Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 126
Filtrar
1.
J Clin Invest ; 134(10)2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38747296

RESUMO

Arrhythmogenic cardiomyopathy (ACM) is an inherited cardiac condition characterized by cardiac remodeling and life-threatening ventricular arrhythmias. In this issue of the JCI, Chelko, Penna, and colleagues mechanistically addressed the intricate contribution of immune-mediated injury in ACM pathogenesis. Inhibition of nuclear factor κ-B (NF-κB) and infiltration of monocyte-derived macrophages expressing C-C motif chemokine receptor-2 (CCR2) alleviated the phenotypic ACM features (i.e., fibrofatty replacement, contractile dysfunction, and ventricular arrhythmias) in desmoglein 2-mutant (Dsg2mut/mut) mice. These findings pave the way for efficacious and targetable immune therapy for patients with ACM.


Assuntos
Desmogleína 2 , Macrófagos , Receptores CCR2 , Animais , Macrófagos/metabolismo , Macrófagos/imunologia , Macrófagos/patologia , Camundongos , Humanos , Desmogleína 2/genética , Desmogleína 2/metabolismo , Desmogleína 2/imunologia , Receptores CCR2/genética , Receptores CCR2/metabolismo , Receptores CCR2/antagonistas & inibidores , NF-kappa B/metabolismo , NF-kappa B/genética , Arritmias Cardíacas/patologia , Arritmias Cardíacas/imunologia , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Displasia Arritmogênica Ventricular Direita/genética , Displasia Arritmogênica Ventricular Direita/patologia , Displasia Arritmogênica Ventricular Direita/metabolismo , Cardiomiopatias/genética , Cardiomiopatias/patologia , Cardiomiopatias/imunologia , Cardiomiopatias/metabolismo
2.
Cell Tissue Res ; 386(1): 79-98, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34236518

RESUMO

The study of a desmoglein 2 murine model of arrhythmogenic cardiomyopathy revealed cardiac inflammation as a key early event leading to fibrosis. Arrhythmogenic cardiomyopathy (AC) is an inherited heart muscle disorder leading to ventricular arrhythmias and heart failure due to abnormalities in the cardiac desmosome. We examined how loss of desmoglein 2 (Dsg2) in the young murine heart leads to development of AC. Apoptosis was an early cellular phenotype, and RNA sequencing analysis revealed early activation of inflammatory-associated pathways in Dsg2-null (Dsg2-/-) hearts at postnatal day 14 (2 weeks) that were absent in the fibrotic heart of adult mice (10 weeks). This included upregulation of iRhom2/ADAM17 and its associated pro-inflammatory cytokines and receptors such as TNFα, IL6R and IL-6. Furthermore, genes linked to specific macrophage populations were also upregulated. This suggests cardiomyocyte stress triggers an early immune response to clear apoptotic cells allowing tissue remodelling later on in the fibrotic heart. Our analysis at the early disease stage suggests cardiac inflammation is an important response and may be one of the mechanisms responsible for AC disease progression.


Assuntos
Arritmias Cardíacas/imunologia , Cardiomiopatias/imunologia , Desmogleína 2/metabolismo , Fibrose/fisiopatologia , Insuficiência Cardíaca/fisiopatologia , Inflamação/complicações , Animais , Modelos Animais de Doenças , Humanos , Inflamação/patologia , Camundongos
3.
J Mol Cell Cardiol ; 160: 121-127, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34303670

RESUMO

Immune checkpoint inhibitors (ICI) have changed the landscape of cancer therapy, but their use carries a high risk of cardiac immune related adverse events (iRAEs). With the expanding utilization of ICI therapy, there is a growing need to understand the underlying mechanisms behind their anti-tumor activity as well as their immune-mediated toxicities. In this review, we will focus on clinical characteristics and immune pathways of ICI cardiotoxicity, with an emphasis on single-cell technologies used to gain insights in this field. We will focus on three key areas of ICI-mediated immune pathways, including the anti-tumor immune response, the augmentation of the immune response by ICIs, and the pathologic "autoimmune" response in some individuals leading to immune-mediated toxicity, as well as local factors in the myocardial immune environment predisposing to autoimmunity. Discerning the underlying mechanisms of these immune pathways is necessary to inform the development of targeted therapies for ICI cardiotoxicities and reduce treatment related morbidity and mortality.


Assuntos
Antineoplásicos/efeitos adversos , Arritmias Cardíacas/induzido quimicamente , Aterosclerose/induzido quimicamente , Inibidores de Checkpoint Imunológico/efeitos adversos , Imunoterapia/métodos , Miocardite/induzido quimicamente , Pericardite/induzido quimicamente , Vasculite/induzido quimicamente , Animais , Arritmias Cardíacas/imunologia , Aterosclerose/imunologia , Autoimunidade/efeitos dos fármacos , Cardiotoxicidade/imunologia , Humanos , Camundongos , Miocardite/imunologia , Pericardite/imunologia , Placa Aterosclerótica/induzido quimicamente , Placa Aterosclerótica/imunologia , Fatores de Risco , Resultado do Tratamento , Vasculite/imunologia
4.
Curr Probl Cardiol ; 46(6): 100820, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33743434

RESUMO

Chagas' disease (ChD) is a parasitic disease endemic to regions of Latin America and with an increasingly global reach. Up to 30% of patients with ChD develop severe dilated cardiomyopathy, ventricular arrhythmias, conduction disorders and/or sudden cardiac death. Autoantibodies against M2 muscarinic acetylcholine receptors (M2 mAChR) have been implicated in the pathogenesis of ChD. We sought to understand whether there was an association between anti-M2 mAChR autoantibody titers in patients with chronic ChD and the presence of distal cardiac conduction disorders or cardiac arrhythmias. We conducted a cross-sectional study in 79 patients from Argentina and Bolivia with chronic ChD without evident structural heart disease. Autoantibody titers were measured using indirect enzyme-linked immunosorbent assay. Elevated anti-M2 mAChR autoantibody titers were associated with the presence of distal conduction disease but not with cardiac arrhythmias. High anti-M2 mAChR autoantibody levels could assist with identifying early structural heart disease in patients with chronic ChD.


Assuntos
Arritmias Cardíacas/epidemiologia , Autoanticorpos/imunologia , Doença do Sistema de Condução Cardíaco/epidemiologia , Doença de Chagas/epidemiologia , Receptor Muscarínico M2/imunologia , Idoso , Argentina/epidemiologia , Arritmias Cardíacas/sangue , Arritmias Cardíacas/etiologia , Arritmias Cardíacas/imunologia , Autoanticorpos/sangue , Bolívia/epidemiologia , Doença do Sistema de Condução Cardíaco/sangue , Doença do Sistema de Condução Cardíaco/etiologia , Doença do Sistema de Condução Cardíaco/imunologia , Doença de Chagas/sangue , Doença de Chagas/complicações , Doença de Chagas/imunologia , Doença Crônica , Estudos Transversais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Prevalência
5.
Nat Rev Cardiol ; 18(8): 547-564, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33654273

RESUMO

Conduction disorders and arrhythmias remain difficult to treat and are increasingly prevalent owing to the increasing age and body mass of the general population, because both are risk factors for arrhythmia. Many of the underlying conditions that give rise to arrhythmia - including atrial fibrillation and ventricular arrhythmia, which frequently occur in patients with acute myocardial ischaemia or heart failure - can have an inflammatory component. In the past, inflammation was viewed mostly as an epiphenomenon associated with arrhythmia; however, the recently discovered inflammatory and non-canonical functions of cardiac immune cells indicate that leukocytes can be arrhythmogenic either by altering tissue composition or by interacting with cardiomyocytes; for example, by changing their phenotype or perhaps even by directly interfering with conduction. In this Review, we discuss the electrophysiological properties of leukocytes and how these cells relate to conduction in the heart. Given the thematic parallels, we also summarize the interactions between immune cells and neural systems that influence information transfer, extrapolating findings from the field of neuroscience to the heart and defining common themes. We aim to bridge the knowledge gap between electrophysiology and immunology, to promote conceptual connections between these two fields and to explore promising opportunities for future research.


Assuntos
Arritmias Cardíacas , Alergia e Imunologia , Arritmias Cardíacas/imunologia , Arritmias Cardíacas/fisiopatologia , Fenômenos Eletrofisiológicos , Humanos
6.
Am J Med Genet A ; 185(1): 213-218, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33044030

RESUMO

Glycosylation is a critical post/peri-translational modification required for the appropriate development and function of the immune system. As an example, abnormalities in glycosylation can cause antibody deficiency and reduced lymphocyte signaling, although the phenotype can be complex given the diverse roles of glycosylation. Human MGAT2 encodes N-acetylglucosaminyltransferase II, which is a critical enzyme in the processing of oligomannose to complex N-glycans. Complex N-glycans are essential for immune system functionality, but only one individual with MGAT2-CDG has been described to have an abnormal immunologic evaluation. MGAT2-CDG (CDG-IIa) is a congenital disorder of glycosylation (CDG) associated with profound global developmental disability, hypotonia, early onset epilepsy, and other multisystem manifestations. Here, we report a 4-year old female with MGAT2-CDG due to a novel homozygous pathogenic variant in MGAT2, a 4-base pair deletion, c.1006_1009delGACA. In addition to clinical features previously described in MGAT2-CDG, she experienced episodic asystole, persistent hypogammaglobulinemia, and defective ex vivo mitogen and antigen proliferative responses, but intact specific vaccine antibody titers. Her infection history has been mild despite the testing abnormalities. We compare this patient to the 15 previously reported patients in the literature, thus expanding both the genotypic and phenotypic spectrum for MGAT2-CDG.


