Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Eur J Heart Fail ; 24(11): 2000-2018, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36065751

RESUMO

Over 10 million doses of COVID-19 vaccines based on RNA technology, viral vectors, recombinant protein, and inactivated virus have been administered worldwide. Although generally very safe, post-vaccine myocarditis can result from adaptive humoral and cellular, cardiac-specific inflammation within days and weeks of vaccination. Rates of vaccine-associated myocarditis vary by age and sex with the highest rates in males between 12 and 39 years. The clinical course is generally mild with rare cases of left ventricular dysfunction, heart failure and arrhythmias. Mild cases are likely underdiagnosed as cardiac magnetic resonance imaging (CMR) is not commonly performed even in suspected cases and not at all in asymptomatic and mildly symptomatic patients. Hospitalization of symptomatic patients with electrocardiographic changes and increased plasma troponin levels is considered necessary in the acute phase to monitor for arrhythmias and potential decline in left ventricular function. In addition to evaluation for symptoms, electrocardiographic changes and elevated troponin levels, CMR is the best non-invasive diagnostic tool with endomyocardial biopsy being restricted to severe cases with heart failure and/or arrhythmias. The management beyond guideline-directed treatment of heart failure and arrhythmias includes non-specific measures to control pain. Anti-inflammatory drugs such as non-steroidal anti-inflammatory drugs, and corticosteroids have been used in more severe cases, with only anecdotal evidence for their effectiveness. In all age groups studied, the overall risks of SARS-CoV-2 infection-related hospitalization and death are hugely greater than the risks from post-vaccine myocarditis. This consensus statement serves as a practical resource for physicians in their clinical practice, to understand, diagnose, and manage affected patients. Furthermore, it is intended to stimulate research in this area.


Assuntos
Vacinas contra COVID-19 , COVID-19 , Adolescente , Adulto , Criança , Humanos , Adulto Jovem , COVID-19/prevenção & controle , Vacinas contra COVID-19/administração & dosagem , SARS-CoV-2
2.
Cardiovasc Res ; 118(2): 573-584, 2022 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-33576779

RESUMO

AIMS: Angiotensin (Ang) II signalling has been suggested to promote cardiac fibrosis in inflammatory heart diseases; however, the underlying mechanisms remain obscure. Using Agtr1a-/- mice with genetic deletion of angiotensin receptor type 1 (ATR1) and the experimental autoimmune myocarditis (EAM) model, we aimed to elucidate the role of Ang II-ATR1 pathway in development of heart-specific autoimmunity and post-inflammatory fibrosis. METHODS AND RESULTS: EAM was induced in wild-type (WT) and Agtr1a-/- mice by subcutaneous injections with alpha myosin heavy chain peptide emulsified in complete Freund's adjuvant. Agtr1a-/- mice developed myocarditis to a similar extent as WT controls at day 21 but showed reduced fibrosis and better systolic function at day 40. Crisscross bone marrow chimaera experiments proved that ATR1 signalling in the bone marrow compartment was critical for cardiac fibrosis. Heart infiltrating, bone-marrow-derived cells produced Ang II, but lack of ATR1 in these cells reduced transforming growth factor beta (TGF-ß)-mediated fibrotic responses. At the molecular level, Agtr1a-/- heart-inflammatory cells showed impaired TGF-ß-mediated phosphorylation of Smad2 and TAK1. In WT cells, TGF-ß induced formation of RhoA-GTP and RhoA-A-kinase anchoring protein-Lbc (AKAP-Lbc) complex. In Agtr1a-/- cells, stabilization of RhoA-GTP and interaction of RhoA with AKAP-Lbc were largely impaired. Furthermore, in contrast to WT cells, Agtr1a-/- cells stimulated with TGF-ß failed to activate canonical Wnt pathway indicated by suppressed activity of glycogen synthase kinase-3 (GSK-3)ß and nuclear ß-catenin translocation and showed reduced expression of Wnts. In line with these in vitro findings, ß-catenin was detected in inflammatory regions of hearts of WT, but not Agtr1a-/- mice and expression of canonical Wnt1 and Wnt10b were lower in Agtr1a-/- hearts. CONCLUSION: Ang II-ATR1 signalling is critical for development of post-inflammatory fibrotic remodelling and dilated cardiomyopathy. Our data underpin the importance of Ang II-ATR1 in effective TGF-ß downstream signalling response including activation of profibrotic Wnt/ß-catenin pathway.


Assuntos
Angiotensina II/metabolismo , Doenças Autoimunes/metabolismo , Autoimunidade , Linfócitos T CD4-Positivos/metabolismo , Miocardite/metabolismo , Miócitos Cardíacos/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Via de Sinalização Wnt , Animais , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Linfócitos T CD4-Positivos/imunologia , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Fibrose , Mediadores da Inflamação/metabolismo , Ativação Linfocitária , Camundongos Endogâmicos BALB C , Camundongos Knockout , Miocardite/genética , Miocardite/imunologia , Miocardite/patologia , Miócitos Cardíacos/imunologia , Miócitos Cardíacos/patologia , Receptor Tipo 1 de Angiotensina/genética , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteína Wnt1/genética , Proteína Wnt1/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
3.
Eur J Heart Fail ; 23(4): 512-526, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33826207

RESUMO

Cardiac amyloidosis is a serious and progressive infiltrative disease that is caused by the deposition of amyloid fibrils at the cardiac level. It can be due to rare genetic variants in the hereditary forms or as a consequence of acquired conditions. Thanks to advances in imaging techniques and the possibility of achieving a non-invasive diagnosis, we now know that cardiac amyloidosis is a more frequent disease than traditionally considered. In this position paper the Working Group on Myocardial and Pericardial Disease proposes an invasive and non-invasive definition of cardiac amyloidosis, addresses clinical scenarios and situations to suspect the condition and proposes a diagnostic algorithm to aid diagnosis. Furthermore, we also review how to monitor and treat cardiac amyloidosis, in an attempt to bridge the gap between the latest advances in the field and clinical practice.


Assuntos
Amiloidose , Cardiologia , Cardiomiopatias , Cardiopatias , Insuficiência Cardíaca , Humanos , Miocárdio
4.
Eur Heart J ; 42(16): 1554-1568, 2021 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-33825853

RESUMO

Cardiac amyloidosis is a serious and progressive infiltrative disease that is caused by the deposition of amyloid fibrils at the cardiac level. It can be due to rare genetic variants in the hereditary forms or as a consequence of acquired conditions. Thanks to advances in imaging techniques and the possibility of achieving a non-invasive diagnosis, we now know that cardiac amyloidosis is a more frequent disease than traditionally considered. In this position paper the Working Group on Myocardial and Pericardial Disease proposes an invasive and non-invasive definition of cardiac amyloidosis, addresses clinical scenarios and situations to suspect the condition and proposes a diagnostic algorithm to aid diagnosis. Furthermore, we also review how to monitor and treat cardiac amyloidosis, in an attempt to bridge the gap between the latest advances in the field and clinical practice.


Assuntos
Amiloidose , Cardiomiopatias , Cardiopatias , Amiloidose/diagnóstico , Amiloidose/terapia , Cardiomiopatias/diagnóstico , Cardiomiopatias/terapia , Coração , Cardiopatias/diagnóstico , Cardiopatias/terapia , Humanos , Miocárdio
5.
Swiss Med Wkly ; 150: w20343, 2020 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-33035354

RESUMO

INTRODUCTION: Sudden cardiac death caused by malignant arrhythmia can be prevented by the use of defibrillators. Although the wearable cardioverter defibrillator (WCD) can prevent such an event, its role in clinical practice is ill defined. We investigated the use of the WCD in Switzerland with emphasis on prescription rate, therapy adherence and treatment rate. MATERIALS AND METHODS: The Swiss WCD Registry is a retrospective observational registry including patients using a WCD. Patients were included from the first WCD use in Switzerland until February 2018. Baseline characteristics and data on WCD usage were examined for the total study population, and separately for each hospital. RESULTS: From 1 December 2011 to 18 February 2018, a total of 456 patients (67.1% of all WCDs prescribed in Switzerland and 81.1% of all prescribed in the participating hospitals) were included in the registry. Up to 2017 there was a yearly increase in the number of prescribed WCDs to a maximum of 271 prescriptions per year. The mean age of patients was 57 years (± 14), 81 (17.8%) were female and mean left ventricular ejection fraction (EF) was 32% (± 13). The most common indications for WCD use were new-onset ischaemic cardiomyopathy (ICM) with EF ≤35% (206 patients, 45.2%), new-onset nonischaemic cardiomyopathy (NICM) with EF ≤35% (115 patients, 25.2%), unknown arrhythmic risk (83 patients, 18.2%), bridging to implantable cardioverter-defibrillator implantation or heart transplant (37 patients, 8.1%) and congenital/inherited heart disease (15 patients, 3.3%). Median wear duration was 58 days (interquartile range [IQR] 31–94) with a median average daily wear time of 22.6 hours (IQR 20–23.2). Seventeen appropriate therapies from the WCD were delivered in the whole population (treatment rate: 3.7%) to a total of 12 patients (2.6% of all patients). The most common underlying heart disease in patients with a treatment was ICM (13/17, 76.5%). There were no inappropriate treatments. CONCLUSION: The use of WCDs has increased in Switzerland over the years for a variety of indications. There is high therapy adherence to the WCD, and a treatment rate comparable to previously published registry data.  .


Assuntos
Desfibriladores Implantáveis , Dispositivos Eletrônicos Vestíveis , Morte Súbita Cardíaca/prevenção & controle , Desfibriladores , Cardioversão Elétrica , Feminino , Humanos , Pessoa de Meia-Idade , Estudos Retrospectivos , Volume Sistólico , Suíça , Função Ventricular Esquerda
6.
Int Immunopharmacol ; 87: 106822, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32738595

RESUMO

C-Cbl-associated protein (CAP), also known as Sorbin and SH3 domain-containing protein 1 (Sorbs1) or ponsin, an adaptor protein of the insulin-signalling pathway, mediates anti-viral and anti-cytotoxic protection in acute viral heart disease. In the present study we describe a novel protective immuno-modulatory function of CAP in inflammation. Among the three members of the Sorbs family of adapter molecules, which include CAP (Sorbs1), ArgBP2 (Sorbs2), and Vinexin (Sorbs3), CAP consistently down-regulated the expression of pro-inflammatory cytokines in mouse fibroblasts, cardiomyocytes, and myeloid-derived leukocytes, after Toll-like receptor (TLR) stimulation. Upon the same TLR stimulation, ArgBP2 partially down-regulated pro-inflammatory cytokine production in mouse fibroblasts and cardiomyocytes, while Vinexin rather promoted their production. Mechanistically, CAP limited pro-inflammatory cytokine expression by suppressing the phosphorylation of Inhibitor of kappa B (IκB) kinase (Iκκ)-α and Iκκ-ß and their downstream NF-κB-dependent signalling pathway. Molecular affinity between CAP and Iκκ-α/ Iκκ-ß was necessary to block the NF-κB pathway. The CAP-dependent inhibitory mechanism - in vivo exclusively IL-6 inhibition - was confirmed after collecting blood from mice with systemic inflammation induced by lipopolysaccharide (LPS) and in the heart tissue collected from mice infected with the cardiotropic Coxsackievirus B3 (CVB3). Taken together, CAP down-regulates pro-inflammatory cytokines by interfering with the normal function of the NF-κB pathway. The promotion of CAP production could support the development of new strategies aiming to limit excessive and detrimental activation of the immune system.


Assuntos
Citocinas/imunologia , NF-kappa B/imunologia , Proteínas Proto-Oncogênicas c-cbl/imunologia , Animais , Linhagem Celular , Infecções por Coxsackievirus/imunologia , Enterovirus Humano B , Fibroblastos , Humanos , Leucócitos , Camundongos Knockout , Miócitos Cardíacos , Proteínas Proto-Oncogênicas c-cbl/genética , Transdução de Sinais
7.
J Mol Cell Cardiol ; 139: 24-32, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31972266

RESUMO

AIMS: Sirtuin 6 (Sirt6) is a NAD+-dependent deacetylase that plays a key role in DNA repair, inflammation and lipid regulation. Sirt6-null mice show severe metabolic defects and accelerated aging. Macrophage-foam cell formation via scavenger receptors is a key step in atherogenesis. We determined the effects of bone marrow-restricted Sirt6 deletion on foam cell formation and atherogenesis using a mouse model. METHODS AND RESULTS: Sirt6 deletion in bone marrow-derived cells increased aortic plaques, lipid content and macrophage numbers in recipient Apoe-/- mice fed a high-cholesterol diet for 12 weeks (n = 12-14, p < .001). In RAW macrophages, Sirt6 overexpression reduced oxidized low-density lipoprotein (oxLDL) uptake, Sirt6 knockdown enhanced it and increased mRNA and protein levels of macrophage scavenger receptor 1 (Msr1), whereas levels of other oxLDL uptake and efflux transporters remained unchanged. Similarly, in human primary macrophages, Sirt6 knockdown increased MSR1 protein levels and oxLDL uptake. Double knockdown of Sirt6 and Msr1 abolished the increase in oxLDL uptake observed upon Sirt6 single knockdown. FACS analyses of macrophages from aortic plaques of Sirt6-deficient bone marrow-transplanted mice showed increased MSR1 protein expression. Double knockdown of Sirt6 and the transcription factor c-Myc in RAW cells abolished the increase in Msr1 mRNA and protein levels; c-Myc overexpression increased Msr1 mRNA and protein levels. CONCLUSIONS: Loss of Sirt6 in bone marrow-derived cells is proatherogenic; hereby macrophages play an important role given a c-Myc-dependent increase in MSR1 protein expression and an enhanced oxLDL uptake in human and murine macrophages. These findings assign endogenous SIRT6 in macrophages an important atheroprotective role.


Assuntos
Aterosclerose/metabolismo , Aterosclerose/patologia , Medula Óssea/patologia , Deleção de Genes , Receptores Depuradores Classe A/metabolismo , Sirtuínas/genética , Sirtuínas/metabolismo , Animais , Aorta/patologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/metabolismo , Transplante de Medula Óssea , Regulação para Baixo , Técnicas de Silenciamento de Genes , Hematopoese , Homozigoto , Humanos , Lipoproteínas LDL/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Modelos Biológicos , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Células RAW 264.7
8.
Thromb Haemost ; 120(1): 168-180, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31858519

RESUMO

Ischemia/reperfusion (I/R) injury in acute myocardial infarction activates several deleterious molecular mechanisms. The transcription factor JunD regulates pathways involved in oxidative stress as well as in cellular proliferation, differentiation, and death. The present study investigated the potential role of JunD as a modulator of myocardial injury pathways in a mouse model of cardiac I/R injury. Infarct size, systemic and local inflammation, and production of reactive oxygen species, as well as cytosolic and mitochondrial apoptotic pathways were investigated in adult males after myocardial I/R. In wild-type (WT) mice, 30 minutes after ischemia and up to 24 hours following reperfusion, cardiac JunD messenger ribonucleic acid expression was reduced while JunB increased. Cardiac-specific JunD overexpressing mice (JunDTg/0 ) displayed larger infarcts compared with WT. However, postischemic inflammatory or oxidative responses did not differ. JunD overexpression reduced Sirt3 transcription by binding to its promoter, thus leading to mitochondrial dysfunction, myocardial cell death, and increased infarct size. On the other hand, JunD silencing reduced, while Sirt3 silencing increased infarct size. In human myocardial autopsy specimens, JunD-positive areas within the infarcted left ventricle staining corresponded to undetectable Sirt3 areas in consecutive sections of the same heart. Cardiac-specific JunD overexpression increases myocardial infarct size following I/R. These effects are mediated via Sirt3 transcriptional repression, mitochondrial swelling, and increased apoptosis, suggesting that JunD is a key regulator of myocardial I/R injury. The present data set the stage for further investigation of the potential role of Sirt3 activation as a novel target for the treatment of acute myocardial infarction.


Assuntos
Mitocôndrias/metabolismo , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/fisiologia , Proteínas Proto-Oncogênicas c-jun/metabolismo , Traumatismo por Reperfusão/metabolismo , Sirtuína 3/metabolismo , Animais , Apoptose , Modelos Animais de Doenças , Regulação para Baixo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infarto do Miocárdio/patologia , Miocárdio/patologia , Especificidade de Órgãos , Proteínas Proto-Oncogênicas c-jun/genética , Traumatismo por Reperfusão/patologia , Sirtuína 3/genética , Regulação para Cima
9.
Science ; 366(6467): 881-886, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31727837

RESUMO

Myocarditis can develop into inflammatory cardiomyopathy through chronic stimulation of myosin heavy chain 6-specific T helper (TH)1 and TH17 cells. However, mechanisms governing the cardiotoxicity programming of heart-specific T cells have remained elusive. Using a mouse model of spontaneous autoimmune myocarditis, we show that progression of myocarditis to lethal heart disease depends on cardiac myosin-specific TH17 cells imprinted in the intestine by a commensal Bacteroides species peptide mimic. Both the successful prevention of lethal disease in mice by antibiotic therapy and the significantly elevated Bacteroides-specific CD4+ T cell and B cell responses observed in human myocarditis patients suggest that mimic peptides from commensal bacteria can promote inflammatory cardiomyopathy in genetically susceptible individuals. The ability to restrain cardiotoxic T cells through manipulation of the microbiome thereby transforms inflammatory cardiomyopathy into a targetable disease.


Assuntos
Doenças Autoimunes/complicações , Bacteroides/imunologia , Cardiomiopatia Dilatada/imunologia , Cardiomiopatia Dilatada/microbiologia , Microbioma Gastrointestinal/imunologia , Miocardite/complicações , Peptídeos/imunologia , beta-Galactosidase/imunologia , Animais , Doenças Autoimunes/imunologia , Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Modelos Animais de Doenças , Humanos , Intestinos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Miocardite/imunologia , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/imunologia , Células Th17/imunologia
10.
Swiss Med Wkly ; 149: w20136, 2019 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-31707723

RESUMO

INTRODUCTION: The wearable cardioverter-defibrillator (WCD) has established itself in treatment of potentially life-threatening ventricular arrhythmias, when implantation of an implantable cardioverter-defibrillator (ICD) is not warranted. Careful patient selection for this therapy is crucial, but unfortunately very little information from randomised controlled trials is available to guide clinical decision-making. Consequently, data from real-world patient registries play a more important role in this context. MATERIALS AND METHODS: A retrospective observational study was conducted at the University Hospital of Zurich and the GZO Regional Healthcare Centre in Wetzikon. Clinical databases were screened for patients with a history of WCD use from the time of its approval in Switzerland in July 2014 until February 2018. Baseline characteristics, WCD data and outcome data, with an emphasis on ICD implantation and ICD therapies, were collected and analysed. RESULTS: Two-hundred and seven patients were included in the primary analysis. Eighty-six percent were male and the mean age was 58 ± 13 years. The underlying heart disease was ischaemic cardiomyopathy (ICM), non-ischaemic cardiomyopathy (NICM) and congenital/inherited heart diseases in 60, 35 and 5%, respectively. The most common indication for WCD use was heart failure with an ejection fraction (EF) <35% due to ICM or NICM (43 and 27%, respectively). Three of the 207 patients received an appropriate shock over a median WCD wear-time of 62 days (interquartile range [IQR] 35–95). No inappropriate shocks were registered. Median average daily wear-time was 22.6 hours (IQR 19.9–23.2) and was significantly shorter for patients for whom WCD discontinuation was due to comfort issues (17 patients, p = 0.003). After the end of WCD therapy, 48% were implanted with an ICD. In those receiving an ICD, the rate of appropriate ICD therapies (either shock or antitachycardia pacing) was 8% during a median follow-up of 110 days (IQR 23–421). CONCLUSION: The WCD is safe and effective in terminating malignant ventricular arrhythmias. A substantial subgroup of patients, however, discontinued WCD use prematurely because of comfort issues. This subset of patients deserves further attention in clinical practice to ensure therapy adherence.  .


Assuntos
Morte Súbita Cardíaca/prevenção & controle , Desfibriladores , Cardioversão Elétrica/instrumentação , Insuficiência Cardíaca/terapia , Dispositivos Eletrônicos Vestíveis , Morte Súbita Cardíaca/etiologia , Eletrocardiografia , Feminino , Insuficiência Cardíaca/mortalidade , Insuficiência Cardíaca/fisiopatologia , Hospitais , Humanos , Masculino , Pessoa de Meia-Idade , Isquemia Miocárdica/complicações , Cooperação do Paciente , Sistema de Registros , Estudos Retrospectivos , Suíça , Taquicardia Ventricular/complicações
12.
J Exp Med ; 216(2): 350-368, 2019 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-30647120

RESUMO

Heart failure due to dilated cardiomyopathy is frequently caused by myocarditis. However, the pathogenesis of myocarditis remains incompletely understood. Here, we report the presence of neutrophil extracellular traps (NETs) in cardiac tissue of patients and mice with myocarditis. Inhibition of NET formation in experimental autoimmune myocarditis (EAM) of mice substantially reduces inflammation in the acute phase of the disease. Targeting the cytokine midkine (MK), which mediates NET formation in vitro, not only attenuates NET formation in vivo and the infiltration of polymorphonuclear neutrophils (PMNs) but also reduces fibrosis and preserves systolic function during EAM. Low-density lipoprotein receptor-related protein 1 (LRP1) acts as the functionally relevant receptor for MK-induced PMN recruitment as well as NET formation. In summary, NETosis substantially contributes to the pathogenesis of myocarditis and drives cardiac inflammation, probably via MK, which promotes PMN trafficking and NETosis. Thus, MK as well as NETs may represent novel therapeutic targets for the treatment of cardiac inflammation.


Assuntos
Doenças Autoimunes/imunologia , Movimento Celular/imunologia , Armadilhas Extracelulares/imunologia , Midkina/imunologia , Miocardite/imunologia , Miocárdio/imunologia , Neutrófilos/imunologia , Animais , Doenças Autoimunes/genética , Doenças Autoimunes/patologia , Movimento Celular/genética , Armadilhas Extracelulares/genética , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/imunologia , Camundongos , Camundongos Transgênicos , Midkina/genética , Miocardite/genética , Miocardite/patologia , Miocárdio/patologia , Neutrófilos/patologia , Receptores de LDL/genética , Receptores de LDL/imunologia , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/imunologia
13.
Eur Heart J ; 38(18): 1413-1425, 2017 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-27099262

RESUMO

AIMS: Myocardial fibrosis critically contributes to cardiac dysfunction in inflammatory dilated cardiomyopathy (iDCM). Activation of transforming growth factor-ß (TGF-ß) signalling is a key-step in promoting tissue remodelling and fibrosis in iDCM. Downstream mechanisms controlling these processes, remain elusive. METHODS AND RESULTS: Experimental autoimmune myocarditis (EAM) was induced in BALB/c mice with heart-specific antigen and adjuvant. Using heart-inflammatory precursors, as well as mouse and human cardiac fibroblasts, we demonstrated rapid secretion of Wnt proteins and activation of Wnt/ß-catenin pathway in response to TGF-ß signalling. Inactivation of extracellular Wnt with secreted Frizzled-related protein 2 (sFRP2) or inhibition of Wnt secretion with Wnt-C59 prevented TGF-ß-mediated transformation of inflammatory precursors and cardiac fibroblasts into pathogenic myofibroblasts. Inhibition of T-cell factor (TCF)/ß-catenin-mediated transcription with ICG-001 or genetic loss of ß-catenin also prevented TGF-ß-induced myofibroblasts formation. Furthermore, blocking of Smad-independent TGF-ß-activated kinase 1 (TAK1) pathway completely abrogated TGF-ß-induced Wnt secretion. Activation of Wnt pathway in the absence of TGF-ß, however, failed to transform precursors into myofibroblasts. The critical role of Wnt axis for cardiac fibrosis in iDCM is also supported by elevated Wnt-1/Wnt-5a levels in human samples from hearts with myocarditis. Accordingly, and as an in vivo proof of principle, inhibition of Wnt secretion or TCF/ß-catenin-mediated transcription abrogated the development of post-inflammatory fibrosis in EAM. CONCLUSION: We identified TAK1-mediated rapid Wnt protein secretion as a novel downstream key mechanism of TGF-ß-mediated myofibroblast differentiation and myocardial fibrosis progression in human and mouse myocarditis. Thus, pharmacological targeting of Wnts might represent a promising therapeutic approach against iDCM in the future.


Assuntos
Doenças Autoimunes/etiologia , Miocardite/etiologia , Miocárdio/patologia , Fator de Crescimento Transformador beta/fisiologia , Proteínas Wnt/metabolismo , Animais , Benzenoacetamidas/farmacologia , Diferenciação Celular/fisiologia , Progressão da Doença , Fibrose/fisiopatologia , Humanos , MAP Quinase Quinase Quinases/metabolismo , MAP Quinase Quinase Quinases/fisiologia , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos BALB C , Miofibroblastos/fisiologia , Piridinas/farmacologia , Transdução de Sinais/fisiologia , Células-Tronco/fisiologia , Fatores de Transcrição TCF/metabolismo , Disfunção Ventricular/fisiopatologia , Proteína Wnt-5a/metabolismo , Proteína Wnt1/metabolismo , beta Catenina/metabolismo
14.
J Am Coll Cardiol ; 68(21): 2348-2364, 2016 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-27884253

RESUMO

Myocarditis is a diverse group of heart-specific immune processes classified by clinical and histopathological manifestations. Up to 40% of dilated cardiomyopathy is associated with inflammation or viral infection. Recent experimental studies revealed complex regulatory roles for several microribonucleic acids and T-cell and macrophage subtypes. Although the prevalence of myocarditis remained stable between 1990 and 2013 at about 22 per 100,000 people, overall mortality from cardiomyopathy and myocarditis has decreased since 2005. The diagnostic and prognostic value of cardiac magnetic resonance has increased with new, higher-sensitivity sequences. Positron emission tomography has emerged as a useful tool for diagnosis of cardiac sarcoidosis. The sensitivity of endomyocardial biopsy may be increased, especially in suspected sarcoidosis, by the use of electrogram guidance to target regions of abnormal signal. Investigational treatments on the basis of mechanistic advances are entering clinical trials. Revised management recommendations regarding athletic participation after acute myocarditis have heightened the importance of early diagnosis.


Assuntos
Cardiomiopatias , Imunidade Celular , Miocardite , Miocárdio/patologia , Linfócitos T/imunologia , Viroses/complicações , Biópsia , Cardiomiopatias/diagnóstico , Cardiomiopatias/epidemiologia , Cardiomiopatias/etiologia , Saúde Global , Humanos , Incidência , Miocardite/diagnóstico , Miocardite/epidemiologia , Miocardite/imunologia , Prognóstico
15.
Eur Heart J ; 36(11): 676-85, 2015 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-23321350

RESUMO

AIMS: Supernatants of serum-free cultured mononuclear cells (MNC) contain a mix of immunomodulating factors (secretome), which have been shown to attenuate detrimental inflammatory responses following myocardial ischaemia. Inflammatory dilated cardiomyopathy (iDCM) is a common cause of heart failure in young patients. Experimental autoimmune myocarditis (EAM) is a CD4+ T cell-dependent model, which mirrors important pathogenic aspects of iDCM. The aim of this study was to determine the influence of MNC secretome on myocardial inflammation in the EAM model. METHODS AND RESULTS: BALB/c mice were immunized twice with an alpha myosin heavy chain peptide together with Complete Freund adjuvant. Supernatants from mouse mononuclear cells were collected, dialysed, and injected i.p. at Day 0, Day 7, or Day 14, respectively. Myocarditis severity, T cell responses, and autoantibody formation were assessed at Day 21. The impact of MNC secretome on CD4+ T cell function and viability was evaluated using in vitro proliferation and cell viability assays. A single high-dose application of MNC secretome, injected at Day 14 after the first immunization, effectively attenuated myocardial inflammation. Mechanistically, MNC secretome induced caspase-8-dependent apoptosis in autoreactive CD4+ T cells. CONCLUSION: MNC secretome abrogated myocardial inflammation in a CD4+ T cell-dependent animal model of autoimmune myocarditis. This anti-inflammatory effect of MNC secretome suggests a novel and simple potential treatment concept for inflammatory heart diseases.


Assuntos
Doenças Autoimunes/prevenção & controle , Linfócitos T CD4-Positivos/fisiologia , Miocardite/prevenção & controle , Cadeias Pesadas de Miosina/farmacologia , Animais , Anticorpos/farmacologia , Apoptose/fisiologia , Autoanticorpos/metabolismo , Relação CD4-CD8 , Ligante de CD40/imunologia , Linfócitos T CD8-Positivos/fisiologia , Inibidores de Caspase/farmacologia , Proliferação de Células/fisiologia , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/fisiologia , Modelos Animais de Doenças , Proteína Ligante Fas/imunologia , Humanos , Camundongos Endogâmicos BALB C , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Baço/citologia
16.
Basic Res Cardiol ; 109(3): 411, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24763933

RESUMO

c-Cbl-associated protein (CAP), also called Sorbs1 or ponsin, has been described as an essential adapter protein in the insulin-signalling pathway. Here, we describe for the first time a unique protective role for CAP in viral myocarditis. Mortality and heart failure development were increased in CAP(-/-) mice compared to CAP(+/+) littermates after Coxsackievirus (CVB3) infection. Mechanistically, CAP protected from tissue apoptosis because of reduced CD8(+) T and natural killer cell cytotoxicity. Despite reduced cytotoxic elimination of CVB3-infected cells in CAP(+/+) hearts, however, CAP enhanced interferon regulatory factor 3 (IRF3)-dependent antiviral type I interferon production and decreased viral proliferation in vitro by binding to the cytoplasmic RIG-I-like receptor melanoma differentiation-associated protein 5 (MDA5). Taken together, these findings reveal a novel modulatory role for CAP in the heart as a key protein stabilizing antiviral type I interferon production, while protecting from excessive cytotoxic responses. Our study will help to define future strategies to develop treatments to limit detrimental responses during viral heart inflammation.


Assuntos
Apoptose , Infecções por Coxsackievirus/prevenção & controle , Enterovirus Humano B/imunologia , Interferon Tipo I/metabolismo , Proteínas dos Microfilamentos/metabolismo , Miocardite/prevenção & controle , Miocárdio/metabolismo , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Células Cultivadas , Infecções por Coxsackievirus/genética , Infecções por Coxsackievirus/imunologia , Infecções por Coxsackievirus/metabolismo , Infecções por Coxsackievirus/patologia , Infecções por Coxsackievirus/virologia , RNA Helicases DEAD-box/metabolismo , Modelos Animais de Doenças , Enterovirus Humano B/genética , Enterovirus Humano B/crescimento & desenvolvimento , Enterovirus Humano B/patogenicidade , Fator Regulador 3 de Interferon/metabolismo , Helicase IFIH1 Induzida por Interferon , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/virologia , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/deficiência , Proteínas dos Microfilamentos/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Miocardite/genética , Miocardite/imunologia , Miocardite/metabolismo , Miocardite/patologia , Miocardite/virologia , Miocárdio/imunologia , Miocárdio/patologia , Fatores de Tempo , Replicação Viral
17.
Circulation ; 128(14): 1542-54, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24030499

RESUMO

BACKGROUND: Viral myocarditis follows a fatal course in ≈30% of patients. Interleukin-1 receptor-associated kinase 4 (IRAK4), a major nodal signal transducer in innate immunity, can play a pivotal role in host inflammatory response. We sought to determine how IRAK4 modulates inflammation and outcome in a mouse model of viral myocarditis. METHODS AND RESULTS: Myocarditis was induced after intraperitoneal inoculation of coxsackievirus B3 into C57Bl/6 IRAK4-deficient mice and their littermate controls. Mortality and viral proliferation were markedly reduced in IRAK4(-/-) mice compared with their IRAK4(+/+) littermates. Disease resistance of IRAK4(-/-) mice paralleled increased amounts of protective heart-infiltrating CCR5(+) monocytes/macrophages and enhanced interferon-α and interferon-γ production 2 days after infection. Competitive bone marrow chimera demonstrated that intact IRAK4 function inhibited heart-specific migration of bone marrow-derived CCR5(+) cells. Mechanistically, lack of IRAK4 resulted in interferon regulatory factor 5 homodimerization via reduced melanoma differentiation-associated protein 5 degradation and enhanced Stat1 and Stat5 phosphorylation. Consequently, antiviral interferon-α and interferon-γ production, as well as CCR5(+) cell recruitment, increased, whereas the overall proinflammatory response was drastically reduced in the absence of IRAK4. CONCLUSIONS: Innate immunity signal transducer IRAK4 exacerbates viral myocarditis through inhibition of interferon production and reduced mobilization of protective CCR5(+) monocytes/macrophages to the heart. The combination of IRAK4 inhibitors and antiviral adjuvants may become an attractive therapeutic approach against viral myocarditis in the future.


Assuntos
Antígeno CD11b/análise , Infecções por Coxsackievirus/imunologia , Interferons/biossíntese , Quinases Associadas a Receptores de Interleucina-1/fisiologia , Monócitos/fisiologia , Miocardite/imunologia , Receptores CCR5/análise , Transferência Adotiva , Animais , Movimento Celular/fisiologia , Quimiocina CCL5/deficiência , Quimiocina CCL5/fisiologia , Infecções por Coxsackievirus/fisiopatologia , Infecções por Coxsackievirus/virologia , RNA Helicases DEAD-box/metabolismo , Dimerização , Resistência à Doença , Enterovirus Humano B/fisiologia , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/fisiopatologia , Fatores Reguladores de Interferon/química , Fatores Reguladores de Interferon/metabolismo , Helicase IFIH1 Induzida por Interferon , Quinases Associadas a Receptores de Interleucina-1/deficiência , Quinases Associadas a Receptores de Interleucina-1/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocardite/fisiopatologia , Miocardite/virologia , Doenças da Imunodeficiência Primária , Processamento de Proteína Pós-Traducional , Quimera por Radiação , Receptores CCR5/deficiência , Receptores CCR5/fisiologia , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT5/metabolismo , Replicação Viral
18.
Circulation ; 127(23): 2285-94, 2013 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-23671208

RESUMO

BACKGROUND: Activation of innate pattern-recognition receptors promotes CD4+ T-cell-mediated autoimmune myocarditis and subsequent inflammatory cardiomyopathy. Mechanisms that counterregulate exaggerated heart-specific autoimmunity are poorly understood. METHODS AND RESULTS: Experimental autoimmune myocarditis was induced in BALB/c mice by immunization with α-myosin heavy chain peptide and complete Freund's adjuvant. Together with interferon-γ, heat-killed Mycobacterium tuberculosis, an essential component of complete Freund's adjuvant, converted CD11b(hi)CD11c(-) monocytes into tumor necrosis factor-α- and nitric oxide synthase 2-producing dendritic cells (TipDCs). Heat-killed M. tuberculosis stimulated production of nitric oxide synthase 2 via Toll-like receptor 2-mediated nuclear factor-κB activation. TipDCs limited antigen-specific T-cell expansion through nitric oxide synthase 2-dependent nitric oxide production. Moreover, they promoted nitric oxide synthase 2 production in hematopoietic and stromal cells in a paracrine manner. Consequently, nitric oxide synthase 2 production by both radiosensitive hematopoietic and radioresistant stromal cells prevented exacerbation of autoimmune myocarditis in vivo. CONCLUSIONS: Innate Toll-like receptor 2 stimulation promotes formation of regulatory TipDCs, which confine autoreactive T-cell responses in experimental autoimmune myocarditis via nitric oxide. Therefore, activation of innate pattern-recognition receptors is critical not only for disease induction but also for counterregulatory mechanisms, protecting the heart from exaggerated autoimmunity.


Assuntos
Doenças Autoimunes/fisiopatologia , Células Dendríticas/metabolismo , Tolerância Imunológica/fisiologia , Interferon gama/fisiologia , Miocardite/fisiopatologia , Óxido Nítrico Sintase Tipo II/biossíntese , Óxido Nítrico/biossíntese , Transdução de Sinais , Linfócitos T Auxiliares-Indutores/patologia , Receptor 2 Toll-Like/fisiologia , Animais , Doenças Autoimunes/imunologia , Cardiomiopatia Dilatada/etiologia , Cardiomiopatia Dilatada/imunologia , Cardiomiopatia Dilatada/prevenção & controle , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas/citologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/imunologia , Indução Enzimática/efeitos dos fármacos , Células-Tronco Hematopoéticas/enzimologia , Células-Tronco Hematopoéticas/efeitos da radiação , Tolerância Imunológica/imunologia , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Monócitos/citologia , Monócitos/efeitos dos fármacos , Mycobacterium tuberculosis/imunologia , Miocardite/imunologia , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Comunicação Parácrina , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/toxicidade , Quimera por Radiação , Tolerância a Radiação , Células Estromais/enzimologia , Células Estromais/efeitos da radiação , Linfócitos T Auxiliares-Indutores/imunologia , Miosinas Ventriculares/imunologia , Miosinas Ventriculares/toxicidade
19.
Cardiovasc Res ; 99(3): 422-31, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23674516

RESUMO

AIMS: Adiponectin (APN) is an immunomodulatory and cardioprotective adipocytokine. Toll-like receptor (TLR) 4 mediates autoimmune reactions that cause myocarditis resulting in inflammation-induced cardiac injury. Here, we investigated whether APN inhibits inflammation and injury in autoimmune myocarditis by interfering with TLR4 signalling. METHODS AND RESULTS: APN overexpression in murine experimental autoimmune myocarditis (EAM) down-regulated cardiac expression of TLR4 and its downstream targets tumour necrosis factor (TNF)α, interleukin (IL)-6, IL-12, CC chemokine ligand (CCL)2, and intercellular adhesion molecule (ICAM)-1 resulting in reduced infiltration with cluster of differentiation (CD)3+, CD14+, and CD45+ immune cells as well as diminished myocardial apoptosis. Expression of TLR4 signalling pathway components was unchanged in hearts and spleens of APN-knockout (APN-KO) mice. In vitro APN had no effect on TLR4 expression in cardiac and immune cells but induced dissociation of APN receptors from the activated TLR4/CD14 signalling complex. APN inhibited the expression of a TLR4-mediated inflammatory phenotype induced by exogenous and endogenous TLR4 ligands as assessed by attenuated nuclear factor (NF)-κB activation and reduced expression of TNFα, IL-6, CCL2, and ICAM-1. Accordingly, following TLR4 ligation, splenocytes from APN-KO mice showed enhanced expression of TNFα, IL-6, IL-12, CCL2, and ICAM-1, whereas dendritic cells (DCs) from APN-KO mice demonstrated increased activation and T-cell priming capacity. Moreover, APN diminished TLR4-mediated splenocyte migration towards cardiac cells as well as cardiomyocyte apoptosis after co-cultivation with splenocytes. Mechanistically, APN inhibited TLR4 signalling through cyclooxygenase (COX)-2, protein kinase A (PKA), and meiosis-specific serine/threonine kinase (MEK)1. CONCLUSION: Our observations indicate that APN protects against inflammation and injury in autoimmune myocarditis by diminishing TLR4 signalling thereby attenuating inflammatory activation and interaction of cardiac and immune cells.


Assuntos
Adiponectina/fisiologia , Miocardite/imunologia , Miocardite/fisiopatologia , Receptor 4 Toll-Like/fisiologia , Adiponectina/deficiência , Adiponectina/genética , Animais , Apoptose/imunologia , Apoptose/fisiologia , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Doenças Autoimunes/fisiopatologia , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ciclo-Oxigenase 2/metabolismo , Feminino , Humanos , MAP Quinase Quinase 1/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocardite/patologia , Miócitos Cardíacos/imunologia , Miócitos Cardíacos/patologia , Miócitos Cardíacos/fisiologia , Cadeias Pesadas de Miosina/imunologia , NF-kappa B/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais
20.
Circ Cardiovasc Imaging ; 6(2): 277-84, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23343515

RESUMO

BACKGROUND: The goal of this study was to characterize the performance of fluorine-19 ((19)F) cardiac magnetic resonance (CMR) for the specific detection of inflammatory cells in a mouse model of myocarditis. Intravenously administered perfluorocarbons are taken up by infiltrating inflammatory cells and can be detected by (19)F-CMR. (19)F-labeled cells should, therefore, generate an exclusive signal at the inflamed regions within the myocardium. METHODS AND RESULTS: Experimental autoimmune myocarditis was induced in BALB/c mice. After intravenous injection of 2×200 µL of a perfluorocarbon on day 19 and 20 (n=9) after immunization, in vivo (19)F-CMR was performed at the peak of myocardial inflammation (day 21). In 5 additional animals, perfluorocarbon combined with FITC (fluorescein isothiocyanate) was administered for postmortem immunofluorescence and flow-cytometry analyses. Control experiments were performed in 9 animals. In vivo (19)F-CMR detected myocardial inflammation in all experimental autoimmune myocarditis-positive animals. Its resolution was sufficient to identify even small inflammatory foci, that is, at the surface of the right ventricle. Postmortem immunohistochemistry and flow cytometry confirmed the presence of perfluorocarbon in macrophages, dendritic cells, and granulocytes, but not in lymphocytes. The myocardial volume of elevated (19)F signal (rs=0.96; P<0.001), the (19)F signal-to-noise ratio (rs=0.92; P<0.001), and the (19)F signal integral (rs=0.96; P<0.001) at day 21 correlated with the histological myocarditis severity score. CONCLUSIONS: In vivo (19)F-CMR was successfully used to visualize the inflammation specifically and robustly in experimental autoimmune myocarditis, and thus allowed for an unprecedented insight into the involvement of inflammatory cells in the disease process.


Assuntos
Doenças Autoimunes/imunologia , Flúor , Fluorocarbonos , Imageamento por Ressonância Magnética , Células Mieloides/imunologia , Miocardite/imunologia , Miocárdio/imunologia , Animais , Doenças Autoimunes/metabolismo , Doenças Autoimunes/patologia , Células Dendríticas/imunologia , Modelos Animais de Doenças , Citometria de Fluxo , Flúor/administração & dosagem , Flúor/farmacocinética , Fluorocarbonos/administração & dosagem , Fluorocarbonos/farmacocinética , Granulócitos/imunologia , Imuno-Histoquímica , Injeções Intravenosas , Linfócitos/imunologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Células Mieloides/metabolismo , Células Mieloides/patologia , Miocardite/metabolismo , Miocardite/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Valor Preditivo dos Testes , Índice de Gravidade de Doença , Razão Sinal-Ruído
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA