Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 494
Filtrar
2.
FEBS J ; 290(6): 1549-1562, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36181338

RESUMO

Sepsis often causes cell death via pyroptosis and hence results in septic cardiomyopathy. Triggering receptors expressed in myeloid cells-1 (TREM-1) may initiate cellular cascade pathways and, in turn, induce cell death and vital organ dysfunction in sepsis, but the evidence is limited. We set to investigate the role of TREM-1 on nucleotide-binding oligomerization domain-like receptors with pyrin domain-3 (NLRP3) inflammasome activation and cardiomyocyte pyroptosis in sepsis models using cardiac cell line (HL-1) and mice. In this study, TREM-1 was found to be significantly increased in HL-1 cells challenged with lipopolysaccharide (LPS). Pyroptosis was also significantly increased in the HL-1 cells challenged with lipopolysaccharide and an NLRP3 inflammasome activator, nigericin. The close interaction between TREM-1 and structural maintenance of chromosome 4 (SMC4) was also identified. Furthermore, inhibition of TREM-1 or SMC4 prevented the upregulation of NLRP3 and decreased Gasdermin-D, IL-1ß and caspase-1 cleavage. In mice subjected to caecal ligation and puncture, the TREM-1 inhibitor LR12 decreased the expression of NLRP3 and attenuated cardiomyocyte pyroptosis, leading to improved cardiac function and prolonged survival of septic mice. Our work demonstrates that, under septic conditions, TREM-1 plays a critical role in cardiomyocyte pyroptosis. Targeting TREM-1 and its associated molecules may therefore lead to novel therapeutic treatments for septic cardiomyopathy.


Assuntos
Inflamassomos , Miócitos Cardíacos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Piroptose , Sepse , Receptor Gatilho 1 Expresso em Células Mieloides , Animais , Humanos , Camundongos , Adenosina Trifosfatases/imunologia , Cardiomiopatias/etiologia , Cardiomiopatias/genética , Cardiomiopatias/imunologia , Caspase 1/genética , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/imunologia , Cromossomos Humanos Par 4/imunologia , Inflamassomos/agonistas , Inflamassomos/genética , Inflamassomos/imunologia , Lipopolissacarídeos/efeitos adversos , Lipopolissacarídeos/farmacologia , Células Mieloides/imunologia , Miócitos Cardíacos/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/agonistas , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Piroptose/genética , Piroptose/imunologia , Sepse/complicações , Sepse/genética , Sepse/imunologia , Receptor Gatilho 1 Expresso em Células Mieloides/antagonistas & inibidores , Receptor Gatilho 1 Expresso em Células Mieloides/genética , Receptor Gatilho 1 Expresso em Células Mieloides/imunologia
3.
BMC Cardiovasc Disord ; 22(1): 49, 2022 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-35152886

RESUMO

BACKGROUND: In a Phase I study treatment with the serum amyloid P component (SAP) depleter miridesap followed by monoclonal antibody to SAP (dezamizumab) showed removal of amyloid from liver, spleen and kidney in patients with systemic amyloidosis. We report results from a Phase 2 study and concurrent immuno-positron emission tomography (PET) study assessing efficacy, pharmacodynamics, pharmacokinetics, safety and cardiac uptake (of dezamizumab) following the same intervention in patients with cardiac amyloidosis. METHODS: Both were uncontrolled open-label studies. After SAP depletion with miridesap, patients received ≤ 6 monthly doses of dezamizumab in the Phase 2 trial (n = 7), ≤ 2 doses of non-radiolabelled dezamizumab plus [89Zr]Zr-dezamizumab (total mass dose of 80 mg at session 1 and 500 mg at session 2) in the immuno-PET study (n = 2). Primary endpoints of the Phase 2 study were changed from baseline to follow-up (at 8 weeks) in left ventricular mass (LVM) by cardiac magnetic resonance imaging and safety. Primary endpoint of the immuno-PET study was [89Zr]Zr-dezamizumab cardiac uptake assessed via PET. RESULTS: Dezamizumab produced no appreciable or consistent reduction in LVM nor improvement in cardiac function in the Phase 2 study. In the immuno-PET study, measurable cardiac uptake of [89Zr]Zr-dezamizumab, although seen in both patients, was moderate to low. Uptake was notably lower in the patient with higher LVM. Treatment-associated rash with cutaneous small-vessel vasculitis was observed in both studies. Abdominal large-vessel vasculitis after initial dezamizumab dosing (300 mg) occurred in the first patient with immunoglobulin light chain amyloidosis enrolled in the Phase 2 study. Symptom resolution was nearly complete within 24 h of intravenous methylprednisolone and dezamizumab discontinuation; abdominal computed tomography imaging showed vasculitis resolution by 8 weeks. CONCLUSIONS: Unlike previous observations of visceral amyloid reduction, there was no appreciable evidence of amyloid removal in patients with cardiac amyloidosis in this Phase 2 trial, potentially related to limited cardiac uptake of dezamizumab as demonstrated in the immuno-PET study. The benefit-risk assessment for dezamizumab in cardiac amyloidosis was considered unfavourable after the incidence of large-vessel vasculitis and development for this indication was terminated. Trial registration NCT03044353 (2 February 2017) and NCT03417830 (25 January 2018).


Assuntos
Amiloidose , Anticorpos Monoclonais , Ácidos Carboxílicos , Cardiomiopatias , Tomografia por Emissão de Pósitrons , Pirrolidinas , Componente Amiloide P Sérico , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Amiloidose/sangue , Amiloidose/diagnóstico por imagem , Amiloidose/tratamento farmacológico , Amiloidose/imunologia , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/uso terapêutico , Ácidos Carboxílicos/efeitos adversos , Ácidos Carboxílicos/uso terapêutico , Cardiomiopatias/sangue , Cardiomiopatias/diagnóstico por imagem , Cardiomiopatias/tratamento farmacológico , Cardiomiopatias/imunologia , Quimioterapia Combinada , Imageamento por Ressonância Magnética , Miocárdio/metabolismo , Miocárdio/patologia , Valor Preditivo dos Testes , Pirrolidinas/efeitos adversos , Pirrolidinas/uso terapêutico , Componente Amiloide P Sérico/antagonistas & inibidores , Componente Amiloide P Sérico/imunologia , Fatores de Tempo , Resultado do Tratamento , Reino Unido , Estados Unidos , Função Ventricular Esquerda/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos
4.
BMC Cardiovasc Disord ; 21(1): 619, 2021 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-34963460

RESUMO

OBJECTIVES: To evaluate the impact of autoantibodies against the M2-muscarinic receptor (anti-M2-R) on the clinical outcomes of patients receiving the standard treatment for peripartum cardiomyopathy (PPCM). METHODS: A total of 107 PPCM patients who received standard heart failure (HF) treatment between January 1998 and June 2020 were enrolled in this study. According to anti-M2-R reactivity, they were classified into negative (n = 59) and positive (n = 48) groups, denoted as the anti-M2-R (-) and anti-M2-R (+) groups. Echocardiography, 6-min walk distance, serum digoxin concentration (SDC), and routine laboratory tests were performed regularly for 2 years. The all-cause mortality, cardiovascular mortality, and rehospitalisation rate for HF were compared between the two groups. RESULTS: A total of 103 patients were included in the final data analysis, with 46 in the anti-M2-R (+) group and 57 in the anti-M2-R (-) group. Heart rate was lower in the anti-M2-R (+) group than in the anti-M2-R (-) group at the baseline (102.7 ± 6.1 bpm vs. 96.0 ± 6.4 bpm, p < 0.001). The initial SDC was higher in the anti-M2-R (+) group than in the anti-M2-R (-) group with the same dosage of digoxin (1.25 ± 0.45 vs. 0.78 ± 0.24 ng/mL, p < 0.001). The dosages of metoprolol and digoxin were higher in the anti-M2-R (-) patients than in the anti-M2-R (+) patients (38.8 ± 4.6 mg b.i.d. vs. 27.8 ± 5.3 mg b.i.d., p < 0.0001, respectively, for metoprolol; 0.12 ± 0.02 mg/day vs. 0.08 ± 0.04 mg/day, p < 0.0001, respectively, for digoxin). Furthermore, there was a greater improvement in cardiac function in the anti-M2-R (-) patients than in the anti-M2-R (+) patients. Multivariate analysis identified negativity for anti-M2-R as the independent predictor for the improvement of cardiac function. Rehospitalisation for HF was lower in the anti-M2-R (-) group, but all-cause mortality and cardiovascular mortality were the same. CONCLUSIONS: There were no differences in all-cause mortality or cardiovascular mortality between the two groups. Rehospitalisation rate for HF decreased in the anti-M2-R (-) group. This difference may be related to the regulation of the autonomic nervous system by anti-M2-R.


Assuntos
Autoanticorpos/sangue , Sistema Nervoso Autônomo/efeitos dos fármacos , Cardiomiopatias/tratamento farmacológico , Fármacos Cardiovasculares/uso terapêutico , Coração/inervação , Complicações Cardiovasculares na Gravidez/tratamento farmacológico , Transtornos Puerperais/tratamento farmacológico , Receptor Muscarínico M2/imunologia , Adulto , Autoimunidade , Sistema Nervoso Autônomo/fisiopatologia , Cardiomiopatias/imunologia , Cardiomiopatias/mortalidade , Cardiomiopatias/fisiopatologia , Feminino , Humanos , Readmissão do Paciente , Período Periparto , Gravidez , Complicações Cardiovasculares na Gravidez/imunologia , Complicações Cardiovasculares na Gravidez/mortalidade , Complicações Cardiovasculares na Gravidez/fisiopatologia , Estudos Prospectivos , Transtornos Puerperais/imunologia , Transtornos Puerperais/mortalidade , Transtornos Puerperais/fisiopatologia , Recuperação de Função Fisiológica , Medição de Risco , Fatores de Risco , Fatores de Tempo , Resultado do Tratamento , Função Ventricular Esquerda/efeitos dos fármacos
5.
Pathol Res Pract ; 227: 153635, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34653913

RESUMO

AIM: Amyloidosis is a systemic or localized disease of protein deposition characterized by amorphous eosinophilic morphology and positivity of Congo Red staining. The typing of amyloidosis is becoming increasingly important because therapeutic agents for each amyloidosis type have been developed. Herein, the authors review the autopsy cases at an institution to reveal the putative Japanese characteristics of each amyloidosis type and evaluate the clinicopathological significance of each type. MATERIALS AND METHODS: A total of 131 autopsy cases of systemic and localized amyloidosis were retrieved for classification by immunohistochemistry. Immunohistochemistry for transthyretin, amyloid A (AA), immunoglobulin light-chain kappa and lambda, and ß2-microglobulin was performed for all cases. RESULTS: The 131 amyloidosis cases were classified as follows: 71 cases (54.2%) of transthyretin amyloidosis, 32 cases (24.4%) of AA amyloidosis, 8 cases (6.1%) of light-chain amyloidosis, and 5 cases (3.8%) of ß2-microglobulin amyloidosis, along with 15 equivocal cases (11.5%). All cases showed myocardial involvement of amyloidosis. Histopathologically, the transthyretin type was significantly associated with the interstitial and nodular patterns, and with the absence of the perivascular and endocardial patterns. The AA type was significantly associated with the perivascular and endocardial patterns, and with the absence of the nodular pattern. CONCLUSION: The authors revealed the putative characteristics of cardiac amyloidosis in Japan by using autopsy cases. About 90% of amyloidosis cases were successfully classified using only commercially available antibodies.


Assuntos
Amiloidose/patologia , Cardiomiopatias/patologia , Imuno-Histoquímica , Miocárdio/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Neuropatias Amiloides Familiares/imunologia , Neuropatias Amiloides Familiares/patologia , Amiloidose/imunologia , Autopsia , Biomarcadores/análise , Cardiomiopatias/imunologia , Feminino , Humanos , Amiloidose de Cadeia Leve de Imunoglobulina/imunologia , Amiloidose de Cadeia Leve de Imunoglobulina/patologia , Cadeias kappa de Imunoglobulina/análise , Cadeias lambda de Imunoglobulina/análise , Japão , Masculino , Pessoa de Meia-Idade , Miocárdio/imunologia , Pré-Albumina/análise , Valor Preditivo dos Testes , Adulto Jovem , Microglobulina beta-2/análise
6.
Front Endocrinol (Lausanne) ; 12: 726967, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34484128

RESUMO

In March 2020, the WHO declared coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a global pandemic. Obesity was soon identified as a risk factor for poor prognosis, with an increased risk of intensive care admissions and mechanical ventilation, but also of adverse cardiovascular events. Obesity is associated with adipose tissue, chronic low-grade inflammation, and immune dysregulation with hypertrophy and hyperplasia of adipocytes and overexpression of pro-inflammatory cytokines. However, to implement appropriate therapeutic strategies, exact mechanisms must be clarified. The role of white visceral adipose tissue, increased in individuals with obesity, seems important, as a viral reservoir for SARS-CoV-2 via angiotensin-converting enzyme 2 (ACE2) receptors. After infection of host cells, the activation of pro-inflammatory cytokines creates a setting conducive to the "cytokine storm" and macrophage activation syndrome associated with progression to acute respiratory distress syndrome. In obesity, systemic viral spread, entry, and prolonged viral shedding in already inflamed adipose tissue may spur immune responses and subsequent amplification of a cytokine cascade, causing worse outcomes. More precisely, visceral adipose tissue, more than subcutaneous fat, could predict intensive care admission; and lower density of epicardial adipose tissue (EAT) could be associated with worse outcome. EAT, an ectopic adipose tissue that surrounds the myocardium, could fuel COVID-19-induced cardiac injury and myocarditis, and extensive pneumopathy, by strong expression of inflammatory mediators that could diffuse paracrinally through the vascular wall. The purpose of this review is to ascertain what mechanisms may be involved in unfavorable prognosis among COVID-19 patients with obesity, especially cardiovascular events, emphasizing the harmful role of excess ectopic adipose tissue, particularly EAT.


Assuntos
COVID-19/metabolismo , Cardiomiopatias/metabolismo , Gordura Intra-Abdominal/metabolismo , Obesidade/metabolismo , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Enzima de Conversão de Angiotensina 2/metabolismo , COVID-19/complicações , COVID-19/imunologia , Cardiomiopatias/imunologia , Cardiomiopatias/patologia , Cardiopatias/imunologia , Cardiopatias/metabolismo , Cardiopatias/patologia , Humanos , Inflamação , Gordura Intra-Abdominal/patologia , Obesidade/complicações , Obesidade/imunologia , Obesidade/patologia , Pericárdio , Prognóstico , SARS-CoV-2/metabolismo , Serina Endopeptidases/metabolismo
7.
Biomed Pharmacother ; 143: 112219, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34560540

RESUMO

Cardiac fibrosis can lead to heart failure, arrhythmia, and sudden cardiac death, representing one of the leading causes of death due to cardiovascular diseases. Cardiac fibrosis involves several multifactorial processes that cannot be effectively controlled by the available therapies. Therefore, current research has focused on the development of novel drugs that can be used to prevent cardiac fibrosis. Recent studies on the functions of inflammasome have provided an in-depth understanding of the regulatory functions of inflammasome in cardiac fibrosis. This review summarizes the latest research on the functions of the NLRP3 inflammasome in various cardiovascular diseases. The latest findings indicate that the NLRP3 inflammasome mediates several inflammatory responses and is associated with pyroptosis, mitochondrial regulation, and myofibroblast differentiation in cardiac fibrosis. These novel findings provide insight into the vital role of the NLRP3 inflammasome in the pathogenesis of cardiac fibrosis, which can be used to identify new targets for its prevention and treatment.


Assuntos
Cardiomiopatias/metabolismo , Inflamassomos/metabolismo , Miocárdio/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Animais , Anti-Inflamatórios/uso terapêutico , Cardiomiopatias/tratamento farmacológico , Cardiomiopatias/imunologia , Cardiomiopatias/patologia , Fibrose , Humanos , Inflamassomos/antagonistas & inibidores , Inflamassomos/imunologia , Mediadores da Inflamação/metabolismo , Mitocôndrias Cardíacas/imunologia , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Cardíacas/patologia , Miocárdio/imunologia , Miocárdio/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Transdução de Sinais
9.
Eur J Pharmacol ; 909: 174438, 2021 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-34437885

RESUMO

Increasing evidence indicates that patients or experimental animals exposure to endotoxin (lipopolysaccharides, LPS) exert deleterious cardiac functions that greatly contribute to morbidity and mortality. The pathophysiologic processes, including NLRP3 inflammasome overactivation and cardiac inflammatory injury, are complicated. Sodium tanshinone IIA sulfonate (STS), a water-soluble derivative of tanshinone IIA, is a naturally occurring compound extracted from Salvia miltiorrhiza and has anti-inflammatory and cardioprotective properties. In this study we examined the effect of STS on endotoxin-induced cardiomyopathy and investigated the underlying mechanisms. An endotoxemic mouse model was established by injecting LPS (10 mg/kg). Different doses of STS were administered intraperitoneally (5, 10, or 50 mg/kg) at different time points (2/12 h, 4/12 h, and 8/12 h) after LPS challenge to assess its effect on survival of mice with endotoxemia. In parallel, cardiac function, myocardial inflammatory cytokines, cardiomyocyte pyroptosis and autophagy were evaluated to determine the extent of myocardial damage due to sepsis in the presence and absence of STS at the optimal dose (10 mg/kg) and time-point (2/12 h). The results demonstrated that STS increased the survival rates, improved the compromised cardiac function and reduced myocardial inflammatory injury associated with enhanced autophagy and mitigated NLRP3 inflammasome activation. Moreover, inhibiting of autophagy or blocking the AMPK pathway reversed STS-elicited prevention of cardiomyopathy and activated the NLRP3 inflammasome in endotoxemic mice. Collectively, our study demonstrates that STS attenuates endotoxemia-induced mortality and cardiomyopathy, which may be associated with promotion of autophagy and inhibition of NLRP3 inflammasome overactivation.


Assuntos
Cardiomiopatias/prevenção & controle , Endotoxemia/tratamento farmacológico , Inflamassomos/antagonistas & inibidores , Fenantrenos/farmacologia , Animais , Autofagia/efeitos dos fármacos , Autofagia/imunologia , Cardiomiopatias/diagnóstico , Cardiomiopatias/imunologia , Cardiomiopatias/microbiologia , Modelos Animais de Doenças , Ecocardiografia , Endotoxemia/complicações , Endotoxemia/imunologia , Endotoxemia/microbiologia , Endotoxinas/sangue , Endotoxinas/imunologia , Ventrículos do Coração/diagnóstico por imagem , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/imunologia , Ventrículos do Coração/patologia , Humanos , Inflamassomos/imunologia , Inflamassomos/metabolismo , Masculino , Camundongos , Miócitos Cardíacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Fenantrenos/uso terapêutico , Piroptose/efeitos dos fármacos , Piroptose/imunologia
10.
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
11.
Viruses ; 13(6)2021 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-34071557

RESUMO

Patients with underlying cardiovascular conditions are particularly vulnerable to severe COVID-19. In this project, we aimed to characterize similarities in dysregulated immune pathways between COVID-19 patients and patients with cardiomyopathy, venous thromboembolism (VTE), or coronary artery disease (CAD). We hypothesized that these similarly dysregulated pathways may be critical to how cardiovascular diseases (CVDs) exacerbate COVID-19. To evaluate immune dysregulation in different diseases, we used four separate datasets, including RNA-sequencing data from human left ventricular cardiac muscle samples of patients with dilated or ischemic cardiomyopathy and healthy controls; RNA-sequencing data of whole blood samples from patients with single or recurrent event VTE and healthy controls; RNA-sequencing data of human peripheral blood mononuclear cells (PBMCs) from patients with and without obstructive CAD; and RNA-sequencing data of platelets from COVID-19 subjects and healthy controls. We found similar immune dysregulation profiles between patients with CVDs and COVID-19 patients. Interestingly, cardiomyopathy patients display the most similar immune landscape to COVID-19 patients. Additionally, COVID-19 patients experience greater upregulation of cytokine- and inflammasome-related genes than patients with CVDs. In all, patients with CVDs have a significant overlap of cytokine- and inflammasome-related gene expression profiles with that of COVID-19 patients, possibly explaining their greater vulnerability to severe COVID-19.


Assuntos
COVID-19/imunologia , COVID-19/fisiopatologia , Cardiomiopatias/imunologia , Doença da Artéria Coronariana/imunologia , Tromboembolia Venosa/imunologia , COVID-19/complicações , COVID-19/genética , Cardiomiopatias/complicações , Cardiomiopatias/genética , Doença da Artéria Coronariana/complicações , Doença da Artéria Coronariana/genética , Citocinas/genética , Conjuntos de Dados como Assunto , Humanos , Hospedeiro Imunocomprometido/genética , Inflamassomos/genética , Contagem de Linfócitos , Gravidade do Paciente , RNA-Seq , Tromboembolia Venosa/complicações
12.
Front Immunol ; 12: 624703, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33692798

RESUMO

Accumulating evidence suggests that the breakdown of immune tolerance plays an important role in the development of myocarditis triggered by cardiotropic microbial infections. Genetic deletion of immune checkpoint molecules that are crucial for maintaining self-tolerance causes spontaneous myocarditis in mice, and cancer treatment with immune checkpoint inhibitors can induce myocarditis in humans. These results suggest that the loss of immune tolerance results in myocarditis. The tissue microenvironment influences the local immune dysregulation in autoimmunity. Recently, tenascin-C (TN-C) has been found to play a role as a local regulator of inflammation through various molecular mechanisms. TN-C is a nonstructural extracellular matrix glycoprotein expressed in the heart during early embryonic development, as well as during tissue injury or active tissue remodeling, in a spatiotemporally restricted manner. In a mouse model of autoimmune myocarditis, TN-C was detectable before inflammatory cell infiltration and myocytolysis became histologically evident; it was strongly expressed during active inflammation and disappeared with healing. TN-C activates dendritic cells to generate pathogenic autoreactive T cells and forms an important link between innate and acquired immunity.


Assuntos
Doenças Autoimunes/metabolismo , Autoimunidade , Cardiomiopatias/metabolismo , Mediadores da Inflamação/metabolismo , Miocardite/metabolismo , Miocárdio/metabolismo , Tenascina/metabolismo , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Cardiomiopatias/imunologia , Cardiomiopatias/patologia , Microambiente Celular , Humanos , Miocardite/imunologia , Miocardite/patologia , Miocárdio/imunologia , Miocárdio/patologia , Tolerância a Antígenos Próprios , Transdução de Sinais
13.
Inflamm Res ; 70(4): 379-387, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33683374

RESUMO

INTRODUCTION: Sepsis-induced myocardial dysfunction (SIMD) is a condition manifested by an intrinsic myocardial systolic and diastolic dysfunction during sepsis, which is associated with worse clinical outcomes and a higher mortality. MATERIALS AND METHODS: Several pathophysiological mechanisms including mitochondrial dysfunction, abnormal body immune reaction, metabolic reprogramming, excessive production of reactive oxygen species (ROS), and disorder of calcium regulation have been involved in SIMD. Mitophagy has potential role in protecting myocardial cells in sepsis, especially in survivors. CONCLUSION: In the current review, we focus on the role of mitochondrial dysfunction and other mitochondria-related mechanisms including immunologic imbalance, energetic reprogramming, mitophagy, and pyroptosis in the mechanisms of SIMD.


Assuntos
Cardiomiopatias/etiologia , Mitocôndrias , Sepse/complicações , Animais , Cardiomiopatias/imunologia , Cardiomiopatias/metabolismo , Humanos , Tolerância Imunológica , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Mitofagia , Miocárdio , Piroptose , Sepse/imunologia , Sepse/metabolismo
14.
Mol Biol Cell ; 32(7): 622-633, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33534612

RESUMO

Dysregulation of immune responses has been linked to the generation of immunoglobulin G (IgG) autoantibodies that target human ß1ARs and contribute to deleterious cardiac outcomes. Given the benefits of ß-blockers observed in patients harboring the IgG3 subclass of autoantibodies, we investigated the role of these autoantibodies in human ß1AR function. Serum and purified IgG3(+) autoantibodies from patients with onset of cardiomyopathy were tested using human embryonic kidney (HEK) 293 cells expressing human ß1ARs. Unexpectedly, pretreatment of cells with IgG3(+) serum or purified IgG3(+) autoantibodies impaired dobutamine-mediated adenylate cyclase (AC) activity and cyclic adenosine monophosphate (cAMP) generation while enhancing biased ß-arrestin recruitment and Extracellular Regulated Kinase (ERK) activation. In contrast, the ß-blocker metoprolol increased AC activity and cAMP in the presence of IgG3(+) serum or IgG3(+) autoantibodies. Because IgG3(+) autoantibodies are specific to human ß1ARs, non-failing human hearts were used as an endogenous system to determine their ability to bias ß1AR signaling. Consistently, metoprolol increased AC activity, reflecting the ability of the IgG3(+) autoantibodies to bias ß-blocker toward G-protein coupling. Importantly, IgG3(+) autoantibodies are specific toward ß1AR as they did not alter ß2AR signaling. Thus, IgG3(+) autoantibody biases ß-blocker toward G-protein coupling while impairing agonist-mediated G-protein activation but promoting G-protein-independent ERK activation. This phenomenon may underlie the beneficial outcomes observed in patients harboring IgG3(+) ß1AR autoantibodies.


Assuntos
Autoanticorpos/imunologia , Imunoglobulina G/imunologia , Receptores Adrenérgicos beta 1/imunologia , Autoanticorpos/sangue , Cardiomiopatias/imunologia , Cardiomiopatias/fisiopatologia , AMP Cíclico , Células HEK293 , Coração/fisiologia , Humanos , Imunoglobulina G/metabolismo , Receptores Adrenérgicos/imunologia , Receptores Adrenérgicos beta 1/metabolismo , Transdução de Sinais , beta-Arrestinas
15.
PLoS Biol ; 19(1): e3001062, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33395408

RESUMO

Lyme carditis is an extracutaneous manifestation of Lyme disease characterized by episodes of atrioventricular block of varying degrees and additional, less reported cardiomyopathies. The molecular changes associated with the response to Borrelia burgdorferi over the course of infection are poorly understood. Here, we identify broad transcriptomic and proteomic changes in the heart during infection that reveal a profound down-regulation of mitochondrial components. We also describe the long-term functional modulation of macrophages exposed to live bacteria, characterized by an augmented glycolytic output, increased spirochetal binding and internalization, and reduced inflammatory responses. In vitro, glycolysis inhibition reduces the production of tumor necrosis factor (TNF) by memory macrophages, whereas in vivo, it produces the reversion of the memory phenotype, the recovery of tissue mitochondrial components, and decreased inflammation and spirochetal burdens. These results show that B. burgdorferi induces long-term, memory-like responses in macrophages with tissue-wide consequences that are amenable to be manipulated in vivo.


Assuntos
Borrelia burgdorferi/imunologia , Cardiomiopatias/etiologia , Memória Imunológica , Doença de Lyme/imunologia , Macrófagos/fisiologia , Animais , Cardiomiopatias/imunologia , Cardiomiopatias/microbiologia , Cardiomiopatias/patologia , Células Cultivadas , Endocardite Bacteriana/complicações , Endocardite Bacteriana/imunologia , Endocardite Bacteriana/microbiologia , Endocardite Bacteriana/patologia , Feminino , Células HEK293 , Coração/microbiologia , Humanos , Doença de Lyme/patologia , Ativação de Macrófagos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/imunologia , Miócitos Cardíacos/microbiologia , Miócitos Cardíacos/patologia , Células RAW 264.7
16.
Nat Rev Cardiol ; 18(3): 169-193, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33046850

RESUMO

Inflammatory cardiomyopathy, characterized by inflammatory cell infiltration into the myocardium and a high risk of deteriorating cardiac function, has a heterogeneous aetiology. Inflammatory cardiomyopathy is predominantly mediated by viral infection, but can also be induced by bacterial, protozoal or fungal infections as well as a wide variety of toxic substances and drugs and systemic immune-mediated diseases. Despite extensive research, inflammatory cardiomyopathy complicated by left ventricular dysfunction, heart failure or arrhythmia is associated with a poor prognosis. At present, the reason why some patients recover without residual myocardial injury whereas others develop dilated cardiomyopathy is unclear. The relative roles of the pathogen, host genomics and environmental factors in disease progression and healing are still under discussion, including which viruses are active inducers and which are only bystanders. As a consequence, treatment strategies are not well established. In this Review, we summarize and evaluate the available evidence on the pathogenesis, diagnosis and treatment of myocarditis and inflammatory cardiomyopathy, with a special focus on virus-induced and virus-associated myocarditis. Furthermore, we identify knowledge gaps, appraise the available experimental models and propose future directions for the field. The current knowledge and open questions regarding the cardiovascular effects associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are also discussed. This Review is the result of scientific cooperation of members of the Heart Failure Association of the ESC, the Heart Failure Society of America and the Japanese Heart Failure Society.


Assuntos
Cardiomiopatias/fisiopatologia , Inflamação/fisiopatologia , Miocardite/fisiopatologia , Viroses/fisiopatologia , Animais , Antivirais/uso terapêutico , Autoimunidade/imunologia , Biópsia , COVID-19/fisiopatologia , COVID-19/terapia , Cardiomiopatias/diagnóstico , Cardiomiopatias/imunologia , Cardiomiopatias/terapia , Cardiomiopatia Dilatada , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/fisiopatologia , Infecções por Coronavirus/terapia , Infecções por Coxsackievirus/imunologia , Infecções por Coxsackievirus/fisiopatologia , Infecções por Coxsackievirus/terapia , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/fisiopatologia , Infecções por Citomegalovirus/terapia , Modelos Animais de Doenças , Infecções por Echovirus/imunologia , Infecções por Echovirus/fisiopatologia , Infecções por Echovirus/terapia , Infecções por Vírus Epstein-Barr/imunologia , Infecções por Vírus Epstein-Barr/fisiopatologia , Infecções por Vírus Epstein-Barr/terapia , Eritema Infeccioso/imunologia , Eritema Infeccioso/fisiopatologia , Eritema Infeccioso/terapia , Infecções por HIV/fisiopatologia , Hepatite C/imunologia , Hepatite C/fisiopatologia , Hepatite C/terapia , Humanos , Imunoglobulinas Intravenosas/uso terapêutico , Fatores Imunológicos/uso terapêutico , Inflamação/diagnóstico , Inflamação/imunologia , Inflamação/terapia , Influenza Humana/imunologia , Influenza Humana/fisiopatologia , Influenza Humana/terapia , Leucócitos/imunologia , Miocardite/diagnóstico , Miocardite/imunologia , Miocardite/terapia , Miocárdio/patologia , Prognóstico , Infecções por Roseolovirus/imunologia , Infecções por Roseolovirus/fisiopatologia
17.
J Cardiovasc Transl Res ; 14(1): 75-87, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32440911

RESUMO

Myocarditis is a multifactorial disorder, characterized by an inflammatory reaction in the myocardium, predominantly triggered by infectious agents, but also by antigen mimicry or autoimmunity in susceptible individuals. Unless spontaneously resolved, a chronic inflammatory course concludes with cardiac muscle dysfunction portrayed by ventricular dilatation, clinically termed inflammatory cardiomyopathy (Infl-CM). Treatment strategies aim to resolve chronic inflammation and preserve cardiac function. Beside standard heart failure treatments, which only play a supportive role in this condition, systemic immunosuppressants are used to diminish inflammatory cell function at the cost of noxious side effects. To date, the treatment protocols are expert-based without large clinical evidence. This review describes concept and contemporary strategies to alleviate myocardial inflammation and sheds light on potential inflammatory targets in an evidence-based order.


Assuntos
Autoimunidade , Cardiomiopatias/tratamento farmacológico , Imunossupressores/uso terapêutico , Inflamação/tratamento farmacológico , Miocárdio/imunologia , Cardiomiopatias/imunologia , Humanos , Inflamação/imunologia
18.
Int Immunopharmacol ; 90: 107160, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33243604

RESUMO

Septic cardiomyopathy (SCM)is common in septic patients and results in cardiovascular failure. The pathogenesis of SCM is complicated, and patients with SCM have high mortality because current treatment methods are limited. The cholinergic anti-inflammatory pathway (CAP) modulates inflammatory responses through vagus nerve stimulation that leads to the release of acetylcholine (ACh), which binds to the alpha7 nicotinic acetylcholine receptor (α7nAChR). Moreover, α7nAChR activation by its agonists at the tissue level inhibits inflammatory mediators and regulates the function of immune cells in sepsis. Therefore, the α7nAChR can maintain balance of the inflammatory-immune response in sepsis. CAP has been elucidated as a critical regulator of anti-inflammation in many diseases, including rheumatoid arthritis, inflammatory boweldisease and SCM. Additionally, some clinical and preclinical trials show therapeutic potential via regulating CAP. There are excellent studies regarding the beneficial role of CAP activation, especially α7nAChR, in experimental SCM. This review aims to discuss the CAP in attenuating inflammation and the potential role of α7nAChR activation in regulating immune and reducing inflammation in SCM.


Assuntos
Cardiomiopatias/imunologia , Cardiomiopatias/metabolismo , Neuroimunomodulação/imunologia , Neuroimunomodulação/fisiologia , Animais , Humanos , Inflamação/metabolismo , Camundongos , Ratos , Sepse , Receptor Nicotínico de Acetilcolina alfa7/metabolismo
19.
Cardiovasc Pathol ; 51: 107315, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33264681

RESUMO

There are few reports on the coexistence of cardiac amyloid light-chain (AL) amyloidosis and light chain deposition disease (LCDD), despite their similar pathophysiologies caused by plasma-cell dyscrasia. Herein, we report the coexistence of these diseases. A 59-year-old man was referred to our hospital because of exertional dyspnea and hypotension. Renal dysfunction of unknown etiology had been present for 4 years and hemodialysis had been introduced. Severe systolic and diastolic cardiac dysfunction was apparent, accompanied with dilatation and granular sparkling, but not with left ventricular hypertrophy. The plasma-free light chain κ was found to be extremely high, with a κ/λ ratio of 1,919. Light microscopic examination of the endomyocardial biopsy revealed spotty and homogenous deposits, which positively stained with Congo red, and exhibited a blazing apple-green color under polarized light. Based on these results, cardiac amyloidosis was diagnosed. In specimens prepared for electron microscopy, no amyloid fibrils could be found. Instead, we observed amorphous nonfibrillar deposits around several small vessels including capillaries and small arteries, which were consistent with light-chain deposits. LCDD was diagnosed based on the systemic increase in κ light chain and the ultrastructural findings of the endomyocardial biopsy specimens. Coexistence of cardiac amyloidosis and LCDD was thus confirmed in our patient. An electron microscopic assessment in addition to Congo red staining may be useful to diagnose latent LCDD in patients with suspected cardiac light-chain amyloidosis.


Assuntos
Cardiomiopatias/patologia , Cadeias Leves de Imunoglobulina/ultraestrutura , Amiloidose de Cadeia Leve de Imunoglobulina/patologia , Miocárdio/ultraestrutura , Biópsia , Cardiomiopatias/imunologia , Cardiomiopatias/terapia , Evolução Fatal , Humanos , Amiloidose de Cadeia Leve de Imunoglobulina/imunologia , Amiloidose de Cadeia Leve de Imunoglobulina/terapia , Masculino , Microscopia Eletrônica , Pessoa de Meia-Idade , Miocárdio/imunologia
20.
Rheumatology (Oxford) ; 60(8): 3809-3816, 2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-33369674

RESUMO

OBJECTIVE: Myocardial involvement (MCI) is known to increase morbidity and mortality in polymyositis (PM) and dermatomyositis (DM). This study aims to investigate whether complicating with ventricular arrhythmia (VA) predicts poor outcomes in patients with PM/DM-related myocardial involvement (PM/DM-MCI). METHODS: We reviewed all PM/DM-MCI patients admitted to Peking Union Medical College Hospital from October 1997 to April 2019. VA and the other possible risk factors for the composite endpoint, including death from any cause and rehospitalization for cardiac causes, were analyzed. RESULTS: A total of 75 PM/DM-MCI patients (44 PM and 31 DM) were enrolled, of which 27 (36%) met the composite endpoint during a median follow-up of 24 months. Independent prognostic factors for the composite endpoint include VA [HR 4.215, 95% CI (1.737, 10.230)], NT-proBNP > 3415 pg/ml [HR 2.606, 95% CI (1.203, 5.646)], interstitial lung disease [HR 2.688, 95% CI (1.209, 5.978)], and anti-cardiac remodelling therapy [HR 0.302, 95% CI (0.115, 0.792)]. The 3-year event-free survival rate of patients without VA was significantly higher than that of patients with VA (63.3% vs 40.7%, P = 0.034). Skin lesions [OR 0.163, 95% CI (0.051, 0.523)] and positive antimitochondrial antibody [OR 3.484, 95% CI (1.192, 10.183)] were independent predictors of VA. CONCLUSION: VA provides prognostic insights for PM/DM-MCI patients and predicts poor outcome. Polymyositis and positive antimitochondrial antibody are closely associated with the presence of VA in PM/DM-MCI.


Assuntos
Arritmias Cardíacas/fisiopatologia , Cardiomiopatias/fisiopatologia , Dermatomiosite/fisiopatologia , Antagonistas Adrenérgicos beta/uso terapêutico , Adulto , Antagonistas de Receptores de Angiotensina/uso terapêutico , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Arritmias Cardíacas/epidemiologia , Autoanticorpos/imunologia , Cardiomiopatias/tratamento farmacológico , Cardiomiopatias/epidemiologia , Cardiomiopatias/imunologia , Dermatomiosite/tratamento farmacológico , Dermatomiosite/epidemiologia , Dermatomiosite/imunologia , Feminino , Humanos , Doenças Pulmonares Intersticiais/epidemiologia , Masculino , Pessoa de Meia-Idade , Antagonistas de Receptores de Mineralocorticoides/uso terapêutico , Mitocôndrias/imunologia , Peptídeo Natriurético Encefálico/sangue , Fragmentos de Peptídeos/sangue , Polimiosite/tratamento farmacológico , Polimiosite/epidemiologia , Polimiosite/imunologia , Polimiosite/fisiopatologia , Prognóstico , Intervalo Livre de Progressão , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Espironolactona/uso terapêutico , Taxa de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...