Assuntos
Arritmias Cardíacas/genética , Defeitos Congênitos da Glicosilação/genética , Doenças do Sistema Imunitário/genética , N-Acetilglucosaminiltransferases/genética , Arritmias Cardíacas/complicações , Arritmias Cardíacas/imunologia , Arritmias Cardíacas/patologia , Pré-Escolar , Defeitos Congênitos da Glicosilação/complicações , Defeitos Congênitos da Glicosilação/imunologia , Defeitos Congênitos da Glicosilação/patologia , Feminino , Glicosilação , Homozigoto , Humanos , Doenças do Sistema Imunitário/complicações , Doenças do Sistema Imunitário/imunologia , Doenças do Sistema Imunitário/patologia , Mutação/genética , N-Acetilglucosaminiltransferases/imunologia , Fenótipo
7.
Respir Physiol Neurobiol ; 283: 103548, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32956843

RESUMO

BACKGROUND: Globally, the current medical emergency for novel coronavirus 2019 (COVID-19) leads to respiratory distress syndrome and death. PURPOSE: This review highlighted the effect of COVID-19 on systemic multiple organ failure syndromes. This review is intended to fill a gap in information about human physiological response to COVID-19 infections. This review may shed some light on other potential mechanisms and approaches in COVID -19 infections towards systemic multiorgan failure syndromes. FINDING: SARS-CoV-2 intervened mainly in the lung with progression to pneumonia and acute respiratory distress syndrome (ARDS) via the angiotensin-converting enzyme 2(ACE2) receptor. Depending on the viral load, infection spread through the ACE2 receptor further to various organs such as heart, liver, kidney, brain, endothelium, GIT, immune cell, and RBC (thromboembolism). This may be aggravated by cytokine storm with the extensive release of proinflammatory cytokines from the deregulating immune system. CONCLUSION: The widespread and vicious combinations of cytokines with organ crosstalk contribute to systemic hyper inflammation and ultimately lead to multiple organ dysfunction (Fig. 1). This comprehensive study comprises various manifestations of different organs in COVID-19 and may assist the clinicians and scientists pertaining to a broad approach to fight COVID 19.


Assuntos
Infecções por Coronavirus/imunologia , Síndrome da Liberação de Citocina/imunologia , Insuficiência de Múltiplos Órgãos/imunologia , Peptidil Dipeptidase A/metabolismo , Pneumonia Viral/imunologia , Síndrome do Desconforto Respiratório/imunologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Injúria Renal Aguda/imunologia , Injúria Renal Aguda/fisiopatologia , Enzima de Conversão de Angiotensina 2 , Arritmias Cardíacas/imunologia , Arritmias Cardíacas/fisiopatologia , Betacoronavirus/metabolismo , COVID-19 , Infecções por Coronavirus/fisiopatologia , Síndrome da Liberação de Citocina/fisiopatologia , Citocinas/imunologia , Endotélio Vascular/metabolismo , Eritrócitos/metabolismo , Gastroenteropatias/imunologia , Gastroenteropatias/fisiopatologia , Trato Gastrointestinal/metabolismo , Insuficiência Cardíaca/imunologia , Insuficiência Cardíaca/fisiopatologia , Humanos , Inflamação/imunologia , Rim/metabolismo , Fígado/metabolismo , Hepatopatias/imunologia , Hepatopatias/fisiopatologia , Pulmão/metabolismo , Insuficiência de Múltiplos Órgãos/fisiopatologia , Miocárdio/metabolismo , Pandemias , Pneumonia Viral/fisiopatologia , Síndrome do Desconforto Respiratório/fisiopatologia , SARS-CoV-2 , Tromboembolia/imunologia , Tromboembolia/fisiopatologia , Carga Viral
8.
Trends Endocrinol Metab ; 31(12): 893-904, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33172748

RESUMO

Coronavirus disease 2019 (COVID-19) patients with pre-existing cardiovascular disease (CVD) or with cardiovascular complications have a higher risk of mortality. The main cardiovascular complications of COVID-19 include acute cardiac injury, acute myocardial infarction (AMI), myocarditis, arrhythmia, heart failure, shock, and venous thromboembolism (VTE)/pulmonary embolism (PE). COVID-19 can cause cardiovascular complications or deterioration of coexisting CVD through direct or indirect mechanisms, including viral toxicity, dysregulation of the renin-angiotensin-aldosterone system (RAAS), endothelial cell damage and thromboinflammation, cytokine storm, and oxygen supply-demand mismatch. We systematically review cardiovascular manifestations, histopathology, and mechanisms of COVID-19, to help to formulate future research goals and facilitate the development of therapeutic management strategies.


Assuntos
COVID-19/fisiopatologia , Doenças Cardiovasculares/fisiopatologia , Enzima de Conversão de Angiotensina 2/metabolismo , Arritmias Cardíacas/imunologia , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatologia , COVID-19/imunologia , COVID-19/metabolismo , Doenças Cardiovasculares/imunologia , Doenças Cardiovasculares/metabolismo , Síndrome da Liberação de Citocina/imunologia , Síndrome da Liberação de Citocina/fisiopatologia , Cardiopatias/imunologia , Cardiopatias/metabolismo , Cardiopatias/fisiopatologia , Insuficiência Cardíaca/imunologia , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/fisiopatologia , Humanos , Hipóxia/imunologia , Hipóxia/metabolismo , Hipóxia/fisiopatologia , Infarto do Miocárdio/imunologia , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/fisiopatologia , Miocardite/imunologia , Miocardite/metabolismo , Miocardite/fisiopatologia , Embolia Pulmonar/imunologia , Embolia Pulmonar/metabolismo , Embolia Pulmonar/fisiopatologia , Sistema Renina-Angiotensina/fisiologia , SARS-CoV-2/imunologia , SARS-CoV-2/metabolismo , Choque/imunologia , Choque/metabolismo , Choque/fisiopatologia , Troponina/metabolismo , Tromboembolia Venosa/imunologia , Tromboembolia Venosa/metabolismo , Tromboembolia Venosa/fisiopatologia
9.
Basic Res Cardiol ; 115(4): 42, 2020 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-32529556

RESUMO

Arrhythmogenic cardiomyopathy (AC) is an incurable genetic disease, whose pathogenesis is poorly understood. AC is characterized by arrhythmia, fibrosis, and cardiodilation that may lead to sudden cardiac death or heart failure. To elucidate AC pathogenesis and to design possible treatment strategies of AC, multiple murine models have been established. Among them, mice carrying desmoglein 2 mutations are particularly valuable given the identification of desmoglein 2 mutations in human AC and the detection of desmoglein 2 auto-antibodies in AC patients. Using two mouse strains producing either a mutant desmoglein 2 or lacking desmoglein 2 in cardiomyocytes, we test the hypothesis that inflammation is a major component of disease pathogenesis. We show that multifocal cardiomyocyte necrosis initiates a neutrophil-dominated inflammatory response, which also involves macrophages and T cells. Increased expression of Ccl2/Ccr2, Ccl3/Ccr5, and Cxcl5/Cxcr2 mRNA reflects the observed immune cell recruitment. During the ensuing acute disease phase, Mmp12+ and Spp1+ macrophages and T cells accumulate in scars, which mature from cell- to collagen-rich. The expression of Cx3cl1/Cx3cr1, Ccl2/Ccr2, and Cxcl10/Cxcr3 dominates this disease phase. We furthermore find that during chronic disease progression macrophages and T cells persist within mature scars and are present in expanding interstitial fibrosis. Ccl12 and Cx3cl1 are predominant chemokines in this disease phase. Together, our observations provide strong evidence that specific immune cell populations and chemokine expression profiles modulate inflammatory and repair processes throughout AC progression.


Assuntos
Arritmias Cardíacas/imunologia , Cardiomiopatias/imunologia , Inflamação/imunologia , Animais , Arritmias Cardíacas/genética , Arritmias Cardíacas/patologia , Cardiomiopatias/genética , Cardiomiopatias/patologia , Desmogleína 2/genética , Inflamação/genética , Inflamação/patologia , Camundongos , Camundongos Mutantes , Mutação
10.
Circulation ; 141(22): 1764-1774, 2020 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-32312099

RESUMO

BACKGROUND: Cardiac arrest is a tragic event that causes 1 death roughly every 90 seconds worldwide. Survivors generally undergo a workup to identify the cause of arrest. However, 5% to 10% of cardiac arrests remain unexplained. Because cardiac arrhythmias underlie most cardiac arrests and increasing evidence strongly supports the involvement of autoantibodies in arrhythmogenesis, a large-panel autoantibody screening was performed in patients with cardiac arrest. METHODS: This is an observational, cross-sectional study of patients from the Montreal Heart Institute hospital cohort, a single-center registry of participants. A peptide microarray was designed to screen for immunoglobulin G targeting epitopes from all known cardiac ion channels with extracellular domains. Plasma samples from 23 patients with unexplained cardiac arrest were compared with those from 22 patients with cardiac arrest cases of ischemic origin and a group of 29 age-, sex-, and body mass index-matched healthy subjects. The false discovery rate, least absolute shrinkage and selection operator logistic regression, and random forest methods were carried out jointly to find significant differential immunoglobulin G responses. RESULTS: The autoantibody against the pore domain of the L-type voltage-gated calcium channel was consistently identified as a biomarker of idiopathic cardiac arrest (P=0.002; false discovery rate, 0.007; classification accuracies ≥0.83). Functional studies on human induced pluripotent stem cell-derived cardiomyocytes demonstrated that the anti-L-type voltage-gated calcium channel immunoglobulin G purified from patients with idiopathic cardiac arrest is proarrhythmogenic by reducing the action potential duration through calcium channel inhibition. CONCLUSIONS: The present report addresses the concept of autoimmunity and cardiac arrest. Hitherto unknown autoantibodies targeting extracellular sequences of cardiac ion channels were detected. Moreover, the study identified an autoantibody signature specific to patients with cardiac arrest.


Assuntos
Autoanticorpos/imunologia , Autoantígenos/imunologia , Canais de Cálcio Tipo L/imunologia , Parada Cardíaca/imunologia , Potenciais de Ação , Adulto , Idoso , Sequência de Aminoácidos , Especificidade de Anticorpos , Arritmias Cardíacas/sangue , Arritmias Cardíacas/imunologia , Arritmias Cardíacas/fisiopatologia , Autoanticorpos/sangue , Biomarcadores , Diferenciação Celular , Células Cultivadas , Estudos Transversais , Feminino , Parada Cardíaca/sangue , Parada Cardíaca/epidemiologia , Sistema de Condução Cardíaco/imunologia , Sistema de Condução Cardíaco/fisiopatologia , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Células-Tronco Pluripotentes Induzidas/citologia , Canais Iônicos/imunologia , Masculino , Pessoa de Meia-Idade , Miócitos Cardíacos/imunologia , Técnicas de Patch-Clamp , Biblioteca de Peptídeos , Análise Serial de Proteínas , Quebeque/epidemiologia , Sistema de Registros
11.
Circ J ; 84(5): 685-694, 2020 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-32101812

RESUMO

Autoimmune diseases (ADs) affect approximately 10% of the world's population. Because ADs are frequently systemic disorders, cardiac involvement is common. In this review we focus on typical arrhythmias and their pathogenesis, arrhythmia-associated mortality, and possible treatment options among selected ADs (sarcoidosis, systemic lupus erythematosus, scleroderma, type 1 diabetes, Graves' disease, rheumatoid arthritis, ankylosing spondylitis [AS], psoriasis, celiac disease [CD], and inflammatory bowel disease [IBD]). Rhythm disorders have different underlying pathophysiologies; myocardial inflammation and fibrosis seem to be the most important factors. Inflammatory processes and oxidative stress lead to cardiomyocyte necrosis, with subsequent electrical and structural remodeling. Furthermore, chronic inflammation is the pathophysiological basis linking AD to autonomic dysfunction, including sympathetic overactivation and a decline in parasympathetic function. Autoantibody-mediated inhibitory effects of cellular events (i.e., potassium or L-type calcium currents, M2muscarinic cholinergic or ß1-adrenergic receptor signaling) can also lead to cardiac arrhythmia. Drug-induced arrhythmias, caused, for example, by corticosteroids, methotrexate, chloroquine, are also observed among AD patients. The most common arrhythmia in most AD presentations is atrial arrhythmia (primarily atrial fibrillation), expect for sarcoidosis and scleroderma, which are characterized by a higher burden of ventricular arrhythmia. Arrhythmia-associated mortality is highest among patients with sarcoidosis and lowest among those with AS; there are scant data related to mortality in patients with psoriasis, CD, and IBD.


Assuntos
Arritmias Cardíacas/fisiopatologia , Doenças Autoimunes/fisiopatologia , Sistema de Condução Cardíaco/fisiopatologia , Frequência Cardíaca , Antiarrítmicos/uso terapêutico , Arritmias Cardíacas/imunologia , Arritmias Cardíacas/mortalidade , Arritmias Cardíacas/prevenção & controle , Doenças Autoimunes/tratamento farmacológico , Doenças Autoimunes/imunologia , Doenças Autoimunes/mortalidade , Sistema de Condução Cardíaco/efeitos dos fármacos , Sistema de Condução Cardíaco/imunologia , Frequência Cardíaca/efeitos dos fármacos , Humanos , Fatores Imunológicos/uso terapêutico , Prevalência , Prognóstico , Fatores de Risco
12.
Circ Heart Fail ; 13(1): e006155, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31957469

RESUMO

Antibodies that activate the ß1-AR (ß1-adrenoreceptor) can induce heart failure in animal models. These antibodies are often found in patients with heart failure secondary to varying etiologies. Their binding to the ß1 receptor leads to prolonged receptor activation with subsequent induction of cellular dysfunction, apoptosis, and arrhythmias. ß-blocker therapy while highly effective for heart failure, may not be sufficient treatment for patients who have ß1 receptor autoantibodies. Removal of these autoantibodies by immunoadsorption has been shown to improve heart failure in small studies. However, immunoadsorption is costly, time consuming, and carries potential risks. An alternative to immunoadsorption is neutralization of autoantibodies through the intravenous application of small soluble molecules, such as peptides or aptamers, which specifically target and neutralize ß1-AR autoantibodies. Peptides may induce immunogenicity. Animal as well as early phase human studies with aptamers have not shown safety concerns to date and have demonstrated effectiveness in reducing autoantibody levels. Novel aptamers have the potential advantage of having a wide spectrum of action, neutralizing a variety of known circulating G-protein coupled receptor autoantibodies. These aptamers, therefore, have the potential to be novel therapeutic option for patients with heart failure who have positive for ß1-AR autoantibodies. However, clinical outcomes trials are needed to assess the clinical utility of this novel approach to treat heart failure.


Assuntos
Antagonistas de Receptores Adrenérgicos beta 1/farmacologia , Autoanticorpos/imunologia , Insuficiência Cardíaca/tratamento farmacológico , Receptores Adrenérgicos beta 1/imunologia , Animais , Arritmias Cardíacas/tratamento farmacológico , Arritmias Cardíacas/imunologia , Insuficiência Cardíaca/fisiopatologia , Humanos
13.
Cells ; 10(1)2020 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-33396359

RESUMO

The immune system plays a pivotal role in the initiation, development and resolution of inflammation following insult or damage to organs. The heart is a vital organ which supplies nutrients and oxygen to all parts of the body. Heart failure (HF) has been conventionally described as a disease associated with cardiac tissue damage caused by systemic inflammation, arrhythmia and conduction defects. Cardiac inflammation and subsequent tissue damage is orchestrated by the infiltration and activation of various immune cells including neutrophils, monocytes, macrophages, eosinophils, mast cells, natural killer cells, and T and B cells into the myocardium. After tissue injury, monocytes and tissue-resident macrophages undergo marked phenotypic and functional changes, and function as key regulators of tissue repair, regeneration and fibrosis. Disturbance in resident macrophage functions such as uncontrolled production of inflammatory cytokines, growth factors and inefficient generation of an anti-inflammatory response or unsuccessful communication between macrophages and epithelial and endothelial cells and fibroblasts can lead to aberrant repair, persistent injury, and HF. Therefore, in this review, we discuss the role of cardiac macrophages on cardiac inflammation, tissue repair, regeneration and fibrosis.


Assuntos
Fibrose/metabolismo , Traumatismos Cardíacos/metabolismo , Macrófagos/metabolismo , Regeneração , Animais , Arritmias Cardíacas/imunologia , Arritmias Cardíacas/metabolismo , Citocinas/metabolismo , Cardiomiopatias Diabéticas/imunologia , Cardiomiopatias Diabéticas/metabolismo , Fibrose/imunologia , Coração/fisiopatologia , Traumatismos Cardíacos/imunologia , Homeostase , Humanos , Hipertensão/imunologia , Hipertensão/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Macrófagos/citologia , Mitocôndrias/imunologia , Miocárdio/citologia , Miocárdio/imunologia
14.
Clin Biochem ; 75: 1-6, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31786205

RESUMO

Cardiac arrhythmias are associated with substantial morbidity and mortality. Recent advances in the pathophysiological understanding of cardiac arrhythmia indicate that inflammation, fibrosis, and even autoimmune mechanisms could facilitate the development of arrhythmias by interfering either with fibroblast activation-related electrical remodeling or with the function of different cardiac ion channels, leading to the emerging concepts of autoimmune and inflammatory channelopathies. In this descriptive review, we considered recent data of the literature focusing on biomarkers reflecting the degree of inflammation, myocardial stretch, fibrosis and sustained B-cell activation as potential additional diagnostic, risk stratification tools and potential therapeutic targets in cardiac arrhythmia.


Assuntos
Arritmias Cardíacas/diagnóstico , Arritmias Cardíacas/sangue , Arritmias Cardíacas/imunologia , Arritmias Cardíacas/patologia , Linfócitos B/imunologia , Biomarcadores/sangue , Fibrose , Humanos , Inflamação/patologia , Ativação Linfocitária , Miocárdio/patologia
15.
J Cardiovasc Transl Res ; 13(5): 722-730, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-31833003

RESUMO

To investigate the role of classical (CLM, CD14++CD16-), intermediate (INTM, CD14++CD16+), and non-classical (Non-CLM, CD14+CD16++) monocytes in scar formation after ST-elevation myocardial infarction (STEMI), evaluated with cardiac magnetic resonance (CMR). One hundred two patients with a first STEMI had serial blood analyses after 1, 3, and 7 days. A CMR was performed at 7 days and 6 months, depicting scar core (CO), border zone (BZ), and the presence of BZ channels. CLM and INTM levels progressively decreased, correlated with the scar mass, CO, and BZ at 7 days and 6 months (p < 0.05), and inversely with left ventricular ejection fraction (LVEF, p < 0.01). Non-CLM levels gradually increased, correlated with BZ mass and the presence of BZ channels at 7 days and 6 months (p < 0.001).CLM and INTM are associated with infarct size and inversely with LVEF, whereas Non-CLM are associated with BZ mass and the presence of potentially arrhythmogenic substrate.


Assuntos
Arritmias Cardíacas/etiologia , Monócitos/imunologia , Miocárdio/patologia , Infarto do Miocárdio com Supradesnível do Segmento ST/complicações , Volume Sistólico , Função Ventricular Esquerda , Remodelação Ventricular , Idoso , Arritmias Cardíacas/imunologia , Arritmias Cardíacas/fisiopatologia , Biomarcadores/sangue , Feminino , Proteínas Ligadas por GPI/sangue , Humanos , Receptores de Lipopolissacarídeos/sangue , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Monócitos/classificação , Intervenção Coronária Percutânea , Estudos Prospectivos , Receptores de IgG/sangue , Fatores de Risco , Infarto do Miocárdio com Supradesnível do Segmento ST/imunologia , Infarto do Miocárdio com Supradesnível do Segmento ST/fisiopatologia , Infarto do Miocárdio com Supradesnível do Segmento ST/terapia , Fatores de Tempo , Resultado do Tratamento
16.
Can J Cardiol ; 35(3): 310-319, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30825952

RESUMO

Long-term survival of HIV-infected patients has significantly improved with the use of antiretroviral therapy (ART). As a consequence, cardiovascular diseases are now emerging as an important clinical problem in this population. Sudden cardiac death is the third leading cause of mortality in HIV patients. Twenty percent of patients with HIV who died of sudden cardiac death had previous cardiac arrhythmias including ventricular tachycardia, atrial fibrillation, and other unspecified rhythm disorders. This review presents a summary of HIV-related arrhythmias, associated risk factors specific to the HIV population, and underlying mechanisms. Compared with the general population, patients with HIV have several cardiac conditions and electrophysiological abnormalities. As a result, they have an increased risk of developing severe arrhythmias, that can lead to sudden cardiac death. Possible explanations may be related to non-ART polypharmacy, electrolyte imbalances, and use of substances observed in HIV-infected patients; many of these conditions are associated with alterations in cardiac electrical activity, increasing the risk of arrhythmia and sudden cardiac death. However, clinical and experimental evidence has also revealed that cardiac arrhythmias occur in HIV-infected patients, even in the absence of drugs. This indicates that HIV itself can change the electrophysiological properties of the heart profoundly and cause cardiac arrhythmias and related sudden cardiac death. The current knowledge of the underlying mechanisms, as well as the emerging role of inflammation in these arrhythmias, are discussed here.


Assuntos
Arritmias Cardíacas , Morte Súbita Cardíaca , Infecções por HIV/complicações , Arritmias Cardíacas/diagnóstico , Arritmias Cardíacas/etiologia , Arritmias Cardíacas/imunologia , Arritmias Cardíacas/fisiopatologia , Morte Súbita Cardíaca/epidemiologia , Morte Súbita Cardíaca/etiologia , Morte Súbita Cardíaca/prevenção & controle , Fenômenos Eletrofisiológicos , Infecções por HIV/tratamento farmacológico , Humanos , Fatores de Risco
17.
Sleep Breath ; 23(1): 95-101, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29744684

RESUMO

OBJECTIVES: We explored relationships between biochemical markers and cardiac responses of children with and without obstructive sleep apnoea (OSA) during exercise. We hypothesised that serum markers of sympathetic nervous system activity and low-grade inflammation would correlate with cardiac responses to exercise in children with or without OSA. METHODOLOGY: The study included 40 of 71 children with previously characterised responses to cardiopulmonary exercise testing. Measures included serum cytokine levels using a multiplex bead-based assay (interleukins IL-1ß, IL-2, IL-4, IL-6, IL-8, IL-10, TNF-α and IFN-γ). Serum amyloid A (SAA) was quantified by nephelometry, and metanephrine/normetanephrine levels were measured by liquid chromatography, mass-spectroscopy. Comparisons were made between children with and without OSA, and with and without obesity. Relationships between biomarkers and various cardiac parameters were explored by linear regression. RESULTS: Amongst the 40 children in this study, OSA was present in 23. Compared to the 17 children without OSA, those with OSA had higher resting serum IL-6 levels compared to those without (median 3.22 pg/ml vs. 2.31, p < 0.05). Regarding correlations with cardiac function after adjusting for OSA, IL-8 negatively correlated to heart rate (HR) response following exercise (p = 0.03) and IFN-γ negatively correlated with Stroke Volume Index (SVI) (p = 0.03). Both metanephrine and normetanephrine levels positively correlated with SVI (p = 0.04, p = 0.047; respectively) and QI (p = 0.04, p = 0.04; respectively) during exercise when adjusting for OSA. CONCLUSIONS: Children with OSA have raised morning levels of serum IL-6. Separately, higher levels of IFN-γ and IL-8 and lower levels of metanephrine and normetanephrine related to poorer cardiac function during exercise.


Assuntos
Arritmias Cardíacas/imunologia , Arritmias Cardíacas/fisiopatologia , Citocinas/sangue , Apneia Obstrutiva do Sono/imunologia , Biomarcadores/sangue , Criança , Estudos Transversais , Exercício Físico/fisiologia , Feminino , Humanos , Masculino , Índice de Gravidade de Doença , Apneia Obstrutiva do Sono/sangue
18.
Circulation ; 139(11): 1407-1421, 2019 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-30586752

RESUMO

BACKGROUND: Arterial hypertension and its organ sequelae show characteristics of T cell-mediated inflammatory diseases. Experimental anti-inflammatory therapies have been shown to ameliorate hypertensive end-organ damage. Recently, the CANTOS study (Canakinumab Antiinflammatory Thrombosis Outcome Study) targeting interleukin-1ß demonstrated that anti-inflammatory therapy reduces cardiovascular risk. The gut microbiome plays a pivotal role in immune homeostasis and cardiovascular health. Short-chain fatty acids (SCFAs) are produced from dietary fiber by gut bacteria and affect host immune homeostasis. Here, we investigated effects of the SCFA propionate in 2 different mouse models of hypertensive cardiovascular damage. METHODS: To investigate the effect of SCFAs on hypertensive cardiac damage and atherosclerosis, wild-type NMRI or apolipoprotein E knockout-deficient mice received propionate (200 mmol/L) or control in the drinking water. To induce hypertension, wild-type NMRI mice were infused with angiotensin II (1.44 mg·kg-1·d-1 subcutaneous) for 14 days. To accelerate the development of atherosclerosis, apolipoprotein E knockout mice were infused with angiotensin II (0.72 mg·kg-1·d-1 subcutaneous) for 28 days. Cardiac damage and atherosclerosis were assessed using histology, echocardiography, in vivo electrophysiology, immunofluorescence, and flow cytometry. Blood pressure was measured by radiotelemetry. Regulatory T cell depletion using PC61 antibody was used to examine the mode of action of propionate. RESULTS: Propionate significantly attenuated cardiac hypertrophy, fibrosis, vascular dysfunction, and hypertension in both models. Susceptibility to cardiac ventricular arrhythmias was significantly reduced in propionate-treated angiotensin II-infused wild-type NMRI mice. Aortic atherosclerotic lesion area was significantly decreased in propionate-treated apolipoprotein E knockout-deficient mice. Systemic inflammation was mitigated by propionate treatment, quantified as a reduction in splenic effector memory T cell frequencies and splenic T helper 17 cells in both models, and a decrease in local cardiac immune cell infiltration in wild-type NMRI mice. Cardioprotective effects of propionate were abrogated in regulatory T cell-depleted angiotensin II-infused mice, suggesting the effect is regulatory T cell-dependent. CONCLUSIONS: Our data emphasize an immune-modulatory role of SCFAs and their importance for cardiovascular health. The data suggest that lifestyle modifications leading to augmented SCFA production could be a beneficial nonpharmacological preventive strategy for patients with hypertensive cardiovascular disease.


Assuntos
Anti-Inflamatórios/farmacologia , Doenças da Aorta/tratamento farmacológico , Arritmias Cardíacas/prevenção & controle , Aterosclerose/tratamento farmacológico , Cardiomegalia/prevenção & controle , Hipertensão/tratamento farmacológico , Propionatos/farmacologia , Angiotensina II , Animais , Doenças da Aorta/genética , Doenças da Aorta/imunologia , Doenças da Aorta/patologia , Arritmias Cardíacas/imunologia , Arritmias Cardíacas/fisiopatologia , Pressão Arterial/efeitos dos fármacos , Aterosclerose/genética , Aterosclerose/imunologia , Aterosclerose/patologia , Cardiomegalia/imunologia , Cardiomegalia/fisiopatologia , Modelos Animais de Doenças , Hipertensão/induzido quimicamente , Hipertensão/imunologia , Hipertensão/fisiopatologia , Masculino , Camundongos Knockout para ApoE , Placa Aterosclerótica , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Células Th17/efeitos dos fármacos , Células Th17/imunologia
19.
Ageing Res Rev ; 48: 40-50, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30300712

RESUMO

An ageing myocardium possesses significant electrophysiological alterations that predisposes the elderly patient to arrhythmic risk. Whilst these alterations are intrinsic to the cardiac myocytes, they are modulated by the cardiac autonomic nervous system (ANS) and consequently, ageing of the cardiac ANS is fundamental to the development of arrhythmias. A systems-based approach that incorporates the influence of the cardiac ANS could lead to better mechanistic understanding of how arrhythmogenic triggers and substrates interact spatially and temporally to produce sustained arrhythmia and why its incidence increases with age. Despite the existence of physiological oscillations of ANS activity on the heart, pathological oscillations can lead to defective activation and recovery properties of the myocardium. Such changes can be attributable to the decrease in functionality and structural alterations to ANS specific receptors in the myocardium with age. These altered ANS adaptive responses can occur either as a normal ageing process or accelerated in the presence of specific cardiac pathologies, such as genetic mutations or neurodegenerative conditions. Targeted intervention that seek to manipulate the ageing ANS influence on the myocardium may prove to be an efficacious approach for the management of arrhythmia in the ageing population.


Assuntos
Envelhecimento/metabolismo , Arritmias Cardíacas/metabolismo , Sistema Nervoso Autônomo/metabolismo , Encéfalo/metabolismo , Miocárdio/metabolismo , Envelhecimento/imunologia , Envelhecimento/patologia , Animais , Arritmias Cardíacas/imunologia , Arritmias Cardíacas/patologia , Sistema Nervoso Autônomo/imunologia , Sistema Nervoso Autônomo/patologia , Encéfalo/imunologia , Encéfalo/patologia , Coração , Humanos , Miocárdio/imunologia , Miocárdio/patologia
20.
Curr Oncol Rep ; 20(6): 44, 2018 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-29644505

RESUMO

PURPOSE OF REVIEW: This review describes cardiotoxicity associated with adoptive T cell therapy and immune checkpoint blockade. RECENT FINDINGS: Cardiotoxicity is a rare but potentially fatal complication associated with novel immunotherapies. Both affinity-enhanced and chimeric antigen receptor T cells have been reported to cause hypotension, arrhythmia, and left ventricular dysfunction, typically in the setting of cytokine release syndrome. Immune checkpoint inhibitors are generally well-tolerated but have the potential to cause myocarditis, with clinical presentations ranging from asymptomatic cardiac biomarker elevation to heart failure, arrhythmia, and cardiogenic shock. Electrocardiography, cardiac biomarker measurement, and cardiac imaging are key components of the diagnostic evaluation. For suspected myocarditis, endomyocardial biopsy is recommended if the diagnosis remains unclear after initial testing. The incidence of immunotherapy-associated cardiotoxicity is likely underestimated and may increase as adoptive T cell therapy and immune checkpoint inhibitors are used in larger populations and for longer durations of therapy. Baseline and serial cardiac evaluation is recommended to facilitate early identification and treatment of cardiotoxicity.


Assuntos
Cardiotoxicidade/imunologia , Imunoterapia/efeitos adversos , Neoplasias/terapia , Arritmias Cardíacas/epidemiologia , Arritmias Cardíacas/etiologia , Arritmias Cardíacas/imunologia , Cardiotoxicidade/etiologia , Cardiotoxicidade/patologia , Insuficiência Cardíaca/epidemiologia , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/imunologia , Humanos , Hipotensão/epidemiologia , Hipotensão/etiologia , Hipotensão/imunologia , Neoplasias/complicações , Neoplasias/imunologia , Receptores de Antígenos de Linfócitos T/antagonistas & inibidores , Receptores de Antígenos de Linfócitos T/imunologia , Disfunção Ventricular Esquerda/epidemiologia , Disfunção Ventricular Esquerda/etiologia , Disfunção Ventricular Esquerda/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA