Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 156
Filtrar
1.
Front Immunol ; 15: 1402468, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38799471

RESUMO

Ischemic heart disease (IHD) is a leading cause of disability and death worldwide, with immune regulation playing a crucial role in its pathogenesis. Various immune cells are involved, and as one of the key immune cells residing in the heart, macrophages play an indispensable role in the inflammatory and reparative processes during cardiac ischemia. Exosomes, extracellular vesicles containing lipids, nucleic acids, proteins, and other bioactive molecules, have emerged as important mediators in the regulatory functions of macrophages and hold promise as a novel therapeutic target for IHD. This review summarizes the regulatory mechanisms of different subsets of macrophages and their secreted exosomes during cardiac ischemia over the past five years. It also discusses the current status of clinical research utilizing macrophages and their exosomes, as well as strategies to enhance their therapeutic efficacy through biotechnology. The aim is to provide valuable insights for the treatment of IHD.


Assuntos
Exossomos , Macrófagos , Isquemia Miocárdica , Exossomos/metabolismo , Exossomos/imunologia , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Isquemia Miocárdica/imunologia , Isquemia Miocárdica/metabolismo , Animais
2.
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
3.
Nat Rev Cardiol ; 18(5): 368-379, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33462421

RESUMO

The lymphatic vasculature has an essential role in maintaining normal fluid balance in tissues and modulating the inflammatory response to injury or pathogens. Disruption of normal development or function of lymphatic vessels can have severe consequences. In the heart, reduced lymphatic function can lead to myocardial oedema and persistent inflammation. Macrophages, which are phagocytic cells of the innate immune system, contribute to cardiac development and to fibrotic repair and regeneration of cardiac tissue after myocardial infarction. In this Review, we discuss the cardiac lymphatic vasculature with a focus on developments over the past 5 years arising from the study of mammalian and zebrafish model organisms. In addition, we examine the interplay between the cardiac lymphatics and macrophages during fibrotic repair and regeneration after myocardial infarction. Finally, we discuss the therapeutic potential of targeting the cardiac lymphatic network to regulate immune cell content and alleviate inflammation in patients with ischaemic heart disease.


Assuntos
Coração , Inflamação , Vasos Linfáticos , Macrófagos , Isquemia Miocárdica , Regeneração , Animais , Modelos Animais de Doenças , Fibrose/imunologia , Fibrose/fisiopatologia , Coração/embriologia , Coração/fisiologia , Coração/fisiopatologia , Humanos , Inflamação/imunologia , Inflamação/fisiopatologia , Vasos Linfáticos/embriologia , Vasos Linfáticos/imunologia , Vasos Linfáticos/fisiologia , Vasos Linfáticos/fisiopatologia , Macrófagos/imunologia , Macrófagos/fisiologia , Isquemia Miocárdica/imunologia , Isquemia Miocárdica/fisiopatologia , Miocárdio/imunologia , Regeneração/imunologia , Regeneração/fisiologia
4.
Clin Transl Sci ; 14(1): 317-325, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32949228

RESUMO

Adipose-derived mesenchymal stem cells (ADSCs) are promising candidates for novel cell therapeutic applications. Hibernating brown bears sustain tissue integrity and function via unknown mechanisms, which might be plasma borne. We hypothesized that plasma from hibernating bears may increase the expression of favorable factors from human ADSCs. In an experimental study, ADSCs from patients with ischemic heart disease were treated with interventional media containing plasma from hibernating and active bears, respectively, and with control medium. Extracted RNA from the ADSCs was sequenced using next generation sequencing. Statistical analyses of differentially expressed genes were performed using fold change analysis, pathway analysis, and gene ontology. As a result, we found that genes associated with inflammation, such as IGF1, PGF, IL11, and TGFA, were downregulated by > 10-fold in ADSCs treated with winter plasma compared with control. Genes important for cardiovascular development, ADM, ANGPTL4, and APOL3, were upregulated in ADSCs when treated with winter plasma compared with summer plasma. ADSCs treated with bear plasma, regardless if it was from hibernating or active bears, showed downregulation of IGF1, PGF, IL11, INHBA, IER3, and HMOX1 compared with control, suggesting reduced cell growth and differentiation. This can be summarized in the conclusion that plasma from hibernating bears suppresses inflammatory genes and activates genes associated with cardiovascular development in human ADSCs. Identifying the involved regulator(s) holds therapeutic potential.


Assuntos
Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/imunologia , Isquemia Miocárdica/terapia , Plasma/imunologia , Ursidae/sangue , Idoso , Idoso de 80 Anos ou mais , Animais , Diferenciação Celular/imunologia , Células Cultivadas , Ponte de Artéria Coronária , Meios de Cultura/metabolismo , Feminino , Hibernação/imunologia , Humanos , Masculino , Isquemia Miocárdica/imunologia , Plasma/metabolismo , Cultura Primária de Células/métodos , Estações do Ano , Gordura Subcutânea/citologia , Transplante Autólogo/métodos , Ursidae/imunologia
5.
J Immunol Res ; 2020: 8632048, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33299899

RESUMO

Novel coronavirus disease 2019 (COVID-19) causes pulmonary and cardiovascular disorders and has become a worldwide emergency. Myocardial injury can be caused by direct or indirect damage, particularly mediated by a cytokine storm, a disordered immune response that can cause myocarditis, abnormal coagulation, arrhythmia, acute coronary syndrome, and myocardial infarction. The present review focuses on the mechanisms of this viral infection, cardiac biomarkers, consequences, and the possible therapeutic role of purinergic and adenosinergic signalling systems. In particular, we focus on the interaction of the extracellular nucleotide adenosine triphosphate (ATP) with its receptors P2X1, P2X4, P2X7, P2Y1, and P2Y2 and of adenosine (Ado) with A2A and A3 receptors, as well as their roles in host immune responses. We suggest that receptors of purinergic signalling could be ideal candidates for pharmacological targeting to protect against myocardial injury caused by a cytokine storm in COVID-19, in order to reduce systemic inflammatory damage to cells and tissues, preventing the progression of the disease by modulating the immune response and improving patient quality of life.


Assuntos
Trifosfato de Adenosina/metabolismo , COVID-19/imunologia , Doenças Cardiovasculares/virologia , Receptores Purinérgicos/metabolismo , SARS-CoV-2 , Agonistas do Receptor A2 de Adenosina/farmacologia , COVID-19/metabolismo , Doenças Cardiovasculares/imunologia , Doenças Cardiovasculares/fisiopatologia , Citocinas/metabolismo , Humanos , Isquemia Miocárdica/imunologia , Isquemia Miocárdica/fisiopatologia , Isquemia Miocárdica/virologia , Pandemias , Antagonistas Purinérgicos/farmacologia , Receptor A2A de Adenosina/metabolismo , Receptor A3 de Adenosina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Tratamento Farmacológico da COVID-19
6.
Zhongguo Zhen Jiu ; 40(6): 635-9, 2020 Jun 12.
Artigo em Chinês | MEDLINE | ID: mdl-32538016

RESUMO

OBJECTIVE: To observe the effects of electroacupuncture (EA) pretreatment on the cardiac ejection fraction (EF), the number of macrophages in spleen and heart, and the expression of nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) and interleukin-1ß (IL-1ß) in myocardium in mice with acute myocardial ischemia, and to explore the possible mechanism of EA pretreatment on promoting myocardial protection. METHODS: A total of 30 male C57BL/6J mice were randomly divided into a control group, a model group and an EA pretreatment group, 10 rats in each group. The acute myocardial ischemia model was established by ligating the left anterior descending branch of the coronary artery in the model group and EA pretreatment group, while threading but no ligating at left anterior descending branch of the coronary artery was applied in the control group. In the EA pretreatment group, mice were intervented with EA at bilateral "Neiguan" (PC 6), disperse-dense wave, frequency of 2 Hz/15 Hz, intensity of 2 mA; each EA treatment last for 20 min, once a day, and 3-day treatment was given before model establishment. The EF value was evaluated by ultrasonic cardiogram; the number of macrophages in spleen and heart was measured by flow cytometry; the expression level of NLRP3 and IL-1ß in myocardium was measured by Western blot. RESULTS: Compared with the control group, the EF value was decreased in the model group (P<0.001), the number of macrophages in the heart and spleen was increased (P<0.001), and the expression level of NLRP3 and IL-1ß in the myocardium was increased (P<0.001, P<0.01). Compared with the model group, the EF value was increased in the EA pretreatment group (P<0.01), the number of macrophages in the heart and spleen was decreased (P<0.01), and the expression level of NLRP3 and IL-1ß in the myocardium was decreased (P<0.01, P<0.05). CONCLUSION: EA pretreatment could reduce the number of macrophages in spleen and heart, down-regulate the expression of NLRP3 and IL-1ß in myocardial tissue in mice with acute myocardial ischemia, which could relieve the local inflammatory response and achieve the myocardial protective effect.


Assuntos
Eletroacupuntura , Coração/fisiologia , Inflamação/imunologia , Isquemia Miocárdica/terapia , Pontos de Acupuntura , Animais , Interleucina-1beta/metabolismo , Macrófagos/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Isquemia Miocárdica/imunologia , Miocárdio , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Distribuição Aleatória , Baço
7.
J Int Med Res ; 48(5): 300060520904835, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32389049

RESUMO

OBJECTIVE: This study aimed to determine whether proinflammatory cytokines have an effect on myocardial cells (MCs) and hepatocytes during myocardial ischemia to induce cyclic AMP-responsive element-binding protein H (CREBH) cleavage, activate the acute phase response in the liver, and cause a superimposed injury in MCs. METHODS: In this study, a hepatocyte-MC transwell co-culture system was used to investigate the relationship between myocardial hypoxia/reperfusion injury and CREBH cleavage. MCs and hepatocytes of neonatal rats were obtained from the ventricles and livers of Sprague-Dawley rats, respectively. MCs were inoculated into the lower chamber of transwell chambers for 12 hours under hypoxia. Levels of the endoplasmic reticulum stress protein glucose-regulated protein 78 in MCs, CREBH in hepatocytes, inflammatory factor (tumor necrosis factor-α and interleukin-6) levels, and cell viability were evaluated. The effect of CREBH knockdown was also studied using a CREBH-specific short hairpin RNA (Ad-CREBHi). RESULTS: We found that proinflammatory cytokines affect MCs and hepatocytes during myocardial ischemia to induce CREBH cleavage, activate the acute phase response in the liver, and cause superimposed injury in MCs. CONCLUSIONS: Expression of CREBH aggravates myocardial injury during myocardial ischemia.


Assuntos
Reação de Fase Aguda/imunologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Hepatócitos/metabolismo , Isquemia Miocárdica/imunologia , Miócitos Cardíacos/metabolismo , Animais , Animais Recém-Nascidos , Hipóxia Celular/imunologia , Sobrevivência Celular/imunologia , Células Cultivadas , Técnicas de Cocultura , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Modelos Animais de Doenças , Estresse do Retículo Endoplasmático/imunologia , Técnicas de Silenciamento de Genes , Proteínas de Choque Térmico/metabolismo , Hepatócitos/imunologia , Humanos , Interleucina-6/metabolismo , Fígado/citologia , Fígado/imunologia , Fígado/patologia , Isquemia Miocárdica/patologia , Miocárdio/citologia , Miocárdio/imunologia , Miocárdio/patologia , Miócitos Cardíacos/imunologia , Cultura Primária de Células , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/imunologia , Fator de Necrose Tumoral alfa/metabolismo
8.
Life Sci ; 248: 117451, 2020 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-32088213

RESUMO

AIMS: Electro-acupuncture pretreatment (EAP) plays a protective role in myocardial ischemia (MI) injury. However, the underlying mechanism remains unclear. A growing body of evidence suggests postinfarction inflammatory response directly affects the remodeling of ventricular function. The purpose of this study was to investigate whether EAP alleviates MI through NLRP3 inflammasome inhibition. MATERIALS AND METHODS: We constructed an AMI model by ligating the left anterior descending (LAD) coronary artery after 3 days of EAP with C57BL/6 mice. Echocardiography and TTC staining were employed to evaluate cardiac function and infarct size after 24 h of ischemia. HE staining and immunohistochemistry were employed to determine inflammatory level. Then, inflammasome activation was detected by western blotting, and macrophage polarization and neutrophil infiltration were observed by flow cytometry. KEY FINDINGS: Our preliminary findings showed that EAP reduced the infarct area and increased fractional shortening (FS) and ejection fraction (EF) and decreased the degree of inflammation after AMI injury. Meanwhile, EAP inhibited the expression of NLRP3, cleaved caspase-1 and IL-1ß in ischemia myocardial tissue, companied by inhibiting the expression of F4/80+, CD11b+, CD206low macrophages and activated M2 macrophage, and decreasing Ly-6G+CD11b+ neutrophils in ischemia myocardial and spleen tissue. SIGNIFICANCE: EAP inhibits the activation of NLRP3 inflammasome, promotes M2 polarization of macrophages and reduces the recruitment of neutrophils in damaged myocardium, thereby decreases the infarct size and improves the cardiac function.


Assuntos
Eletroacupuntura/métodos , Inflamassomos/imunologia , Precondicionamento Isquêmico Miocárdico , Isquemia Miocárdica/genética , Isquemia Miocárdica/terapia , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Animais , Antígenos Ly/genética , Antígenos Ly/imunologia , Antígeno CD11b/genética , Antígeno CD11b/imunologia , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/imunologia , Caspase 1/genética , Caspase 1/imunologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Inflamassomos/genética , Inflamação , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Lectinas Tipo C/genética , Lectinas Tipo C/imunologia , Macrófagos/imunologia , Macrófagos/patologia , Masculino , Receptor de Manose , Lectinas de Ligação a Manose/genética , Lectinas de Ligação a Manose/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Isquemia Miocárdica/imunologia , Isquemia Miocárdica/patologia , Miocárdio/imunologia , Miocárdio/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Infiltração de Neutrófilos , Neutrófilos/imunologia , Neutrófilos/patologia , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/imunologia , Transdução de Sinais
9.
JCI Insight ; 52019 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-31287805

RESUMO

Podoplanin, a small mucine-type transmembrane glycoprotein, has been recently shown to be expressed by lymphangiogenic, fibrogenic and mesenchymal progenitor cells in the acutely and chronically infarcted myocardium. Podoplanin binds to CLEC-2, a C-type lectin-like receptor 2 highly expressed by CD11bhigh cells following inflammatory stimuli. Why podoplanin expression appears only after organ injury is currently unknown. Here, we characterize the role of podoplanin in different stages of myocardial repair after infarction and propose a podoplanin-mediated mechanism in the resolution of post-MI inflammatory response and cardiac repair. Neutralization of podoplanin led to significant improvements in the left ventricular functions and scar composition in animals treated with podoplanin neutralizing antibody. The inhibition of the interaction between podoplanin and CLEC-2 expressing immune cells in the heart enhances the cardiac performance, regeneration and angiogenesis post MI. Our data indicates that modulating the interaction between podoplanin positive cells with the immune cells after myocardial infarction positively affects immune cell recruitment and may represent a novel therapeutic target to augment post-MI cardiac repair, regeneration and function.


Assuntos
Cicatriz/metabolismo , Insuficiência Cardíaca/metabolismo , Hipertrofia Ventricular Esquerda/metabolismo , Glicoproteínas de Membrana/metabolismo , Infarto do Miocárdio/metabolismo , Remodelação Ventricular/genética , Angiotensina II/toxicidade , Animais , Anticorpos Neutralizantes , Cardiomiopatias/imunologia , Cardiomiopatias/metabolismo , Cardiomiopatias/cirurgia , Sobrevivência Celular/imunologia , Cicatriz/imunologia , Ecocardiografia , Fibrose , Insuficiência Cardíaca/induzido quimicamente , Insuficiência Cardíaca/imunologia , Transplante de Coração , Hemodinâmica , Humanos , Hipertrofia Ventricular Esquerda/induzido quimicamente , Hipertrofia Ventricular Esquerda/imunologia , Inflamação/imunologia , Macrófagos/imunologia , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/imunologia , Camundongos , Monócitos/imunologia , Infarto do Miocárdio/imunologia , Isquemia Miocárdica/imunologia , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/cirurgia , Miócitos Cardíacos , Regeneração/imunologia , Vasoconstritores/toxicidade , Função Ventricular Esquerda , Remodelação Ventricular/imunologia
10.
Front Immunol ; 10: 1130, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31191522

RESUMO

Atherosclerosis is a chronic autoimmune inflammatory disease that can cause coronary artery disease, stroke, peripheral artery disease, depending on which arteries are affected. At the beginning of atherosclerosis plasma lipoproteins accumulate in the sub-endothelial space. In response, monocytes migrate from the circulation through the endothelium into the intima where they differentiate into macrophages. These early events trigger a complex immune response that eventually involves many cellular subtypes of both innate and adaptive immunity. The Notch signaling pathway is an evolutionary conserved cell signaling system that mediates cell-to-cell communication. Recent studies have revealed that Notch modulate atherosclerosis by controlling macrophages polarization into M1 or M2 subtypes. Furthermore, it is known that Notch signaling controls differentiation and activity of T-helper and cytotoxic T-cells in inflammatory diseases. In this review, we will discuss the role of Notch in modulating immunity in the context of atherosclerosis and whether targeting Notch may represent a therapeutic strategy.


Assuntos
Aterosclerose/imunologia , Receptores Notch/imunologia , Animais , Progressão da Doença , Humanos , Macrófagos/imunologia , Isquemia Miocárdica/imunologia , Transdução de Sinais , Linfócitos T/imunologia
11.
Circ J ; 83(7): 1422-1430, 2019 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31178469

RESUMO

In 1997, the seminal manuscript by Asahara, Murohara, Isner et al outlined the evidence for the existence of circulating, bone marrow-derived cells capable of stimulating and contributing to the formation of new blood vessels. Consistent with the paradigm shift that this work represented, it triggered much scientific debate and controversy, some of which persists 2 decades later. In contrast, the clinical application of autologous CD34 cell therapy has been marked by a track record of consistent safety and clinical benefit in multiple ischemic conditions. In this review, we summarize the preclinical and clinical evidence from over 700 patients in clinical trials of CD34 cell therapy.


Assuntos
Antígenos CD34/imunologia , Transtornos Cerebrovasculares/cirurgia , Células Progenitoras Endoteliais/transplante , Extremidade Inferior/irrigação sanguínea , Isquemia Miocárdica/cirurgia , Neovascularização Fisiológica , Doença Arterial Periférica/cirurgia , Regeneração , Transplante de Células-Tronco , Animais , Transtornos Cerebrovasculares/diagnóstico , Transtornos Cerebrovasculares/imunologia , Transtornos Cerebrovasculares/fisiopatologia , Células Progenitoras Endoteliais/imunologia , Humanos , Isquemia Miocárdica/diagnóstico , Isquemia Miocárdica/imunologia , Isquemia Miocárdica/fisiopatologia , Doença Arterial Periférica/diagnóstico , Doença Arterial Periférica/imunologia , Doença Arterial Periférica/fisiopatologia , Recuperação de Função Fisiológica , Transplante de Células-Tronco/efeitos adversos , Resultado do Tratamento
12.
Int J Cardiol ; 293: 238-247, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31230935

RESUMO

BACKGROUND: A sustained low grade inflammatory state is a recognized feature of various diseases, including cardiovascular disease. This state of chronic inflammation involves activation of Toll-like receptor (TLR) signaling. However, little is known regarding the genetic profile of TLR components in cardiac tissue from patients with cardiac disease. METHODS: In this study we investigated the genetic profile of 84 TLR markers in a unique set of cardiac tissue from patients that had undergone either coronary artery bypass grafting (CABG) or aortic valve replacement (AVR). In addition, we compared the gene data from the cardiac tissue with the same gene profile in blood as well as circulating cytokines to elucidate possible targets in blood that could be used to estimate the inflammatory state of the heart in cardiac disease. RESULTS: We found a marked upregulation of TLR-induced inflammation in cardiac tissue from both patient groups compared to healthy controls. The inflammation appeared to be primarily mediated through TLR1, 3, 7, 8 and 10, resulting in a marked induction of mediators of the innate immune response. Furthermore, the gene expression data in combination with unbiased multivariate analysis suggested a difference in inflammatory response in ischemic cardiac tissue compared to non-ischemic cardiac tissue. Serum levels of IL-13 were significantly elevated in both CABG and AVR patients compared to controls, whereas other cytokines did not appear to coincide with cardiac TLR-induced inflammation. CONCLUSIONS: We propose that cardiac disease in humans may be mediated by local cardiac TLR signaling under both ischemic and non-ischemic conditions.


Assuntos
Cardiopatias , Inflamação/imunologia , Isquemia Miocárdica/imunologia , Receptores Toll-Like , Valva Aórtica/cirurgia , Ponte de Artéria Coronária/métodos , Feminino , Perfilação da Expressão Gênica/métodos , Cardiopatias/etiologia , Cardiopatias/imunologia , Cardiopatias/cirurgia , Humanos , Imunidade Inata/genética , Mediadores da Inflamação/imunologia , Masculino , Pessoa de Meia-Idade , Transdução de Sinais/imunologia , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia , Regulação para Cima
13.
Transplantation ; 103(8): 1582-1590, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31107828

RESUMO

BACKGROUND: Although allogeneic-induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CMs) exhibit potential in cardiomyogenesis for heart failure, whether major histocompatibility complex (MHC)-matched allogenic iPSC implantation (MMAI) minimizes immune rejection for cell survival or functional recovery remains unknown. We herein explored whether MMAI with an iPSC-CM sheet is stable for a longer period and therapeutically more effective than MHC-mismatched AI in a primate ischemic cardiomyopathy model. METHODS: Green fluorescent protein-transfected iPSC-CM sheets, derived from cynomolgus macaques with homozygous MHC haplotypes ''HT1,'' were transplanted on the left ventricle, generated by ligating the left anterior descending artery for 2 weeks in an ischemic model with or without heterozygous HT1 as MMAI and MHC-mismatched AI. Sham models were made by opening the chest at 14 days after left anterior descending ligation without any treatment. RESULTS: Stereomicroscopy revealed that at 4 months after transplantation, green fluorescent protein intensity was higher in the MMAI group than in the MHC-mismatched AI group and the sham group. Immunohistochemistry staining revealed that host immune reaction with CD3-positive cells was stronger in MHC-mismatched AI than in MMAI at 3 months. Cardiac function improved both in MMAI and MHC-mismatched AI at 1 month after transplantation and was preserved until 6 months, whereas in the sham group, functional deterioration progressed over time. CONCLUSIONS: Although MHC-homo-iPSCs are preferred to avoid immune rejection, MHC-mismatched iPSC-CMs can also induce comparable cardiac functional recovery at late follow-up, suggesting that MHC-mismatched iPSC-based cardiac regenerative therapy with immunosuppressants is a feasible option for treating heart failure in clinical settings.


Assuntos
Cardiomiopatias/terapia , Ventrículos do Coração/fisiopatologia , Células-Tronco Pluripotentes Induzidas/citologia , Isquemia Miocárdica/terapia , Miócitos Cardíacos/patologia , Transplante de Células-Tronco/métodos , Função Ventricular Esquerda/fisiologia , Animais , Cardiomiopatias/imunologia , Cardiomiopatias/fisiopatologia , Diferenciação Celular , Modelos Animais de Doenças , Ventrículos do Coração/diagnóstico por imagem , Macaca fascicularis , Microscopia de Fluorescência , Tomografia Computadorizada Multidetectores , Isquemia Miocárdica/imunologia , Isquemia Miocárdica/fisiopatologia , Miócitos Cardíacos/imunologia , Recuperação de Função Fisiológica , Transplante Homólogo
14.
Front Immunol ; 10: 634, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30984196

RESUMO

Innate lymphoid cells (ILC) are a subset of leukocytes with lymphoid properties that lack antigen specific receptors. They can be stimulated by and exert their effect via specific cytokine axes, whereas Natural Killers (NK) cells are the only known cytotoxic member of this family. ILCs are considered key in linking the innate and adaptive response in physiologic and pathologic environments. In this study, we investigated the properties of non-cytotoxic cardiac ILCs in physiologic, inflammatory, and ischemic conditions. We found that in healthy humans and mice, non-cytotoxic cardiac ILCs are predominantly a type 2-committed population with progenitor-like features, such as an absence of type-specific immunophenotype, intermediate GATA3 expression, and capacity to transiently express Pro-myelocytic Leukemia Zinc Finger protein (PLZF) upon activation. During myocarditis and ischemia, in both human and mice, cardiac ILCs differentiated into conventional ILC2s. We found that cardiac ILCs lack IL-25 receptor and cannot become inflammatory ILC2s. We found a strong correlation between IL-33 production in the heart and the ability of cardiac ILCs to become conventional ILC2s. The main producer of IL-33 was a subset of CD29+Sca-1+ cardiac fibroblasts. ILC2 expansion and fibroblast-derived IL-33 production were significantly increased in the heart in mouse models of infarction and myocarditis. Despite its progenitor-like status in healthy hearts, cardiac ILCs were unable to become ILC1 or ILC3 in vivo and in vitro. Using adoptive transfer and parabiosis, we demonstrated that the heart, unlike other organs such as lung, cannot be infiltrated by circulating ILCs in adulthood even during cardiac inflammation or ischemia. Thus, the ILC2s present during inflammatory conditions are derived from the heart-resident and quiescent steady-state population. Non-cytotoxic cardiac ILCs are a resident population of ILC2-commited cells, with undifferentiated progenitor-like features in steady-state conditions and an ability to expand and develop pro-inflammatory type 2 features during inflammation or ischemia.


Assuntos
Imunidade Inata , Células Matadoras Naturais/imunologia , Isquemia Miocárdica/imunologia , Miocardite/imunologia , Miocárdio/imunologia , Animais , Feminino , Fibroblastos/imunologia , Fibroblastos/patologia , Fator de Transcrição GATA3/imunologia , Regulação da Expressão Gênica/imunologia , Humanos , Interleucina-33/imunologia , Células Matadoras Naturais/patologia , Masculino , Camundongos , Camundongos Knockout , Isquemia Miocárdica/patologia , Miocardite/patologia , Miocárdio/patologia , Proteína com Dedos de Zinco da Leucemia Promielocítica/imunologia
15.
J Transl Med ; 17(1): 95, 2019 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-30894181

RESUMO

BACKGROUND: Oxidized low-density lipoproteins and scavenger receptors (SRs) play an important role in the formation and development of atherosclerotic plaques. However, little is known about their presence in epicardial adipose tissue (EAT). The objective of the study was to evaluate the mRNA expression of different SRs in EAT of patients with ischemic heart disease (IHD), stratifying by diabetes status and its association with clinical and biochemical variables. METHODS: We analyzed the mRNA expression of SRs (LOX-1, MSR1, CXCL16, CD36 and CL-P1) and macrophage markers (CD68, CD11c and CD206) in EAT from 45 patients with IHD (23 with type 2 diabetes mellitus (T2DM) and 22 without T2DM) and 23 controls without IHD or T2DM. RESULTS: LOX-1, CL-P1, CD68 and CD11c mRNA expression were significantly higher in diabetic patients with IHD when compared with those without T2DM and control patients. MSR1, CXCL16, CD36 and CD206 showed no significant differences. In IHD patients, LOX-1 (OR 2.9; 95% CI 1.6-6.7; P = 0.019) and CD68 mRNA expression (OR 1.7; 95% CI 0.98-4.5; P = 0.049) were identified as independent risk factors associated with T2DM. Glucose and glycated hemoglobin were also shown to be risk factors. CONCLUSIONS: SRs mRNA expression is found in EAT. LOX-1 and CD68 and were higher in IHD patients with T2DM and were identified as a cardiovascular risk factor of T2DM. This study suggests the importance of EAT in coronary atherosclerosis among patients with T2DM.


Assuntos
Tecido Adiposo , Diabetes Mellitus Tipo 2 , Macrófagos/fisiologia , Isquemia Miocárdica , Pericárdio/imunologia , Pericárdio/metabolismo , Receptores Depuradores/genética , Tecido Adiposo/imunologia , Tecido Adiposo/metabolismo , Idoso , Estudos de Casos e Controles , Movimento Celular , Doença da Artéria Coronariana/genética , Doença da Artéria Coronariana/imunologia , Doença da Artéria Coronariana/metabolismo , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/imunologia , Diabetes Mellitus Tipo 2/metabolismo , Cardiomiopatias Diabéticas/complicações , Cardiomiopatias Diabéticas/genética , Cardiomiopatias Diabéticas/imunologia , Cardiomiopatias Diabéticas/metabolismo , Feminino , Humanos , Lipoproteínas LDL/metabolismo , Masculino , Pessoa de Meia-Idade , Isquemia Miocárdica/complicações , Isquemia Miocárdica/genética , Isquemia Miocárdica/imunologia , Isquemia Miocárdica/metabolismo , Receptores Depuradores/metabolismo , Regulação para Cima/genética
16.
Trans Am Clin Climatol Assoc ; 129: 202-214, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30166715

RESUMO

The role of innate and adaptive immunity in heart failure (HF) is poorly understood. We discovered that mice with ischemic HF exhibit robust expansion of proinflammatory monocytes/macrophages, classical and plasmacytoid dendritic cells, and CD4+ and CD8+ T cells. There was profound splenic remodeling indicative of heightened antigen processing, and expanded antigen-experienced effector and memory CD4+ T cell populations. A series of subsequent studies in HF mice that incorporated splenectomy, adoptive transfer of both unselected splenocytes and splenic CD4+ T-cells, and antibody-mediated CD4+ T-cell depletion indicated that intensely activated splenic immune cells: 1) underlie the chronic inflammatory response in HF, 2) traffic and home to the failing heart, and 3) exhibit immune memory and are primed to induce tissue injury that promotes pathological cardiac remodeling. Hence, we propose that ischemic cardiomyopathy is in part an immune-mediated disease, against as-of-yet unidentified cardiac antigens, with a central role for the spleen in this process.


Assuntos
Imunidade Adaptativa , Cardiomiopatias/imunologia , Insuficiência Cardíaca/imunologia , Imunidade Inata , Isquemia Miocárdica/imunologia , Miocárdio/imunologia , Baço/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Cardiomiopatias/fisiopatologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Humanos , Memória Imunológica , Macrófagos/imunologia , Macrófagos/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Isquemia Miocárdica/fisiopatologia , Miocárdio/metabolismo , Miocárdio/patologia , Transdução de Sinais , Baço/metabolismo , Remodelação Ventricular
17.
Front Immunol ; 9: 832, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29780380

RESUMO

Over the recent years, much has been unraveled about the pro-inflammatory properties of various mitochondrial molecules once they are leaving the mitochondrial compartment. On entering the cytoplasm or the extracellular space, mitochondrial DAMPs (also known as mitochondrial alarmins) can become pro-inflammatory and initiate innate and adaptive immune responses by activating cell surface and intracellular receptors. Current evidence indicates that uncontrolled and excessive release of mitochondrial DAMPs is associated with severity, has prognosis value in human diseases, and contributes to the dysregulated process observed in numerous inflammatory and autoimmune conditions, as well as in ischemic heart disease and cancer. Herein, we review that the expanding research field of mitochondrial DAMPs in innate immune responses and the current knowledge on the association between mitochondrial DAMPs and human diseases.


Assuntos
Alarminas/imunologia , Imunidade Inata , Mitocôndrias/patologia , Transdução de Sinais/imunologia , Animais , Doenças Autoimunes/imunologia , Citocinas/imunologia , Humanos , Inflamação , Camundongos , Isquemia Miocárdica/imunologia , Neoplasias/imunologia
18.
Cell Immunol ; 330: 105-113, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29650244

RESUMO

Heart disease is the major cause of death and it is broadly recognized that the immune system plays a central role in healthy and injured heart. Here, we focus on the contribution of various subsets of mononuclear phagocytes in the cardiac system. Macrophages and dendritic cells reside in the healthy myocardium to fulfill homeostatic functions and rapidly increase in numbers in diseases like myocardial ischemia and myocarditis to contribute to disease or resolve it. Recent experiments have revealed the extraordinary heterogeneity of cardiac mononuclear phagocytes that differ in origin, lifespan, phenotype and function. Although many studies described cardiac phagocytes in the mouse, subsets of cardiac mononuclear phagocytes can also be broadly found in the human heart, opening up the potential of selective targeting of these cells in a therapeutic setting. Before this goal can be achieved we need better understanding not only of the detrimental but also beneficial functions of these highly diverse cells in the heart.


Assuntos
Células Dendríticas/imunologia , Macrófagos/imunologia , Isquemia Miocárdica/imunologia , Miocardite/imunologia , Miocárdio/imunologia , Animais , Humanos , Modelos Cardiovasculares , Modelos Imunológicos , Fagócitos/imunologia
19.
J Immunol ; 199(6): 2106-2117, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28768728

RESUMO

Tissue ischemia, such as transient myocardial ischemia, leads to release of cellular RNA including microRNA(miRNA) into the circulation and extracellular (ex-) space, but the biological function of the ex-RNA is poorly understood. We recently reported that cardiac RNA of both human and rodent origins induced cytokine production and immune cell activation. However, the identity of the ex-RNA responsible for the proinflammatory effect remains unclear. In the current study, using an miRNA array, we profiled the plasma miRNAs 4 h after transient myocardial ischemia (45 min) or sham procedure. Among 38 plasma miRNAs that were elevated following ischemia, eight were tested for their ability to induce cytokine response in macrophages and cardiomyocytes. We found that six miRNA mimics (miR-34a, -122, -133a, -142, -146a, and -208a) induced cytokine production in a dose-dependent manner. The effects of miRNAs (miR-133a, -146a, and -208a) were diminished by uridine→adenosine mutation and by RNase pretreatment. The miRNA-induced cytokine (MIP-2, TNF-α, and IL-6) production was abolished in cells deficient of TLR7 or MyD88, or by a TLR7 antagonist, but remained the same in TLR3- or Trif-deficient cells. In vivo, mice i.p. injected with miR-133a or miR-146a had marked peritoneal neutrophil and monocyte migration, which was significantly attenuated in TLR7-/- mice. Moreover, locked nucleic acid anti-miRNA inhibitors of these six miRNAs markedly reduced cardiac RNA-induced cytokine production. Taken together, these data demonstrate that ex-miRNA mimics (miR-34a, -122, -133a, -142, -146a, and -208a) are potent innate immune activators and that the miRNAs most likely induce cytokine production and leukocyte migration through TLR7 signaling.


Assuntos
Espaço Extracelular/metabolismo , Leucócitos/imunologia , Glicoproteínas de Membrana/metabolismo , MicroRNAs/metabolismo , Isquemia Miocárdica/imunologia , Receptor 7 Toll-Like/metabolismo , Animais , Células Cultivadas , Citocinas/metabolismo , Perfilação da Expressão Gênica , Humanos , Imunidade Inata , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/genética , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Receptor 7 Toll-Like/genética
20.
Wiad Lek ; 70(2): 221-223, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28511164

RESUMO

INTRODUCTION: The metabolic syndrome has become pandemic nature and tends to rejuvenation in the world. Elucidation of the pathogenic mechanisms on membrane-cell level will optimize the treatment of patients with metabolic syndrome and prevention of metabolic syndrome on the level of pre-clinical manifestations. AIM: to study the immunological status and lipid metabolism in the patients with metabolic syndrome and the pathogenetic mechanisms of metabolic syndrome were established. MATERIAL AND METHODS: There were 4 groups of pacients (110) with: metabolic syndrome, ischemic heart disease and hypertension, hypertention; control group. General clinical, instrumental, laboratory and statistical methods were used. RESULTS: The levels of immune factors - interleukin-6 in supernatants of mononuclear cells by 65%, sICAM-1 by 20% is elevated in patients with metabolic syndrome compared with the control group. The increasing of the content of saturated fatty acids by 9.4% and polyunsaturated fatty acid by 36.6% lead to fundamental breach of structural and functional properties of membranes. There is significant common carotid artery intima media thickness on average twice at the patients with metabolic syndrome and with ischemic heart disease and hypertension. CONCLUSIONS: The immunoinflammatory reactions were more revealed in the group of patients with metabolic syndrome than in other groups. The lipid state at patients with metabolic syndrome was changed more than in patients with hypertension or patients with ischemic heart disease and hypertension both. Moreover our data indicate the presence of structural changes in the vessel wall in patients with metabolic syndrome.


Assuntos
Ácidos Graxos/sangue , Hipertensão/sangue , Interleucina-6/sangue , Leucócitos Mononucleares/metabolismo , Síndrome Metabólica/sangue , Isquemia Miocárdica/sangue , Adulto , Idoso , Espessura Intima-Media Carotídea , Feminino , Humanos , Hipertensão/imunologia , Masculino , Síndrome Metabólica/imunologia , Pessoa de Meia-Idade , Isquemia Miocárdica/imunologia , Ucrânia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA