Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Front Immunol ; 15: 1396800, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39100680

RESUMO

Introduction: Bone marrow embolization may complicate orthopedic surgery, potentially causing fat embolism syndrome. The inflammatory potential of bone marrow emboli is unclear. We aimed to investigate the inflammatory response to femoral intramedullary nailing, specifically the systemic inflammatory effects in plasma, and local tissue responses. Additionally, the plasma response was compared to that following intravenous injection of autologous bone marrow. Methods: Twelve pigs underwent femoral nailing (previously shown to have fat emboli in lung and heart), four received intravenous bone marrow, and four served as sham controls. Blood samples were collected hourly and tissue samples postmortem. Additionally, we incubated bone marrow and blood, separately and in combination, from six pigs in vitro. Complement activation was detected by C3a and the terminal C5b-9 complement complex (TCC), and the cytokines TNF, IL-1ß, IL-6 and IL-10 as well as the thrombin-antithrombin complexes (TAT) were all measured using enzyme-immunoassays. Results: After nailing, plasma IL-6 rose 21-fold, compared to a 4-fold rise in sham (p=0.0004). No plasma differences in the rest of the inflammatory markers were noted across groups. However, nailing yielded 2-3-times higher C3a, TCC, TNF, IL-1ß and IL-10 in lung tissue compared to sham (p<0.0001-0.03). Similarly, heart tissue exhibited 2-times higher TCC and IL-1ß compared to sham (p<0.0001-0.03). Intravenous bone marrow yielded 8-times higher TAT than sham at 30 minutes (p<0.0001). In vitro, incubation of bone marrow for four hours resulted in 95-times higher IL-6 compared to whole blood (p=0.03). Discussion: A selective increase in plasma IL-6 was observed following femoral nailing, whereas lung and heart tissues revealed a broad local inflammatory response not reflected systemically. In vitro experiments may imply bone marrow to be the primary IL-6 source.


Assuntos
Embolia Gordurosa , Interleucina-6 , Pulmão , Animais , Suínos , Interleucina-6/sangue , Embolia Gordurosa/etiologia , Embolia Gordurosa/sangue , Embolia Gordurosa/imunologia , Pulmão/imunologia , Pulmão/patologia , Pulmão/metabolismo , Medula Óssea/metabolismo , Fixação Intramedular de Fraturas/efeitos adversos , Miocárdio/metabolismo , Miocárdio/patologia , Miocárdio/imunologia , Inflamação/sangue , Inflamação/imunologia , Feminino , Citocinas/sangue , Citocinas/metabolismo , Pinos Ortopédicos , Ativação do Complemento , Fêmur/metabolismo , Modelos Animais de Doenças
2.
Nat Commun ; 15(1): 6480, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-39090108

RESUMO

Regulatory T cells (Tregs) are key immune regulators that have shown promise in enhancing cardiac repair post-MI, although the mechanisms remain elusive. Here, we show that rapidly increasing Treg number in the circulation post-MI via systemic administration of exogenous Tregs improves cardiac function in male mice, by limiting cardiomyocyte death and reducing fibrosis. Mechanistically, exogenous Tregs quickly home to the infarcted heart and adopt an injury-specific transcriptome that mediates repair by modulating monocytes/macrophages. Specially, Tregs lead to a reduction in pro-inflammatory Ly6CHi CCR2+ monocytes/macrophages accompanied by a rapid shift of macrophages towards a pro-repair phenotype. Additionally, exogenous Treg-derived factors, including nidogen-1 and IL-10, along with a decrease in cardiac CD8+ T cell number, mediate the reduction of the pro-inflammatory monocyte/macrophage subset in the heart. Supporting the pivotal role of IL-10, exogenous Tregs knocked out for IL-10 lose their pro-repair capabilities. Together, this study highlights the beneficial use of a Treg-based therapeutic approach for cardiac repair with important mechanistic insights that could facilitate the development of novel immunotherapies for MI.


Assuntos
Interleucina-10 , Macrófagos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio , Linfócitos T Reguladores , Animais , Infarto do Miocárdio/imunologia , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Linfócitos T Reguladores/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Interleucina-10/metabolismo , Interleucina-10/genética , Fenótipo , Miocárdio/patologia , Miocárdio/imunologia , Miocárdio/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/imunologia , Fibrose , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Camundongos Knockout
3.
Front Immunol ; 15: 1371477, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39007149

RESUMO

Background: A high-fat diet (HFD) contributes to various metabolic disorders and obesity, which are major contributors to cardiovascular disease. As an essential regulator for heart homeostasis, cardiac resident macrophages may go awry and contribute to cardiac pathophysiology upon HFD. Thus, to better understand how HFD induced cardiac dysfunction, this study intends to explore the transcriptional and functional changes in cardiac resident macrophages of HFD mice. Methods: C57BL/6J female mice that were 6 weeks old were fed with HFD or normal chow diet (NCD) for 16 weeks. After an evaluation of cardiac functions by echocardiography, mouse hearts were harvested and cardiac resident CCR2- macrophages were sorted, followed by Smart sequencing. Bioinformatics analysis including GO, KEGG, and GSEA analyses were employed to elucidate transcriptional and functional changes. Results: Hyperlipidemia and obesity were observed easily upon HFD. The mouse hearts also displayed more severe fibrosis and diastolic dysfunction in HFD mice. Smart sequencing and functional analysis revealed metabolic dysfunctions, especially lipid-related genes and pathways. Besides this, antigen-presentation-related gene such as Ctsf and inflammation, particularly for NF-κB signaling and complement cascades, underwent drastic changes in cardiac resident macrophages. GO cellular compartment analysis was also performed and showed specific organelle enrichment trends of the involved genes. Conclusion: Dysregulated metabolism intertwines with inflammation in cardiac resident macrophages upon HFD feeding in mice, and further research on crosstalk among organelles could shed more light on potential mechanisms.


Assuntos
Dieta Hiperlipídica , Macrófagos , Camundongos Endogâmicos C57BL , Miocárdio , Animais , Dieta Hiperlipídica/efeitos adversos , Camundongos , Macrófagos/imunologia , Macrófagos/metabolismo , Feminino , Miocárdio/metabolismo , Miocárdio/imunologia , Obesidade/imunologia , Obesidade/metabolismo , Hiperlipidemias/imunologia , Hiperlipidemias/metabolismo
4.
Heart Fail Rev ; 29(5): 1049-1063, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38985383

RESUMO

Improvements in therapies for heart failure with preserved ejection fraction (HFpEF) are crucial for improving patient outcomes and quality of life. Although HFpEF is the predominant heart failure type among older individuals, its prognosis is often poor owing to the lack of effective therapies. The roles of the spleen and bone marrow are often overlooked in the context of HFpEF. Recent studies suggest that the spleen and bone marrow could play key roles in HFpEF, especially in relation to inflammation and immune responses. The bone marrow can increase production of certain immune cells that can migrate to the heart and contribute to disease. The spleen can contribute to immune responses that either protect or exacerbate heart failure. Extramedullary hematopoiesis in the spleen could play a crucial role in HFpEF. Increased metabolic activity in the spleen, immune cell production and mobilization to the heart, and concomitant cytokine production may occur in heart failure. This leads to systemic chronic inflammation, along with an imbalance of immune cells (macrophages) in the heart, resulting in chronic inflammation and progressive fibrosis, potentially leading to decreased cardiac function. The bone marrow and spleen are involved in altered iron metabolism and anemia, which also contribute to HFpEF. This review presents the concept of an interplay between the heart, spleen, and bone marrow in the setting of HFpEF, with a particular focus on extramedullary hematopoiesis in the spleen. The aim of this review is to discern whether the spleen can serve as a new therapeutic target for HFpEF.


Assuntos
Medula Óssea , Insuficiência Cardíaca , Hematopoese Extramedular , Baço , Humanos , Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/metabolismo , Hematopoese Extramedular/fisiologia , Baço/imunologia , Baço/metabolismo , Volume Sistólico/fisiologia , Miocárdio/metabolismo , Miocárdio/patologia , Miocárdio/imunologia , Inflamação
5.
Int J Mol Sci ; 25(11)2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38891767

RESUMO

Myocarditis is characterized by an influx of inflammatory cells, predominantly of myeloid lineage. The progression of myocarditis to a dilated cardiomyopathy is markedly influenced by TGF-ß signalling. Here, we investigate the role of TGF-ß signalling in inflammatory cardiac macrophages in the development of myocarditis and post-inflammatory fibrosis. Experimental autoimmune myocarditis (EAM) was induced in the LysM-Cre × R26-stop-EYFP × Tgfbr2-fl/fl transgenic mice showing impaired TGF-ß signalling in the myeloid lineage and the LysM-Cre × R26-stop-EYFP control mice. In EAM, immunization led to acute myocarditis on day 21, followed by cardiac fibrosis on day 40. Both strains showed a similar severity of myocarditis and the extent of cardiac fibrosis. On day 21 of EAM, an increase in cardiac inflammatory macrophages was observed in both strains. These cells were sorted and analysed for differential gene expression using whole-genome transcriptomics. The analysis revealed activation and regulation of the inflammatory response, particularly the production of both pro-inflammatory and anti-inflammatory cytokines and cytokine receptors as TGF-ß-dependent processes. The analysis of selected cytokines produced by bone marrow-derived macrophages confirmed their suppressed secretion. In conclusion, our findings highlight the regulatory role of TGF-ß signalling in cytokine production within inflammatory cardiac macrophages during myocarditis.


Assuntos
Doenças Autoimunes , Citocinas , Macrófagos , Camundongos Transgênicos , Miocardite , Transdução de Sinais , Fator de Crescimento Transformador beta , Animais , Miocardite/metabolismo , Miocardite/imunologia , Miocardite/patologia , Miocardite/etiologia , Fator de Crescimento Transformador beta/metabolismo , Camundongos , Macrófagos/metabolismo , Macrófagos/imunologia , Doenças Autoimunes/metabolismo , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Citocinas/metabolismo , Modelos Animais de Doenças , Miocárdio/metabolismo , Miocárdio/patologia , Miocárdio/imunologia , Fibrose , Masculino
6.
Circ Res ; 134(12): 1824-1840, 2024 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-38843291

RESUMO

Immunometabolism is an emerging field at the intersection of immunology and metabolism. Immune cell activation plays a critical role in the pathogenesis of cardiovascular diseases and is integral for regeneration during cardiac injury. We currently possess a limited understanding of the processes governing metabolic interactions between immune cells and cardiomyocytes. The impact of this intercellular crosstalk can manifest as alterations to the steady state flux of metabolites and impact cardiac contractile function. Although much of our knowledge is derived from acute inflammatory response, recent work emphasizes heterogeneity and flexibility in metabolism between cardiomyocytes and immune cells during pathological states, including ischemic, cardiometabolic, and cancer-associated disease. Metabolic adaptation is crucial because it influences immune cell activation, cytokine release, and potential therapeutic vulnerabilities. This review describes current concepts about immunometabolic regulation in the heart, focusing on intercellular crosstalk and intrinsic factors driving cellular regulation. We discuss experimental approaches to measure the cardio-immunologic crosstalk, which are necessary to uncover unknown mechanisms underlying the immune and cardiac interface. Deeper insight into these axes holds promise for therapeutic strategies that optimize cardioimmunology crosstalk for cardiac health.


Assuntos
Miócitos Cardíacos , Humanos , Animais , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/imunologia , Metabolismo Energético , Cardiomiopatias/metabolismo , Cardiomiopatias/imunologia , Miocárdio/metabolismo , Miocárdio/imunologia , Miocárdio/patologia
8.
Cardiovasc Diabetol ; 23(1): 169, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38750502

RESUMO

Diabetic heart disease (DHD) is a serious complication in patients with diabetes. Despite numerous studies on the pathogenic mechanisms and therapeutic targets of DHD, effective means of prevention and treatment are still lacking. The pathogenic mechanisms of DHD include cardiac inflammation, insulin resistance, myocardial fibrosis, and oxidative stress. Macrophages, the primary cells of the human innate immune system, contribute significantly to these pathological processes, playing an important role in human disease and health. Therefore, drugs targeting macrophages hold great promise for the treatment of DHD. In this review, we examine how macrophages contribute to the development of DHD and which drugs could potentially be used to target macrophages in the treatment of DHD.


Assuntos
Cardiomiopatias Diabéticas , Macrófagos , Estresse Oxidativo , Transdução de Sinais , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Cardiomiopatias Diabéticas/imunologia , Cardiomiopatias Diabéticas/metabolismo , Cardiomiopatias Diabéticas/tratamento farmacológico , Cardiomiopatias Diabéticas/etiologia , Animais , Estresse Oxidativo/efeitos dos fármacos , Fibrose , Anti-Inflamatórios/uso terapêutico , Miocárdio/patologia , Miocárdio/metabolismo , Miocárdio/imunologia , Resistência à Insulina , Mediadores da Inflamação/metabolismo , Terapia de Alvo Molecular
9.
Front Immunol ; 15: 1327372, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38736889

RESUMO

Introduction: Growing evidence from animal models indicates that the myocardium hosts a population of B cells that play a role in the development of cardiomyopathy. However, there is minimal data on human myocardial B cells in the context of cardiomyopathy. Methods: We integrated single-cell and single-nuclei datasets from 45 healthy human hearts, 70 hearts with dilated cardiomyopathy (DCM), and 8 hearts with arrhythmogenic right ventricular cardiomyopathy (ARVC). Interactions between B cells and other cell types were investigated using the CellChat Package. Differential gene expression analysis comparing B cells across conditions was performed using DESeq2. Pathway analysis was performed using Ingenuity, KEGG, and GO pathways analysis. Results: We identified 1,100 B cells, including naive B cells and plasma cells. Cells showed an extensive network of interactions within the healthy myocardium that included outgoing signaling to macrophages, T cells, endothelial cells, and pericytes, and incoming signaling from endothelial cells, pericytes, and fibroblasts. This niche relied on ECM-receptor, contact, and paracrine interactions; and changed significantly in the context of cardiomyopathy, displaying disease-specific features. Differential gene expression analysis showed that in the context of DCM both naive and plasma B cells upregulated several pathways related to immune activation, including upregulation of oxidative phosphorylation, upregulation of leukocyte extravasation, and, in naive B cells, antigen presentation. Discussion: The human myocardium contains naive B cells and plasma cells, integrated into a diverse and dynamic niche that has distinctive features in healthy, DCM, and ARVC. Naive myocardial-associated B cells likely contribute to the pathogenesis of human DCM.


Assuntos
Displasia Arritmogênica Ventricular Direita , Linfócitos B , Cardiomiopatia Dilatada , Miocárdio , Humanos , Cardiomiopatia Dilatada/imunologia , Cardiomiopatia Dilatada/genética , Displasia Arritmogênica Ventricular Direita/genética , Displasia Arritmogênica Ventricular Direita/metabolismo , Linfócitos B/imunologia , Linfócitos B/metabolismo , Miocárdio/metabolismo , Miocárdio/imunologia , Miocárdio/patologia , Masculino , Feminino , Comunicação Celular/imunologia , Perfilação da Expressão Gênica , Pessoa de Meia-Idade , Adulto , Transcriptoma , Regulação da Expressão Gênica
10.
Elife ; 122024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38775664

RESUMO

Cardiac macrophages are heterogenous in phenotype and functions, which has been associated with differences in their ontogeny. Despite extensive research, our understanding of the precise role of different subsets of macrophages in ischemia/reperfusion (I/R) injury remains incomplete. We here investigated macrophage lineages and ablated tissue macrophages in homeostasis and after I/R injury in a CSF1R-dependent manner. Genomic deletion of a fms-intronic regulatory element (FIRE) in the Csf1r locus resulted in specific absence of resident homeostatic and antigen-presenting macrophages, without affecting the recruitment of monocyte-derived macrophages to the infarcted heart. Specific absence of homeostatic, monocyte-independent macrophages altered the immune cell crosstalk in response to injury and induced proinflammatory neutrophil polarization, resulting in impaired cardiac remodeling without influencing infarct size. In contrast, continuous CSF1R inhibition led to depletion of both resident and recruited macrophage populations. This augmented adverse remodeling after I/R and led to an increased infarct size and deterioration of cardiac function. In summary, resident macrophages orchestrate inflammatory responses improving cardiac remodeling, while recruited macrophages determine infarct size after I/R injury. These findings attribute distinct beneficial effects to different macrophage populations in the context of myocardial infarction.


Assuntos
Macrófagos , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos , Animais , Macrófagos/imunologia , Camundongos , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Isquemia Miocárdica/imunologia , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/imunologia , Masculino , Traumatismo por Reperfusão Miocárdica/imunologia , Traumatismo por Reperfusão Miocárdica/patologia , Camundongos Endogâmicos C57BL , Miocárdio/patologia , Miocárdio/imunologia , Modelos Animais de Doenças
11.
J Am Heart Assoc ; 13(9): e032172, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38700022

RESUMO

BACKGROUND: The purpose of this study was to investigate a therapeutic approach targeting the inflammatory response and consequent remodeling from ischemic myocardial injury. METHODS AND RESULTS: Coronary thrombus aspirates were collected from patients at the time of ST-segment-elevation myocardial infarction and subjected to array-based proteome analysis. Clinically indistinguishable at myocardial infarction (MI), patients were stratified into vulnerable and resilient on the basis of 1-year left ventricular ejection fraction and death. Network analysis from coronary aspirates revealed prioritization of tumor necrosis factor-α signaling in patients with worse clinical outcomes. Infliximab, a tumor necrosis factor-α inhibitor, was infused intravenously at reperfusion in a porcine MI model to assess whether infliximab-mediated immune modulation impacts post-MI injury. At 3 days after MI (n=7), infliximab infusion increased proregenerative M2 macrophages in the myocardial border zone as quantified by immunofluorescence (24.1%±23.3% in infliximab versus 9.29%±8.7% in sham; P<0.01). Concomitantly, immunoassays of coronary sinus samples quantified lower troponin I levels (41.72±7.34 pg/mL versus 58.11±10.75 pg/mL; P<0.05) and secreted protein analysis revealed upregulation of injury-modifying interleukin-2, -4, -10, -12, and -18 cytokines in the infliximab-treated cohort. At 4 weeks (n=12), infliximab treatment resulted in significant protective influence, improving left ventricular ejection fraction (53.9%±5.4% versus 36.2%±5.3%; P<0.001) and reducing scar size (8.31%±10.9% versus 17.41%±12.5%; P<0.05). CONCLUSIONS: Profiling of coronary thrombus aspirates in patients with ST-segment-elevation MI revealed highest association for tumor necrosis factor-α in injury risk. Infliximab-mediated immune modulation offers an actionable pathway to alter MI-induced inflammatory response, preserving contractility and limiting adverse structural remodeling.


Assuntos
Modelos Animais de Doenças , Infliximab , Remodelação Ventricular , Infliximab/uso terapêutico , Infliximab/farmacologia , Animais , Humanos , Masculino , Pessoa de Meia-Idade , Remodelação Ventricular/efeitos dos fármacos , Feminino , Infarto do Miocárdio com Supradesnível do Segmento ST/tratamento farmacológico , Infarto do Miocárdio com Supradesnível do Segmento ST/imunologia , Função Ventricular Esquerda/efeitos dos fármacos , Suínos , Idoso , Fator de Necrose Tumoral alfa/metabolismo , Volume Sistólico/efeitos dos fármacos , Trombose Coronária/prevenção & controle , Trombose Coronária/tratamento farmacológico , Miocárdio/patologia , Miocárdio/metabolismo , Miocárdio/imunologia , Troponina I/sangue , Troponina I/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo
12.
Cell Signal ; 119: 111169, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38599440

RESUMO

Cardiac resident macrophages (CRMs) are essential in maintaining the balance of the immune homeostasis in the heart. One of the main factors in the progression of cardiovascular diseases, such as myocarditis, myocardial infarction(MI), and heart failure(HF), is the imbalance in the regulatory mechanisms of CRMs. Recent studies have reported novel heterogeneity and spatiotemporal complexity of CRMs, and their role in maintaining cardiac immune homeostasis and treating cardiovascular diseases. In this review, we focus on the functions of CRMs, including immune surveillance, immune phagocytosis, and immune metabolism, and explore the impact of CRM's homeostasis imbalance on cardiac injury and cardiac repair. We also discuss the therapeutic approaches linked to CRMs. The immunomodulatory strategies targeting CRMs may be a therapeutic approach for the treatment of cardiovascular disease.


Assuntos
Homeostase , Macrófagos , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Animais , Miocárdio/imunologia , Miocárdio/metabolismo , Miocárdio/patologia , Fagocitose , Doenças Cardiovasculares/imunologia , Doenças Cardiovasculares/patologia , Doenças Cardiovasculares/metabolismo
13.
Cardiovasc Res ; 120(7): 681-698, 2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38630620

RESUMO

Mast cells are tissue-resident immune cells strategically located in different compartments of the normal human heart (the myocardium, pericardium, aortic valve, and close to nerves) as well as in atherosclerotic plaques. Cardiac mast cells produce a broad spectrum of vasoactive and proinflammatory mediators, which have potential roles in inflammation, angiogenesis, lymphangiogenesis, tissue remodelling, and fibrosis. Mast cells release preformed mediators (e.g. histamine, tryptase, and chymase) and de novo synthesized mediators (e.g. cysteinyl leukotriene C4 and prostaglandin D2), as well as cytokines and chemokines, which can activate different resident immune cells (e.g. macrophages) and structural cells (e.g. fibroblasts and endothelial cells) in the human heart and aorta. The transcriptional profiles of various mast cell populations highlight their potential heterogeneity and distinct gene and proteome expression. Mast cell plasticity and heterogeneity enable these cells the potential for performing different, even opposite, functions in response to changing tissue contexts. Human cardiac mast cells display significant differences compared with mast cells isolated from other organs. These characteristics make cardiac mast cells intriguing, given their dichotomous potential roles of inducing or protecting against cardiovascular diseases. Identification of cardiac mast cell subpopulations represents a prerequisite for understanding their potential multifaceted roles in health and disease. Several new drugs specifically targeting human mast cell activation are under development or in clinical trials. Mast cells and/or their subpopulations can potentially represent novel therapeutic targets for cardiovascular disorders.


Assuntos
Doenças Cardiovasculares , Mastócitos , Humanos , Mastócitos/metabolismo , Mastócitos/imunologia , Mastócitos/efeitos dos fármacos , Mastócitos/patologia , Animais , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/patologia , Doenças Cardiovasculares/imunologia , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/genética , Transdução de Sinais , Fenótipo , Miocárdio/patologia , Miocárdio/metabolismo , Miocárdio/imunologia , Fármacos Cardiovasculares/uso terapêutico , Fármacos Cardiovasculares/farmacologia , Plasticidade Celular/efeitos dos fármacos , Mediadores da Inflamação/metabolismo
14.
J Clin Invest ; 134(10)2024 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-38564300

RESUMO

Nuclear factor κ-B (NFκB) is activated in iPSC-cardiac myocytes from patients with arrhythmogenic cardiomyopathy (ACM) under basal conditions, and inhibition of NFκB signaling prevents disease in Dsg2mut/mut mice, a robust mouse model of ACM. Here, we used genetic approaches and single-cell RNA-Seq to define the contributions of immune signaling in cardiac myocytes and macrophages in the natural progression of ACM using Dsg2mut/mut mice. We found that NFκB signaling in cardiac myocytes drives myocardial injury, contractile dysfunction, and arrhythmias in Dsg2mut/mut mice. NFκB signaling in cardiac myocytes mobilizes macrophages expressing C-C motif chemokine receptor-2 (CCR2+ cells) to affected areas within the heart, where they mediate myocardial injury and arrhythmias. Contractile dysfunction in Dsg2mut/mut mice is caused both by loss of heart muscle and negative inotropic effects of inflammation in viable muscle. Single nucleus RNA-Seq and cellular indexing of transcriptomes and epitomes (CITE-Seq) studies revealed marked proinflammatory changes in gene expression and the cellular landscape in hearts of Dsg2mut/mut mice involving cardiac myocytes, fibroblasts, and CCR2+ macrophages. Changes in gene expression in cardiac myocytes and fibroblasts in Dsg2mut/mut mice were dependent on CCR2+ macrophage recruitment to the heart. These results highlight complex mechanisms of immune injury and regulatory crosstalk between cardiac myocytes, inflammatory cells, and fibroblasts in the pathogenesis of ACM.


Assuntos
Desmogleína 2 , Modelos Animais de Doenças , Macrófagos , NF-kappa B , Receptores CCR2 , Transdução de Sinais , Animais , Camundongos , Macrófagos/metabolismo , Macrófagos/patologia , Macrófagos/imunologia , Receptores CCR2/genética , Receptores CCR2/metabolismo , Desmogleína 2/genética , Desmogleína 2/metabolismo , NF-kappa B/metabolismo , NF-kappa B/genética , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Miócitos Cardíacos/imunologia , Humanos , Displasia Arritmogênica Ventricular Direita/genética , Displasia Arritmogênica Ventricular Direita/metabolismo , Displasia Arritmogênica Ventricular Direita/patologia , Miocárdio/patologia , Miocárdio/metabolismo , Miocárdio/imunologia
15.
Cardiovasc Pathol ; 72: 107646, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38677634

RESUMO

BACKGROUND: Pathologic antibody mediated rejection (pAMR) remains a major driver of graft failure in cardiac transplant patients. The endomyocardial biopsy remains the primary diagnostic tool but presents with challenges, particularly in distinguishing the histologic component (pAMR-H) defined by 1) intravascular macrophage accumulation in capillaries and 2) activated endothelial cells that expand the cytoplasm to narrow or occlude the vascular lumen. Frequently, pAMR-H is difficult to distinguish from acute cellular rejection (ACR) and healing injury. With the advent of digital slide scanning and advances in machine deep learning, artificial intelligence technology is widely under investigation in the areas of oncologic pathology, but in its infancy in transplant pathology. For the first time, we determined if a machine learning algorithm could distinguish pAMR-H from normal myocardium, healing injury and ACR. MATERIALS AND METHODS: A total of 4,212 annotations (1,053 regions of normal, 1,053 pAMR-H, 1,053 healing injury and 1,053 ACR) were completed from 300 hematoxylin and eosin slides scanned using a Leica Aperio GT450 digital whole slide scanner at 40X magnification. All regions of pAMR-H were annotated from patients confirmed with a previous diagnosis of pAMR2 (>50% positive C4d immunofluorescence and/or >10% CD68 positive intravascular macrophages). Annotations were imported into a Python 3.7 development environment using the OpenSlide™ package and a convolutional neural network approach utilizing transfer learning was performed. RESULTS: The machine learning algorithm showed 98% overall validation accuracy and pAMR-H was correctly distinguished from specific categories with the following accuracies: normal myocardium (99.2%), healing injury (99.5%) and ACR (99.5%). CONCLUSION: Our novel deep learning algorithm can reach acceptable, and possibly surpass, performance of current diagnostic standards of identifying pAMR-H. Such a tool may serve as an adjunct diagnostic aid for improving the pathologist's accuracy and reproducibility, especially in difficult cases with high inter-observer variability. This is one of the first studies that provides evidence that an artificial intelligence machine learning algorithm can be trained and validated to diagnose pAMR-H in cardiac transplant patients. Ongoing studies include multi-institutional verification testing to ensure generalizability.


Assuntos
Rejeição de Enxerto , Transplante de Coração , Miocárdio , Valor Preditivo dos Testes , Humanos , Transplante de Coração/efeitos adversos , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/patologia , Rejeição de Enxerto/diagnóstico , Biópsia , Miocárdio/patologia , Miocárdio/imunologia , Reprodutibilidade dos Testes , Interpretação de Imagem Assistida por Computador/métodos , Resultado do Tratamento , Aprendizado de Máquina , Aprendizado Profundo , Macrófagos/imunologia , Macrófagos/patologia , Estudos Retrospectivos
16.
Circulation ; 150(1): 49-61, 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38506045

RESUMO

BACKGROUND: Viral infections can cause acute respiratory distress syndrome (ARDS), systemic inflammation, and secondary cardiovascular complications. Lung macrophage subsets change during ARDS, but the role of heart macrophages in cardiac injury during viral ARDS remains unknown. Here we investigate how immune signals typical for viral ARDS affect cardiac macrophage subsets, cardiovascular health, and systemic inflammation. METHODS: We assessed cardiac macrophage subsets using immunofluorescence histology of autopsy specimens from 21 patients with COVID-19 with SARS-CoV-2-associated ARDS and 33 patients who died from other causes. In mice, we compared cardiac immune cell dynamics after SARS-CoV-2 infection with ARDS induced by intratracheal instillation of Toll-like receptor ligands and an ACE2 (angiotensin-converting enzyme 2) inhibitor. RESULTS: In humans, SARS-CoV-2 increased total cardiac macrophage counts and led to a higher proportion of CCR2+ (C-C chemokine receptor type 2 positive) macrophages. In mice, SARS-CoV-2 and virus-free lung injury triggered profound remodeling of cardiac resident macrophages, recapitulating the clinical expansion of CCR2+ macrophages. Treating mice exposed to virus-like ARDS with a tumor necrosis factor α-neutralizing antibody reduced cardiac monocytes and inflammatory MHCIIlo CCR2+ macrophages while also preserving cardiac function. Virus-like ARDS elevated mortality in mice with pre-existing heart failure. CONCLUSIONS: Our data suggest that viral ARDS promotes cardiac inflammation by expanding the CCR2+ macrophage subset, and the associated cardiac phenotypes in mice can be elicited by activating the host immune system even without viral presence in the heart.


Assuntos
COVID-19 , Cardiomiopatias , Síndrome do Desconforto Respiratório , SARS-CoV-2 , COVID-19/imunologia , COVID-19/complicações , COVID-19/patologia , Animais , Humanos , Síndrome do Desconforto Respiratório/imunologia , Síndrome do Desconforto Respiratório/etiologia , Síndrome do Desconforto Respiratório/patologia , Síndrome do Desconforto Respiratório/virologia , Camundongos , Masculino , Feminino , Cardiomiopatias/imunologia , Cardiomiopatias/etiologia , Cardiomiopatias/patologia , Cardiomiopatias/virologia , Macrófagos/imunologia , Macrófagos/patologia , Macrófagos/metabolismo , Inflamação/patologia , Pessoa de Meia-Idade , Miocárdio/patologia , Miocárdio/imunologia , Camundongos Endogâmicos C57BL , Idoso
17.
Cardiovasc Pathol ; 70: 107624, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38412903

RESUMO

This study aimed to assess the frequency and association between transthyretin-derived (ATTR) amyloidosis and sarcoidosis in a large autopsy cohort including many cases of sudden cardiac death (SCD). We identified 73 sporadic ATTR amyloidosis cases and 11 sarcoidosis cases, among which we found two cases with concomitant ATTR amyloidosis and sarcoidosis (2.4% of all cases; 2.7% within the sporadic ATTR group). The first case involved a 92-year-old man who experienced SCD. In this patient's heart, we observed ATTR deposition and noncaseating epithelioid granulomas consistent with sarcoidosis. Focally, ATTR deposits and granulomas co-localized, with histiocyte phagocytosis of transthyretin-immunoreactive fragments. However, in most lesions, they were distributed independently. The second case was that of an 86-year-old woman who also experienced SCD. In this patient, we detected ATTR deposition in the heart and lung, while noncaseating epithelioid granulomas were only observed in the lung, liver, kidney, and thyroid. Furthermore, no co-localization of the two lesions was observed. Based on these findings, we concluded that the coexistence of ATTR amyloidosis and sarcoidosis was likely coincidental. Nevertheless, despite the rarity of the combination of these two diseases, it should be recognized as a potential cause of SCD, especially among elderly people.


Assuntos
Neuropatias Amiloides Familiares , Granuloma , Sarcoidose , Humanos , Idoso de 80 Anos ou mais , Feminino , Masculino , Granuloma/patologia , Granuloma/metabolismo , Sarcoidose/patologia , Sarcoidose/metabolismo , Sarcoidose/complicações , Neuropatias Amiloides Familiares/patologia , Neuropatias Amiloides Familiares/metabolismo , Neuropatias Amiloides Familiares/complicações , Idoso , Autopsia , Miocárdio/patologia , Miocárdio/metabolismo , Miocárdio/imunologia , Morte Súbita Cardíaca/etiologia , Morte Súbita Cardíaca/patologia , Pessoa de Meia-Idade , Pré-Albumina/análise , Pré-Albumina/metabolismo , Cardiomiopatias/patologia , Cardiomiopatias/metabolismo , Cardiomiopatias/etiologia , Cardiomiopatias/imunologia
18.
Int. j. cardiovasc. sci. (Impr.) ; 35(3): 410-418, May-June 2022. graf
Artigo em Inglês | LILACS | ID: biblio-1375650

RESUMO

Abstract An acute respiratory syndrome caused by SARS-CoV2 was declared a pandemic by the World Health Organization. Current data in the world and in Brazil show that approximately 40% of patients who died have some type of cardiac comorbidity. There are also robust reports showing an increase in IL-6 / IL-1B / TNF-alpha and the presence of lymphopenia in patients with COVID-19. Our team and others have shown that increased cytokines are the link between arrhythmias/Left ventricular dysfunction and the immune system in different diseases. In addition, it has been well demonstrated that lymphopenia can not only be a good marker, but also a factor that causes heart failure. Thus, the present review focused on the role of the immune system upon the cardiac alterations observed in the SARS-CoV2 infection. Additionally, it was well described that SARS-CoV-2 is able to infect cardiac cells. Therefore, here it will be reviewed in deep.


Assuntos
Arritmias Cardíacas/complicações , SARS-CoV-2/patogenicidade , COVID-19/complicações , Insuficiência Cardíaca/etiologia , Miocárdio/imunologia , Arritmias Cardíacas/fisiopatologia , Citocinas , Citocinas/imunologia , Coronavirus/patogenicidade , Disfunção Ventricular Esquerda/fisiopatologia , Miócitos Cardíacos/patologia , Síndrome Respiratória Aguda Grave , Insuficiência Cardíaca/complicações , Linfopenia/complicações
19.
Rev. bras. parasitol. vet ; 26(1): 100-103, Jan.-Mar. 2017.
Artigo em Inglês | LILACS | ID: biblio-844123

RESUMO

Abstract Sarcocystis spp., Neospora spp., and Toxoplasma gondii are Apicomplexa protozoa that can infect horses. This study aimed to investigate the occurrence of antibodies against Sarcocystis spp., Neospora spp., and T. gondii in horses slaughtered in southern Brazil. The presence of histological lesions, tissue cysts, and Sarcocystis spp. DNA in the hearts of these horses was also investigated. A total of 197 paired serum and heart samples were evaluated by serology and direct microscopic examination; 50 of these samples were subjected to histopathological and PCR analyses. Antibodies against at least one of the protozoa were detected in 146 (74.1%) of the serum samples. The frequencies of positive serology were: 36% (71/197) against Sarcocystis spp., 39.1% (77/197) against Neospora spp., and 47.2% (93/197) against T. gondii. No cysts, Sarcocystis spp. DNA, or histopathological lesions were observed in myocardial tissue samples. The frequencies of antibody seropositivity against Sarcocystis spp., Neospora spp., and T. gondii showed that horses are frequently infected by these parasites in southern Brazil. The absence of sarcocysts in horse tissues is compatible with their role as aberrant/accidental hosts in the life cycle of Sarcocystis spp..


Resumo Sarcocystis spp., Neospora spp. e Toxoplasma gondii são protozoários que pertencem ao filo Apicomplexa e que podem afetar equinos. O objetivo deste estudo foi investigar a ocorrência de anticorpos contra Sarcocystis spp., Neospora spp. e T. gondii. A presença de lesões histológicas, cistos teciduais e DNA de Sarcocystis spp. no miocárdio de equinos abatidos no sul do Brasil também foi investigado. Um total de 197 amostras de soro juntamente com as respectivas amostras de coração, foram avaliadas por sorologia e exame microscópico direto. Destas amostras, 50 foram selecionadas e submetidas a análise histopatológica e PCR. Anticorpos contra pelo menos um dos protozoários foi detectado em 146 (74,1%) das amostras de soro. As frequências de sorologia positiva foram: 36% (71/197) para Sarcocystis spp., 39,1% (77/197) para Neospora spp. e 47,2% (93/197) para T. gondii. Não foram encontradas lesões histopatológicas, cistos e DNA de Sarcocystis spp. nas amostras de miocárdio dos equinos. As frequências de soropositividade para Sarcocystis spp., Neospora spp. e T. gondii mostra que os equinos podem ser frequentemente infectados por estes parasitas no sul do Brasil. A ausência de sarcocistose no coração dos equinos é compatível com seu papel como hospedeiro errático/acidental no ciclo de vida deste protozoário.


Assuntos
Animais , Toxoplasma/imunologia , Anticorpos Antiprotozoários/análise , Sarcocystis/imunologia , Neospora/imunologia , Coração/parasitologia , Doenças dos Cavalos/parasitologia , Brasil , Estudos Soroepidemiológicos , Sarcocistose/veterinária , Coccidiose/veterinária , Doenças dos Cavalos/imunologia , Cavalos , Miocárdio/imunologia
20.
Biomédica (Bogotá) ; 34(4): 514-520, oct.-dic. 2014. ilus, tab
Artigo em Espanhol | LILACS | ID: lil-730934

RESUMO

El panorama epidemiológico del dengue ha empeorado durante la última década. Las dificultades para prevenir su transmisión, así como la ausencia de una vacuna o tratamiento específico, lo convierten en un riesgo que desafía las medidas de salud pública y desborda la capacidad de los centros de salud y los sistemas de investigación a muchos niveles. Actualmente, la mayoría de los estudios sobre la patogenia de la infección centran su atención en la respuesta inmunitaria de las células T casi exclusivamente en infecciones secundarias y están dirigidos a identificar los mecanismos implicados en el desarrollo de la permeabilidad vascular y de los eventos hemorrágicos que lo acompañan. En este reporte se describe el caso de una menor de 45 días de edad con signos clínicos de dengue grave, cuyo diagnóstico se confirmó por reacción en cadena de la polimerasa de transcripción inversa en muestras de tejido post mórtem y por herramientas de apoyo diagnóstico de inmunohistoquímica, las cuales detectaron antígenos virales en todos los órganos obtenidos en la necropsia. Este caso subraya la importancia del estudio de las infecciones primarias asociadas a dengue grave, particularmente en niños, en quienes es más probable el desarrollo de la forma grave de la enfermedad sin una infección previa, y, además, pone de relieve la importancia de un diagnóstico que no se limite a las muestras de tejido hepático en el estudio de la patogenia de la infección viral.


The epidemiological situation of dengue has worsened over the last decade. The difficulties in preventing its transmission and the absence of a vaccine or specific treatment have made dengue a serious risk to public health, health centers and research systems at different levels. Currently, most studies on the pathogenesis of dengue infection focus on the T-cell immune response almost exclusively in secondary infections and are aimed at identifying the mechanisms involved in the development of vascular permeability and bleeding events that accompany the infection. This report describes the case of a baby girl less than 45 days of age with clinical signs of severe dengue, whose diagnosis was confirmed by reverse transcription polymerase chain reaction in post-mortem tissue samples and by the ancillary diagnostic use of immunohistochemistry, which detected viral antigens in all organs obtained at autopsy. This case highlights the importance of studying primary infections associated with severe dengue, particularly in children, who are more likely to develop the severe form of the disease without previous infection, and it further stresses the importance of a diagnosis that should not be based solely on the examination of liver tissue samples when studying the pathogenesis of the viral infection.


Assuntos
Feminino , Humanos , Lactente , Antígenos Virais/análise , Autopsia/métodos , Vírus da Dengue/imunologia , Dengue/patologia , Técnicas Imunoenzimáticas , DNA Viral/análise , Vírus da Dengue/genética , Vírus da Dengue/isolamento & purificação , Dengue/diagnóstico , Dengue/virologia , Coração/virologia , Rim/imunologia , Rim/patologia , Rim/virologia , Fígado/imunologia , Fígado/patologia , Fígado/virologia , Miocárdio/imunologia , Miocárdio/patologia , Especificidade de Órgãos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/imunologia , Baço/patologia , Baço/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA