B/genética , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Transdução de Sinais , Fator 2 Associado a Receptor de TNF/genética , Fator 2 Associado a Receptor de TNF/metabolismo , Fator 6 Associado a Receptor de TNF/genética , Fator 6 Associado a Receptor de TNF/metabolismo , Fatores de Tempo , Transfecção , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima
18.
Clin Hemorheol Microcirc ; 51(3): 177-91, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22240383

RESUMO

QiShen YiQi Pills(®) (QSYQ) is a compound Chinese medicine used for treatment of cardiovascular diseases. However, the potential of QSYQ to inhibit cardiac fibrosis in left ventricle hypertrophy is not explored to date. We investigated the effects of post-treatment with QSYQ on rat myocardial fibrosis in left ventricle hypertrophy induced by pressure over-load through ascending aortic stenosis. QSYQ was administrated 4 weeks after the surgery, at a dose of 0.8 g/kg/day over the next 4 weeks, while echocardiography was performed 4 and 8 weeks, respectively, after the surgery. Eight weeks after the surgery, myocardial blood flow was determined by Laser-Doppler Perfusion Imager and the ratio of heart weight to body weight (HW/BW) was estimated, in concurrent evaluation of myocardial histology and ultrastructure, as well as collagen content by sirius red staining, and immunohistochemistry staining for CD68 and transforming growth factor beta 1. Post-treatment with QSYQ significantly alleviated left ventricular posterior wall end diastolic thickness and the HW/BW, increased left ventricle ejection fraction and left ventricle fractional shortening. QSYQ also decreased myocardial fibrosis size. The expression of CD68 and transforming growth factor beta 1 were obviously suppressed after QSYQ treatment. The results suggest that post-treatment with QSYQ attenuates pressure over-load-induced cardiac hypertrophy and myocardial fibrosis through interfering in inflammatory process.


Assuntos
Cardiomegalia/tratamento farmacológico , Cardiomegalia/patologia , Medicamentos de Ervas Chinesas/uso terapêutico , Coração/efeitos dos fármacos , Miocárdio/patologia , Animais , Antígenos CD/análise , Antígenos CD/imunologia , Antígenos de Diferenciação Mielomonocítica/análise , Antígenos de Diferenciação Mielomonocítica/imunologia , Cardiomegalia/imunologia , Ecocardiografia/efeitos dos fármacos , Fibrose , Masculino , Miocárdio/imunologia , Ratos , Ratos Sprague-Dawley , Fator de Crescimento Transformador beta1/análise , Fator de Crescimento Transformador beta1/imunologia
19.
Arterioscler Thromb Vasc Biol ; 31(5): 1116-23, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21330604

RESUMO

OBJECTIVE: Cyclophilin A (CyPA, encoded by Ppia) is a proinflammatory protein secreted in response to oxidative stress in mice and humans. We recently demonstrated that CyPA increased angiotensin II (Ang II)-induced reactive oxygen species (ROS) production in the aortas of apolipoprotein E (Apoe)-/- mice. In this study, we sought to evaluate the role of CyPA in Ang II-induced cardiac hypertrophy. METHODS AND RESULTS: Cardiac hypertrophy was not significantly different between Ppia+/+ and Ppia-/- mice infused with Ang II (1000 ng/min per kg for 4 weeks). Therefore, we investigated the effect of CyPA under conditions of high ROS and inflammation using the Apoe-/- mice. In contrast to Apoe-/- mice, Apoe-/-Ppia-/- mice exhibited significantly less Ang II-induced cardiac hypertrophy. Bone marrow cell transplantation showed that CyPA in cells intrinsic to the heart plays an important role in the cardiac hypertrophic response. Ang II-induced ROS production, cardiac fibroblast proliferation, and cardiac fibroblast migration were markedly decreased in Apoe-/-Ppia-/- cardiac fibroblasts. Furthermore, CyPA directly induced the hypertrophy of cultured neonatal cardiac myocytes. CONCLUSIONS: CyPA is required for Ang II-mediated cardiac hypertrophy by directly potentiating ROS production, stimulating the proliferation and migration of cardiac fibroblasts, and promoting cardiac myocyte hypertrophy.


Assuntos
Apolipoproteínas E/deficiência , Cardiomegalia/enzimologia , Ciclofilina A/metabolismo , Miocárdio/enzimologia , Angiotensina II , Animais , Animais Recém-Nascidos , Apolipoproteínas E/genética , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Cardiomegalia/induzido quimicamente , Cardiomegalia/genética , Cardiomegalia/imunologia , Cardiomegalia/patologia , Cardiomegalia/prevenção & controle , Comunicação Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Ciclofilina A/deficiência , Ciclofilina A/genética , Modelos Animais de Doenças , Fibroblastos/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/imunologia , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Estresse Oxidativo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes/metabolismo , Fatores de Tempo
20.
Am J Physiol Regul Integr Comp Physiol ; 298(4): R1089-97, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20147609

RESUMO

Activation of the immune system by ANG II contributes to the pathogenesis of hypertension, and pharmacological suppression of lymphocyte responses can ameliorate hypertensive end-organ damage. Therefore, to examine the mechanisms through which lymphocytes mediate blood pressure elevation, we studied ANG II-dependent hypertension in scid mice lacking lymphocyte responses and wild-type controls. Scid mice had a blunted hypertensive response to chronic ANG II infusion and accordingly developed less cardiac hypertrophy. Moreover, lymphocyte deficiency led to significant reductions in heart and kidney injury following 4 wk of angiotensin. The muted hypertensive response in the scid mice was associated with increased sodium excretion, urine volumes, and weight loss beginning on day 5 of angiotensin infusion. To explore the mechanisms underlying alterations in blood pressure and renal sodium handling, we measured gene expression for vasoactive mediators in the kidney after 4 wk of ANG II administration. Scid mice and controls had similar renal expression for interferon-gamma, interleukin-1beta, and interleukin-6. By contrast, lymphocyte deficiency (i.e., scid mice) during ANG II infusion led to upregulation of tumor necrosis factor-alpha, endothelial nitric oxide synthase (eNOS), and cyclooxygenase-2 (COX-2) in the kidney. In turn, this enhanced eNOS and COX-2 expression in the scid kidneys was associated with exaggerated renal generation of nitric oxide, prostaglandin E(2), and prostacyclin, all of which promote natriuresis. Thus, the absence of lymphocyte activity protects from hypertension by allowing blood pressure-induced sodium excretion, possibly via stimulation of eNOS- and COX-2-dependent pathways.


Assuntos
Angiotensina II/farmacologia , Hipertensão/induzido quimicamente , Linfócitos/fisiologia , Angiotensina II/efeitos adversos , Animais , Peso Corporal/efeitos dos fármacos , Cardiomegalia/induzido quimicamente , Cardiomegalia/imunologia , Cardiomegalia/fisiopatologia , Cruzamentos Genéticos , Progressão da Doença , Coração/efeitos dos fármacos , Coração/fisiopatologia , Hemodinâmica/fisiologia , Hipertensão/patologia , Hipertensão/fisiopatologia , Rim/efeitos dos fármacos , Rim/lesões , Rim/fisiopatologia , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos SCID/imunologia , Tamanho do Órgão/efeitos dos fármacos , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA