Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 21(19)2020 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-32998408

RESUMO

Despite relevant advances made in therapies for cardiovascular diseases (CVDs), they still represent the first cause of death worldwide. Cardiac fibrosis and excessive extracellular matrix (ECM) remodeling are common end-organ features in diseased hearts, leading to tissue stiffness, impaired myocardial functional, and progression to heart failure. Although fibrosis has been largely recognized to accompany and complicate various CVDs, events and mechanisms driving and governing fibrosis are still not entirely elucidated, and clinical interventions targeting cardiac fibrosis are not yet available. Immune cell types, both from innate and adaptive immunity, are involved not just in the classical response to pathogens, but they take an active part in "sterile" inflammation, in response to ischemia and other forms of injury. In this context, different cell types infiltrate the injured heart and release distinct pro-inflammatory cytokines that initiate the fibrotic response by triggering myofibroblast activation. The complex interplay between immune cells, fibroblasts, and other non-immune/host-derived cells is now considered as the major driving force of cardiac fibrosis. Here, we review and discuss the contribution of inflammatory cells of innate immunity, including neutrophils, macrophages, natural killer cells, eosinophils and mast cells, in modulating the myocardial microenvironment, by orchestrating the fibrogenic process in response to tissue injury. A better understanding of the time frame, sequences of events during immune cells infiltration, and their action in the injured inflammatory heart environment, may provide a rationale to design new and more efficacious therapeutic interventions to reduce cardiac fibrosis.


Assuntos
Comunicação Celular/imunologia , Fibrose Endomiocárdica/imunologia , Imunidade Inata , Traumatismo por Reperfusão Miocárdica/imunologia , Miocárdio/imunologia , Miofibroblastos/imunologia , Imunidade Adaptativa , Animais , Citocinas/imunologia , Citocinas/metabolismo , Fibrose Endomiocárdica/metabolismo , Fibrose Endomiocárdica/patologia , Eosinófilos/imunologia , Eosinófilos/metabolismo , Eosinófilos/patologia , Matriz Extracelular/química , Matriz Extracelular/imunologia , Matriz Extracelular/metabolismo , Humanos , Inflamação , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Mastócitos/imunologia , Mastócitos/metabolismo , Mastócitos/patologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio/patologia , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Neutrófilos/patologia
2.
Matrix Biol ; 91-92: 167-175, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32438054

RESUMO

Inflammation contributes to the development of heart failure (HF) through multiple mechanisms including regulating extracellular matrix (ECM) degradation and deposition. Interactions between cells in the myocardium orchestrates the magnitude and duration of inflammatory cell recruitment and ECM remodeling events associated with HF. More recently, studies have shown T-cells have signficant roles in post-MI wound healing. T-cell biology in HF illustrates the complexity of cross-talk between inflammatory cell types and resident fibroblasts. This review will focus on T-cell recruitment to the myocardium and T-cell specific factors that might influence cardiac wound healing and fibrosis in the heart with consideration of age and sex as important factors in T-cell activity.


Assuntos
Fibrose Endomiocárdica/imunologia , Matriz Extracelular/imunologia , Fibroblastos/imunologia , Insuficiência Cardíaca/imunologia , Infarto do Miocárdio/imunologia , Linfócitos T/imunologia , Fatores Etários , Antígenos CD/genética , Antígenos CD/imunologia , Comunicação Celular/genética , Comunicação Celular/imunologia , Citocinas/genética , Citocinas/imunologia , Fibrose Endomiocárdica/genética , Fibrose Endomiocárdica/metabolismo , Fibrose Endomiocárdica/patologia , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Feminino , Fibroblastos/patologia , Regulação da Expressão Gênica , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Humanos , Inflamação , Macrófagos/imunologia , Macrófagos/patologia , Masculino , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/imunologia , Miocárdio/patologia , Miócitos Cardíacos/imunologia , Miócitos Cardíacos/patologia , Transdução de Sinais , Linfócitos T/classificação , Linfócitos T/patologia
3.
Drug Dev Res ; 81(1): 114-126, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31605544

RESUMO

Aliskiren is an oral antihypertensive medication that acts by directly inhibiting renin. High levels of circulating renin and prorenin activate the pathological signaling pathway of fibrosis. This drug also reduces oxidative stress. Thus, the aim of this systematic review is to analyze experimental studies that show the actions of aliskiren on fibrosis. PubMed and LILACS databases were consulted using the keywords aliskiren and fibrosis within the period between 2005 and 2017. Fifty-three articles were analyzed. In the heart, aliskiren attenuated remodeling, hypertrophy, inflammatory cytokines, collagen deposition, and oxidative stress. In the kidneys, there was a reduction in interstitial fibrosis, the infiltration of inflammatory cells, apoptosis, proteinuria, and in the recruitment of macrophages. In diabetic models, an improvement in the albumin/creatinine relationship and in the insulin pathway in skeletal muscles was observed; aliskiren was beneficial to pancreatic function and glucose tolerance. In the liver, aliskiren reduced fibrosis, steatosis, inflammatory cytokines, and collagen deposition. In the lung and peritoneal tissues, there was a reduction in fibrosis. Many studies have reported on the beneficial effects of aliskiren on endothelial function and arterial rigidity. A reduction in fibrosis in different organs is cited by many authors, which complies with the results found in this review. However, studies diverge on the use of the drug in diabetic patients. Aliskiren has antifibrotic potential in several experimental models, interfering with the levels of fibrogenic cytokines and oxidative stress. Therefore, its use in diseases in which fibrosis plays an important pathophysiological role is suggested.


Assuntos
Amidas/administração & dosagem , Fibrose Endomiocárdica/tratamento farmacológico , Fumaratos/administração & dosagem , Nefrite Intersticial/tratamento farmacológico , Amidas/farmacologia , Animais , Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/farmacologia , Modelos Animais de Doenças , Reposicionamento de Medicamentos , Fibrose Endomiocárdica/imunologia , Fibrose Endomiocárdica/patologia , Fibrose , Fumaratos/farmacologia , Humanos , Nefrite Intersticial/imunologia , Nefrite Intersticial/patologia , Estresse Oxidativo/efeitos dos fármacos
4.
Nature ; 573(7774): 430-433, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31511695

RESUMO

Fibrosis is observed in nearly every form of myocardial disease1. Upon injury, cardiac fibroblasts in the heart begin to remodel the myocardium by depositing excess extracellular matrix, resulting in increased stiffness and reduced compliance of the tissue. Excessive cardiac fibrosis is an important factor in the progression of various forms of cardiac disease and heart failure2. However, clinical interventions and therapies that target fibrosis remain limited3. Here we demonstrate the efficacy of redirected T cell immunotherapy to specifically target pathological cardiac fibrosis in mice. We find that cardiac fibroblasts that express a xenogeneic antigen can be effectively targeted and ablated by adoptive transfer of antigen-specific CD8+ T cells. Through expression analysis of the gene signatures of cardiac fibroblasts obtained from healthy and diseased human hearts, we identify an endogenous target of cardiac fibroblasts-fibroblast activation protein. Adoptive transfer of T cells that express a chimeric antigen receptor against fibroblast activation protein results in a significant reduction in cardiac fibrosis and restoration of function after injury in mice. These results provide proof-of-principle for the development of immunotherapeutic drugs for the treatment of cardiac disease.


Assuntos
Linfócitos T CD8-Positivos , Fibrose Endomiocárdica/terapia , Imunoterapia Adotiva , Animais , Antígenos de Superfície/imunologia , Linfócitos T CD8-Positivos/imunologia , Fibrose Endomiocárdica/imunologia , Fibroblastos/imunologia , Humanos , Masculino , Camundongos , Ovalbumina/imunologia , Cicatrização
5.
Int J Mol Sci ; 19(12)2018 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-30467294

RESUMO

Heart failure (HF) is a cardiovascular syndrome characterized by maladaptive changes with an underlying inflammatory mediated pathogenesis. Nevertheless, current therapy is aimed at the heart workload and neurohormonal axis; thus, prognosis remains poor. To continue improving treatment, we rely on murine models for a better understanding of HF pathophysiology. Among them, pressure overload HF (PO-HF) animal models are a common strategy. Development of PO-HF is characterized by monocyte infiltration, which orchestrates a cascade of events leading to sustained inflammation and maladaptive changes. Here, we divide the PO-HF model progression into four phases and describe the inflammatory, structural, and gene expression profiles. This division is relevant due to its similarities with clinical hypertensive heart disease progression to HF. Evidence shows improvement in hemodynamic and other local parameters by altering the inflammatory response in a specific immune response at a specific point of time. Thus, it is relevant to focus on the time-dependent immune response interaction in order to provide more effective therapy. This review summarizes the pathogenesis of PO-HF murine models, highlighting the inflammatory events in a time frame view. By this approach, we expect to provide researchers with a better understanding of the intertwining time-dependent events that occur in PO-HF.


Assuntos
Linfócitos B/imunologia , Insuficiência Cardíaca/imunologia , Hipertensão/imunologia , Monócitos/imunologia , Linfócitos T/imunologia , Angiotensina II/administração & dosagem , Angiotensina II/efeitos adversos , Animais , Aorta/imunologia , Aorta/patologia , Linfócitos B/patologia , Cardiomegalia/imunologia , Cardiomegalia/patologia , Movimento Celular , Constrição Patológica/imunologia , Constrição Patológica/patologia , Citocinas/biossíntese , Citocinas/imunologia , Modelos Animais de Doenças , Fibrose Endomiocárdica/imunologia , Fibrose Endomiocárdica/patologia , Insuficiência Cardíaca/induzido quimicamente , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/patologia , Humanos , Hipertensão/complicações , Hipertensão/patologia , Camundongos , Monócitos/patologia , Linfócitos T/patologia , Fatores de Tempo , Disfunção Ventricular Esquerda/imunologia , Disfunção Ventricular Esquerda/patologia
6.
Biochem Biophys Res Commun ; 486(4): 930-936, 2017 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-28347819

RESUMO

We attempted to investigate the therapeutic effects of deferiprone on DC rats and explore the underlying mechanism. Total 24 6-week-old male Wistar rats (weighing from 180 g to 220 g) were subjected to DC model construction and then randomly divided to three groups (8 rats per group): DC group, DC + 50 mg, and DC + 100 mg deferiprone treatment group. The 8 normal rats were considered as controls. After deferiprone treatment for 20 weeks, the blood samples were collected for the biochemical parameters test, including fasting glucose, HOMA-IR (homeostasis model assessment of the insulin resistance), serum iron, ferritin and transferrin saturation (TS). The oxidative stress was assessed by detecting the level of malondialdehyde (MDA) and superoxide dismutase (SOD). Histopathologic changes were determined by Masson's trichrome staining and electron microscopy imaging. The expression levels of NF-κB (nuclear factor kappa B), COX2 (cytochrome c oxidase), tenascin C, collagen IV were measured by RT-PCR and western blotting. The expression of nitrotyrosine and MCP-1 (monocyte chemotactic protein 1) were determined by immunohistochemistry. Deferiprone treatment reduced iron deposition and IR in DC rats except for blood glucose. After deferiprone treatment, MDA level was significantly decreased and SOD level was increased significantly. The level of NF-κB, cyclooxygenase-2, tenascin C, collagen IV MCP-1 and nitrotyrosine were significantly reduced. There was no significant difference in the effect of deferiprone at 50 and 100 mg doses. Deferiprone showed therapeutic effects on DC by regulating the pro-inflammatory and pro-fibrotic factors.


Assuntos
Cardiomiopatias Diabéticas/tratamento farmacológico , Cardiomiopatias Diabéticas/imunologia , Miocardite/tratamento farmacológico , Miocardite/imunologia , Piridonas/administração & dosagem , Espécies Reativas de Oxigênio/imunologia , Animais , Deferiprona , Cardiomiopatias Diabéticas/patologia , Relação Dose-Resposta a Droga , Fibrose Endomiocárdica/tratamento farmacológico , Fibrose Endomiocárdica/imunologia , Fibrose Endomiocárdica/patologia , Fatores Imunológicos/imunologia , Quelantes de Ferro/administração & dosagem , Masculino , Miocardite/patologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/imunologia , Ratos , Ratos Wistar , Resultado do Tratamento
7.
Sci Rep ; 6: 32430, 2016 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-27577254

RESUMO

Chronic sustained stimulation of ß-adrenoceptor is closely related to cardiac fibrosis which is bad for cardiac function. Growing evidence showed that the high prevalence of ß1-adrenoceptor autoantibody (ß1-AA) in the sera of patients with various types of cardiovascular diseases decreased cardiac function. In the current study, we demonstrated that ß1-AA impaired the cardiac function evaluated by echocardiography and that ß1-AA triggered cardiac fibrosis in terms of increased expression of α-smooth muscle actin as the marker of myofibroblast and collagen deposition in a passive ß1-AA immunized mice model during 16 weeks. Further, we showed that ß1-AA activated ß1-AR/cAMP/PKA pathway and promoted proliferation in primary cardiac fibroblasts through specific binding to ß1-AR but not to ß2-AR. Moreover, ß1-AA was also likely to promote proliferation in cardiac fibroblasts through activating p38MAPK and ERK1/2 as p38MAPK inhibitor SB203580 and ERK1/2 inhibitor PD98059 partially reversed the proliferative effect. The persistent activating signalling of PKA and P38MAPK in 1 h induced by ß1-AA was associated with lacking agonist-induced desensitization phenomena. The conditioned medium from ß1-AA-stimulated cardiac fibroblasts induced cardiomyocyte apoptosis, which indicated that ß1-AA changed the secretion of cardiac fibroblasts contributing to cardiac injury. These findings will contribute to our understanding of the pathological mechanisms of ß1-AA.


Assuntos
Fibrose Endomiocárdica/imunologia , Fibroblastos/imunologia , Miocárdio/imunologia , Miócitos Cardíacos/imunologia , Receptores Adrenérgicos beta 1/genética , Actinas/genética , Actinas/imunologia , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Autoanticorpos/administração & dosagem , Autoanticorpos/biossíntese , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Ecocardiografia , Fibrose Endomiocárdica/etiologia , Fibrose Endomiocárdica/genética , Fibrose Endomiocárdica/patologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Flavonoides/farmacologia , Regulação da Expressão Gênica , Imidazóis/farmacologia , Imunização Passiva , Camundongos , Camundongos Endogâmicos BALB C , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/imunologia , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/imunologia , Miocárdio/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Cultura Primária de Células , Piridinas/farmacologia , Ratos , Receptores Adrenérgicos beta 1/imunologia , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia
9.
Zhongguo Zhong Yao Za Zhi ; 39(12): 2314-7, 2014 Jun.
Artigo em Chinês | MEDLINE | ID: mdl-25244766

RESUMO

To establish neonatal rat cardiac fibroblast inflammatory secretion model by using LPS 100 microg x L(-1) combined with ATP 5 mmol x L(-1), in order to study the inhibitory effect of quercetin on the secretion of inflammatory factors TNF-alpha, IL-1beta and IL-6 of cardiac fibroblasts, further investigate the effect of quercetin on the protein expression of p-NF-kappaB p65 (S276) and p-Akt (S473) by western blot, and discuss the inhibitory effect of quercetin on the inflammatory secretion of cardiac fibroblasts. According to the findings, quercetin with the concentrations between 51.74 micromol x L(-1) and 827.81 micromol x L(-1) had no significant effect on the activity of cardiac fibroblasts. Quercetin with the concentrations of 82.78, 41.39, 20.70 micromol x L(-1) could notably inhibit the increase of TNF-alpha and IL-1beta induced by LPS 100 microg x L(-1) for 3 h and then ATP 5 mmol x L(-1) for 36 h (P < 0.01 or P < 0.05). Quercetin with the concentrations of 82.78, 41.39 micromol x L(-1) could notably inhibit the increase of IL-6 induced LPS 100 microg x L(-1) for 3 h and then ATP 5 mmol x L(-1) for 36 h (P < 0.05), without any notable effect of quercetin with the concentration of 20.70 micromol x L(-1). Quercetin with the concentrations of 82.78, 41.39, 20. 70 micromol x L(-1) could notably inhibit the NF-kappaB p65 (S276) activation induced by LPS 100 microg x L(-1) for 3 h and then ATP 5 mmol x L(-1) for 15 min, with the most significant effect in 20.70 micromol x L(-1). Quercetin with the concentrations of 82.78, 41.39, 20.70 micromol x L(-1) could notably inhibit the increase of p-Akt(473) expression induced by LPS 100 microg x L(-1) for 3 h and then ATP 5 mmol x L(-1) for 240 min (P < 0.05). Therefore, this study believes that quercetin could attenuate the secretion of inflammatory factors TNF-alpha, IL-1beta and IL-6 of cardiac fibroblasts by inhibiting the activation of NF-kappaB p65 (S276) and Akt (473).


Assuntos
Medicamentos de Ervas Chinesas/administração & dosagem , Fibrose Endomiocárdica/tratamento farmacológico , Fibroblastos/efeitos dos fármacos , Quercetina/administração & dosagem , Animais , Células Cultivadas , Fibrose Endomiocárdica/genética , Fibrose Endomiocárdica/imunologia , Feminino , Fibroblastos/imunologia , Coração/efeitos dos fármacos , Humanos , Interleucina-6/genética , Interleucina-6/imunologia , Masculino , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/imunologia , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
10.
Clin Dev Immunol ; 2012: 361730, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22811738

RESUMO

Chagas' disease is caused by the protozoan parasite Trypanosoma cruzi. The immune system plays an important role in the reduction of parasite load, but may also contribute to the development of lesions observed during the chronic phase of the disease. We analyzed cytokines produced by inflammatory heart cells in 21 autopsy samples obtained from patients with Chagas' disease divided according to the presence or absence of heart failure (HF). Left ventricular sections were analyzed by immunohistochemistry using antibodies against human IL-4, IFN-γ, TGF-ß, TNF-α, and NOS2. In situ mRNA expression was quantified by a Low Density Array. The number of IFN-γ-positive cells was significantly higher than IL-4 positive cells. TNF-α, TGF-ß and NOS2 were detected in 65%, 62% and 94% of samples respectively. There was an association between TNF-α-producing cells and the presence of HF. Subjects with HF presented higher levels of STAT4 mRNA, whereas FoxP3 and STAT6 levels were similar in the two groups. A Th1 cytokine pattern predominated in the cardiac inflammatory cell infiltrate of Chagas' disease patients associated with HF. High degree of fibrosis was associated with low NOS2 expression. These results support the idea that Th1 immune responses are involved in heart lesions of Chagas' disease patients.


Assuntos
Doença de Chagas/complicações , Doença de Chagas/imunologia , Citocinas/metabolismo , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/imunologia , Doença de Chagas/genética , Citocinas/genética , Citocinas/imunologia , Fibrose Endomiocárdica/imunologia , Fibrose Endomiocárdica/patologia , Insuficiência Cardíaca/genética , Humanos , Interferon gama/genética , Interferon gama/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Células Th1/imunologia , Células Th1/metabolismo , Células Th2/imunologia , Células Th2/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
11.
J Mol Cell Cardiol ; 53(1): 6-14, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22446162

RESUMO

Extracellular cyclophilin A (CyPA) and its receptor Extracellular Matrix Metalloproteinase Inducer (EMMPRIN, CD147) modulate inflammatory processes beyond metalloproteinase (MMP) activity. Recently, we have shown that CyPA and CD147 are upregulated in patients with inflammatory cardiomyopathy. Here we investigate the role of CyPA and CD147 in murine coxsackievirus B3 (CVB3)-induced myocarditis. CVB3-infected CyPA(-/-) mice (129S6/SvEv) revealed a significantly reduced T-cell and macrophage recruitment at 8 days p.i. compared to wild-type mice. In A.BY/SnJ mice, treatment with the cyclophilin-inhibitor NIM811 was associated with a reduction of inflammatory lesions and MMP-9 expression but with enhanced virus replication 8 days p.i. At 28 days p.i. the extent of lesion areas was not affected bei NIM811, whereas the collagen content was reduced. Initiation of NIM811-treatment on day 12 (after an effective virus defense) resulted in an even more pronounced reduction of myocardial fibrosis. In conclusion, in CVB3-induced myocarditis CyPA is important for macrophage and T cell recruitment and effective virus defense and may represent a pharmacological target to modulate myocardial remodeling in myocarditis.


Assuntos
Infecções por Coxsackievirus/imunologia , Ciclofilina A/metabolismo , Fibrose Endomiocárdica/imunologia , Enterovirus Humano B/imunologia , Miocardite/imunologia , Animais , Basigina/metabolismo , Linhagem Celular , Ciclofilina A/antagonistas & inibidores , Ciclofilina A/deficiência , Ciclosporina/farmacologia , Fibrose Endomiocárdica/etiologia , Fibrose Endomiocárdica/patologia , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Miocardite/complicações , Miocardite/virologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Remodelação Ventricular/efeitos dos fármacos
12.
J Med Food ; 15(3): 269-77, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22181074

RESUMO

Studies have suggested that 1-α-OH-vitamin D3 and green tea polyphenols (GTPs) are promising dietary supplements for mitigating chronic inflammation-induced fibrosis of vessels because of their anti-inflammatory properties. This study evaluated (1) the impact of 1-α-OH-vitamin D3 on myocardial fibrosis in female rats with chronic inflammation and (2) if 1-α-OH-vitamin D3 and GTPs have an additive or synergistic effect to attenuate myocardial fibrosis in these female rats. A 3-month study of a 2 (no 1-α-OH-vitamin D3 vs. 0.05 µg/kg 1-α-OH-vitamin D3, five times per week) ×2 (no GTPs vs. 0.5% GTPs in drinking water) factorial design in lipopolysaccharide (LPS)-administered female rats was performed. Additionally, a group receiving placebo administration was used to compare with a group receiving LPS administration only to evaluate the effect of LPS. Masson's Trichrome staining evaluated myocardial fibrosis in coronary vessels and surrounding myocardium. Spleen cyclooxygenase-2 mRNA expression was determined by real-time polymerase chain reaction. Total lipid profiles were also determined. Whole blood was used for differential cell counts. Data were analyzed by two-way analysis of variance followed by mean separation procedures. At 3 months LPS administration induced myocardial fibrosis in vessels and surrounding myocardium, spleen cyclooxygenase-2 mRNA expression, and elevated leukocyte counts, whereas both 1-α-OH-vitamin D3 administration and GTPs supplementation significantly attenuated these pro-inflammatory events. The inhibitory effects of 1-α-OH-vitamin D3 and GTPs seem to be an individual effect, instead of an additive or synergistic effect. 1-α-OH-vitamin D3 and GTPs lowered red blood cell counts, hematocrit, and hemoglobin. Neither 1-α-OH-vitamin D3 nor GTPs affected lipid profiles. In summary, both 1-α-OH-vitamin D3 administration and GTPs supplementation mitigate myocardial fibrosis through suppression of a chronic inflammation innate immune response.


Assuntos
Anti-Inflamatórios/uso terapêutico , Suplementos Nutricionais , Fibrose Endomiocárdica/imunologia , Fibrose Endomiocárdica/prevenção & controle , Hidroxicolecalciferóis/uso terapêutico , Polifenóis/uso terapêutico , Chá/química , Animais , Vasos Coronários/imunologia , Vasos Coronários/patologia , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Fibrose Endomiocárdica/metabolismo , Fibrose Endomiocárdica/patologia , Feminino , Regulação Enzimológica da Expressão Gênica , Imunidade Inata , Leucocitose/imunologia , Leucocitose/prevenção & controle , Miocárdio/imunologia , Miocárdio/patologia , Fitoterapia , RNA Mensageiro/metabolismo , Distribuição Aleatória , Ratos , Ratos Endogâmicos , Baço/imunologia , Baço/metabolismo
13.
Circulation ; 124(19): 2106-16, 2011 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-22025605

RESUMO

BACKGROUND: Transgenic mice with cardiac-restricted overexpression of tumor necrosis factor (MHCsTNF mice) develop progressive myocardial fibrosis, diastolic dysfunction, and adverse cardiac remodeling. Insofar as tumor necrosis factor (TNF) does not directly stimulate fibroblast collagen synthesis, we asked whether TNF-induced fibrosis was mediated indirectly through interactions between mast cells and cardiac fibroblasts. METHODS AND RESULTS: Cardiac mast cell number increased 2 to 3 fold (P<0.001) in MHCsTNF mice compared with littermate controls. Outcrossing MHCsTNF mice with mast cell-deficient (c-kit(-/-)) mice showed that the 11-fold increase (P<0.001) in collagen volume fraction in MHCsTNF/c-kit(+/-) mice was abrogated in MHCsTNF/c-kit(-/-) mice, and that the leftward shifted left ventricular pressure-volume curve in the MHCsTNF/c-kit(+/-) mice was normalized in the MHCsTNF/c-kit(-/-) hearts. Furthermore, the increase in transforming growth factor ß1 and type I transforming growth factor ß receptor messenger RNA levels was significantly (P=0.03, P=0.01, respectively) attenuated in MHCsTNF/c-kit(-/-) when compared with MHCsTNF/c-kit(+/-) mice. Coculture of fibroblasts with mast cells resulted in enhanced α-smooth muscle actin expression, increased proliferation and collagen messenger RNA expression, and increased contraction of 3-dimensional collagen gels in MHCsTNF fibroblasts compared with littermate fibroblasts. The effects of mast cells were abrogated by type I transforming growth factor ß receptor antagonist NP-40208. CONCLUSIONS: These results suggest that increased mast cell density with resultant mast cell-cardiac fibroblast cross-talk is required for the development of myocardial fibrosis in inflammatory cardiomyopathy. Cardiac fibroblasts exposed to sustained inflammatory signaling exhibit an increased repertoire of profibrotic phenotypic responses in response to mast cell mediators.


Assuntos
Comunicação Celular/imunologia , Fibrose Endomiocárdica/patologia , Fibroblastos/patologia , Mastócitos/patologia , Miocardite/patologia , Miocárdio/patologia , Fator de Necrose Tumoral alfa/genética , Animais , Fibrose Endomiocárdica/imunologia , Fibrose Endomiocárdica/fisiopatologia , Fibroblastos/imunologia , Expressão Gênica/imunologia , Mastócitos/imunologia , Camundongos , Camundongos Transgênicos , Miocardite/imunologia , Miocardite/fisiopatologia , Miocárdio/imunologia , Fenótipo , Cultura Primária de Células , Pteridinas/farmacologia , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Fator de Crescimento Transformador beta/metabolismo , Fator de Necrose Tumoral alfa/imunologia
14.
J Cardiovasc Pharmacol ; 57(6): 625-9, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21633248

RESUMO

Atrial fibrillation (AF) is the most common arrhythmia in humans. It affects 5% of the population older than age 65 years and is projected to rise as the population ages. Experimental data from animal models of AF show that AF is associated with progressive structural and electrical remodeling of the atria. Atrial fibrosis alters atrial electrical conduction and excitability and provides a substrate for AF maintenance. However, whether fibrosis is causally related to AF or an epiphenomenon and the precise mechanisms underlying atrial fibrosis remain unclear. A variety of signaling systems involving angiotensin II and related mediators are centrally involved in atrial fibrosis. This article reviews the role that atrial fibrosis plays in AF, the mechanisms of atrial fibrosis, and emerging therapeutic approaches to AF aimed at attenuating atrial fibrosis.


Assuntos
Fibrilação Atrial/etiologia , Fibrilação Atrial/prevenção & controle , Cardiomiopatias/tratamento farmacológico , Cardiomiopatias/fisiopatologia , Átrios do Coração/patologia , Antagonistas de Receptores de Angiotensina/uso terapêutico , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Animais , Fibrilação Atrial/imunologia , Cardiomiopatias/imunologia , Cardiomiopatias/patologia , Fibrose Endomiocárdica/etiologia , Fibrose Endomiocárdica/imunologia , Fibrose , Átrios do Coração/efeitos dos fármacos , Humanos , Antagonistas de Receptores de Mineralocorticoides , Sistema Renina-Angiotensina/efeitos dos fármacos , Fator de Crescimento Transformador beta1/metabolismo
15.
Eur J Pharmacol ; 652(1-3): 126-35, 2011 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-21115000

RESUMO

Excess cytokine produced by inflammatory stimuli contributes to the progression of myocardial damage in myocarditis. Angiotensin-II has been shown to play a pivotal role in the pathophysiology of various organs, especially the cardiovascular system. Some angiotensin II type 1 receptor antagonists are reported to inhibit proinflammatory cytokine production in vitro and in vivo. We investigated whether telmisartan, an angiotensin II type 1 receptor antagonist protects against experimental autoimmune myocarditis by suppression of inflammatory cytokines and oxidative stress. Experimental autoimmune myocarditis was induced in Lewis rats by immunization with porcine cardiac myosin. The rats were divided into two groups and treated with either telmisartan (10mg/kg/day) or vehicle for 21days. Age-matched normal rats without immunization were also included in this study. Myocardial functional parameters were significantly improved by treatment with telmisartan compared with vehicle-treated rats. Increased myocardial mRNA expressions of inflammatory cytokines [interleukin (IL-6), IL-1ß, tumor necrosis factor-α and interferon-γ] were also suppressed by telmisartan treatment compared with vehicle-treated rats. Myocardial protein expressions of NADPH oxidase subunits p47phox, Nox-4, and gp91phox, myocardial levels of 8-hydroxydeoxyguanosine and 4-hydroxy-2-nonenal, and myocardial apoptosis were also significantly decreased by telmisartan treatment compared with vehicle-treated rats. Further, telmisartan significantly decreased endoplasmic reticulum stress markers in experimental autoimmune myocarditis rats. These findings suggest that telmisartan protects against experimental autoimmune myocarditis in rats, at least in part by suppressing inflammatory cytokines and oxidative stress; however, further investigations are needed before clinical use.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Doenças Autoimunes/tratamento farmacológico , Benzimidazóis/uso terapêutico , Benzoatos/uso terapêutico , Inflamação/tratamento farmacológico , Miocardite/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Benzimidazóis/farmacologia , Benzoatos/farmacologia , Western Blotting , Citocinas/metabolismo , Modelos Animais de Doenças , Fibrose Endomiocárdica/tratamento farmacológico , Fibrose Endomiocárdica/imunologia , Fibrose Endomiocárdica/patologia , Ensaio de Imunoadsorção Enzimática , Inflamação/imunologia , Inflamação/patologia , Masculino , Miocardite/imunologia , Miocardite/patologia , Ratos , Ratos Endogâmicos Lew , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Telmisartan
16.
Circulation ; 122(14): 1396-404, 2010 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-20855659

RESUMO

BACKGROUND: Experimental autoimmune myocarditis (EAM), a mouse model of post-infectious cardiomyopathy, reflects mechanisms of inflammatory cardiomyopathy in humans. EAM is characterized by an infiltration of inflammatory cells into the myocardium that can be followed by myocyte fibrosis, edema, and necrosis, leading to ventricular wall dysfunction and heart failure. Different data indicate that CD69 exerts an important immunoregulatory effect in vivo. However, the possible role of CD69 in autoimmune myocarditis has not been studied. METHODS AND RESULTS: We have explored the role of the leukocyte regulatory molecule CD69 in the inflammation that leads to cardiac dysfunction after myocardial injury in EAM. We have found that after induction of EAM, the draining lymph nodes from CD69-deficient mice developed an exacerbated Th17 inflammatory response, resulting in increases in the numbers of infiltrating leukocytes in the myocardium. In the chronic phase of EAM, transthoracic echocardiography revealed a significantly reduced left ventricular fractional shortening and a decreased ejection fraction in CD69-deficient mice, indicative of an impaired cardiac contractility. This condition was accompanied by a greater extent of myocardial fibrosis, an elevated number of sinus pauses on ECG, and an enhanced ratio of heart weight to body weight in CD69-/- mice. Moreover, both bone marrow transplantation and adoptive transfer of Th17 cells isolated from immunized CD69-/- mice with EAM into naive wild-type recipients reproduced the severity of the disease, demonstrating that CD69 exerts its function within the lymphocyte compartment. CONCLUSION: Our findings indicate that CD69 negatively regulates heart-specific Th17 responses, cardiac inflammation, and heart failure progression in EAM.


Assuntos
Doenças Autoimunes/fisiopatologia , Lectinas Tipo C/deficiência , Miocardite/fisiopatologia , Animais , Antígenos CD/genética , Antígenos de Diferenciação de Linfócitos T/genética , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Cruzamentos Genéticos , Fibrose Endomiocárdica/imunologia , Fibrose Endomiocárdica/fisiopatologia , Feminino , Citometria de Fluxo , Humanos , Lectinas Tipo C/genética , Linfonodos/citologia , Linfonodos/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Miocardite/genética , Miocardite/imunologia , Cadeias Pesadas de Miosina/imunologia , Fragmentos de Peptídeos/imunologia , Índice de Gravidade de Doença , Baço/citologia , Baço/fisiologia
17.
PLoS Negl Trop Dis ; 4(4): e661, 2010 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-20422043

RESUMO

BACKGROUND: Endomyocardial Fibrosis (EMF) is a tropical restrictive cardiomyopathy of unknown etiology with high prevalence in Sub-Saharan Africa, for which it is unclear whether the primary target of injury is the endocardial endothelium, the subendocardial fibroblast, the coronary microcirculation or the myocyte. In an attempt to explore the possibility of endocardial lesions being a result of an immune response against the myocyte we assessed the presence and frequency of circulating anti-myocardial antibodies in EMF patients. METHODOLOGY/PRINCIPAL FINDINGS: EMF classification, assessment of severity and staging was based on echocardiography. We used sodium dodecylsulfate polyacrylamide gel electrophoresis (SDS-PAGE) of myocardial proteins followed by western blotting to screen serum samples for antiheart antibodies G and M classes. The degree of serum reactivity was correlated with the severity and activity of EMF. We studied 56 EMF patients and 10 healthy controls. IgG reactivity against myocardial proteins was stronger and more frequent in patients with EMF when compared to controls (30/56; 53.6% vs. 1/10; 10%, respectively). IgM reactivity was weak in both groups, although higher in EMF patients (11/56; 19.6%) when compared to controls (n = 0). EMF patients showed greater frequency and reactivity of IgG antibodies against myocardial proteins of molecular weights 35 kD, 42 kD and 70 kD (p values <0.01, <0.01 and <0.05 respectively). CONCLUSIONS: The presence of antibodies against myocardial proteins was demonstrated in a subset of EMF patients. These immune markers seem to be related with activity and might provide an adjunct tool for diagnosis and classification of EMF, therefore improving its management by identifying patients who may benefit from immunosuppressive therapy. Further research is needed to clarify the role of autoimmunity in the pathogenesis of EMF.


Assuntos
Autoanticorpos/sangue , Fibrose Endomiocárdica/imunologia , Fibrose Endomiocárdica/patologia , Miosinas/imunologia , Adolescente , Adulto , Western Blotting , Criança , Eletroforese em Gel de Poliacrilamida , Feminino , Humanos , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Masculino , Miocárdio/química , Proteínas/imunologia , Proteínas/isolamento & purificação , Adulto Jovem
19.
Rom J Morphol Embryol ; 49(3): 315-20, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18758635

RESUMO

This study was undertaken to assess several histopathological and immunohistochemical markers regarding some lesional aspects of ischemically and hypoxically damaged myocardium in sudden cardiac death. Tissue samples of myocardium from 17 middle age and young patients with sudden cardiac death, following acute or chronic cardio(myo)pathies, were analyzed using standard HE stain and indirect tristadial ABC peroxidase immunohistochemical method, for a panel of 12 antibodies grouped in three categories: antibodies involved in programmed cell death (bcl-2, p53, Fas/CD95, Fas-L, bax, caspase 9), muscular markers (Myo-D1, myogenin, desmin, actin) and growth factor receptors (b-FGF, VEGF, NGF). Myogenin was more sensitive in identifying the ischemic perilesional myocardic fibers than Myo-D1, but less specific, while desmin had a greater sensitivity than myogenin and Myo-D1 taken separately, but with no specificity for myocardic fibers. Fas-L, caspase 9 and bax were expressed in more than 75% of cases in perilesional residual cardiomyocytes, correlating to each other (r = 0.45, respectively r = 0.6, p<0.05). b-FGF, VEGF and NGF had a focally variable expression in subendocardial and subepicardial cardiomyocytes and were statistically independent. Even if there was a polymorphic expression of antibodies in the studied batch, our findings indicate that some parameters (Fas-L, b-FGF, Myo-D1) might be independent markers for predicting sudden cardiac death in patients with previously damaged myocardium.


Assuntos
Morte Súbita Cardíaca/patologia , Imunofenotipagem , Adulto , Apoptose/fisiologia , Biomarcadores/metabolismo , Desmina/metabolismo , Fibrose Endomiocárdica/imunologia , Fibrose Endomiocárdica/metabolismo , Fibrose Endomiocárdica/patologia , Proteína Ligante Fas/metabolismo , Feminino , Humanos , Masculino , Miocárdio/imunologia , Miocárdio/patologia , Miogenina/metabolismo , Adulto Jovem
20.
Herz ; 28(8): 692-701, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14689103

RESUMO

Hypertension is an important determinant of heart failure. Ventricular systolic and/or diastolic dysfunction can eventuate in an activation of the renin-angiotensin-aldosterone system (RAAS). Circulating RAAS effector hormones lead to: the appearance of the congestive heart failure syndrome; and a systemic illness that features oxi/nitrosative stress in various tissues, including blood, together with circulating pro-inflammatory cytokines, anorexia and, ultimately, cachexia. In addition to its well-known endocrine properties, expressed in epithelial cells of classic target tissues, aldosterone (ALDO) has an emerging portfolio of actions in nonclassic target tissues, such as circulating lymphocytes and monocytes (or peripheral blood mononuclear cells [PBMC]). This neuroendocrine-immune interface is based on Na(+)-dependent, ALDO-induced iterations in PBMC cytosolic free [Mg(2+)](i) and [Ca(2+)](i). Ca(2+) loading contributes to an induction of oxi/nitrosative stress and activation of PBMC transcriptome. This immunostimulatory state begets a pro-inflammatory/fibrogenic vascular phenotype involving the heart and systemic organs and can be prevented by either ALDO receptor antagonism or antioxidant. The established efficacy of ALDO receptor antagonism as an integral component of the overall management of symptomatic heart failure may include its immunomodulatory properties. This brief review traces studies that led to and then identified the neuroendocrine-immune interface that accompanies aldosteronism.


Assuntos
Citocinas/sangue , Insuficiência Cardíaca/imunologia , Hiperaldosteronismo/imunologia , Imunidade Celular/imunologia , Aldosterona/fisiologia , Antioxidantes/uso terapêutico , Fibrose Endomiocárdica/tratamento farmacológico , Fibrose Endomiocárdica/imunologia , Insuficiência Cardíaca/tratamento farmacológico , Humanos , Hiperaldosteronismo/tratamento farmacológico , Hipertensão/complicações , Hipertensão/fisiopatologia , Imunidade Celular/efeitos dos fármacos , Mediadores da Inflamação/sangue , Antagonistas de Receptores de Mineralocorticoides , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Receptores de Mineralocorticoides/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...