Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 882
Filter
Add more filters

Publication year range
1.
Circ Res ; 134(12): 1752-1766, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38843295

ABSTRACT

Heart failure (HF) is characterized by a progressive decline in cardiac function and represents one of the largest health burdens worldwide. Clinically, 2 major types of HF are distinguished based on the left ventricular ejection fraction (EF): HF with reduced EF and HF with preserved EF. While both types share several risk factors and features of adverse cardiac remodeling, unique hallmarks beyond ejection fraction that distinguish these etiologies also exist. These differences may explain the fact that approved therapies for HF with reduced EF are largely ineffective in patients suffering from HF with preserved EF. Improving our understanding of the distinct cellular and molecular mechanisms is crucial for the development of better treatment strategies. This article reviews the knowledge of the immunologic mechanisms underlying HF with reduced and preserved EF and discusses how the different immune profiles elicited may identify attractive therapeutic targets for these conditions. We review the literature on the reported mechanisms of adverse cardiac remodeling in HF with reduced and preserved EF, as well as the immune mechanisms involved. We discuss how the knowledge gained from preclinical models of the complex syndrome of HF as well as from clinical data obtained from patients may translate to a better understanding of HF and result in specific treatments for these conditions in humans.


Subject(s)
Heart Failure , Stroke Volume , Ventricular Remodeling , Humans , Heart Failure/physiopathology , Heart Failure/immunology , Animals , Myocarditis/physiopathology , Myocarditis/immunology , Ventricular Function, Left , Myocardium/pathology , Myocardium/metabolism , Myocardium/immunology
2.
Inflamm Res ; 73(5): 709-723, 2024 May.
Article in English | MEDLINE | ID: mdl-38546848

ABSTRACT

A role for inflammation in the development and progression of heart failure (HF) has been proposed for decades. Multiple studies have demonstrated the potential involvement of several groups of cytokines and chemokines in acute and chronic HF, though targeting these pathways in early therapeutic trials have produced mixed results. These studies served to highlight the complexity and nuances of how pro-inflammatory pathways contribute to the pathogenesis of HF. More recent investigations have highlighted how inflammation may play distinct roles based on HF syndrome phenotypes, findings that may guide the development of novel therapies. In this review, we propose a contemporary update on the role of inflammation mediated by the innate and adaptive immune systems with HF, highlighting differences that exist across the ejection fraction spectrum. This will specifically be looked at through the lens of established and novel biomarkers of inflammation. Subsequently, we review how improvements in inflammatory pathways may mediate clinical benefits of existing guideline-directed medical therapies for HF, as well as future therapies in the pipeline targeting HF and inflammation.


Subject(s)
Heart Failure , Inflammation , Humans , Heart Failure/physiopathology , Heart Failure/immunology , Heart Failure/drug therapy , Inflammation/immunology , Inflammation/physiopathology , Animals , Cytokines/immunology , Cytokines/metabolism
3.
Artif Organs ; 48(6): 636-645, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38133151

ABSTRACT

BACKGROUND: Neutrophils take part in the innate immune response, phagocytosis, and pro-inflammatory cytokine release. The phagocytic capacity of circulating neutrophils in patients on continuous flow (CF) ventricular assist device (VAD) has not been well studied. METHODS: Blood samples from 14 patients undergoing CF-VAD implantation were collected and analyzed preoperatively (at baseline) and on postoperative days (POD) 3, 7, 14, and 28. Flow cytometry was used to assess the surface expression levels of CD62L, CD162, and macrophage antigen-1 (MAC-1) and neutrophil phagocytic capacity. Interleukin 1 (IL1), IL6, IL8, TNF-α, neutrophil elastase, and myeloperoxidase in plasma were measured using enzyme-linked immunosorbent assays. RESULTS: Among the 14 patients, seven patients had preoperative bridge device support. Relative to baseline, patients with no bridge device had elevated leukocyte count and neutrophil elastase by POD3 which normalized by POD7. Neutrophil activation level, IL6, IL8, and TNF-α increased by POD3 and sustained elevated levels for 7-14 days postoperatively. Elevated neutrophil phagocytic capacity persisted even until POD28. Similar patterns were observed in patients on a preoperative bridge device. CONCLUSIONS: Neutrophil activation and phagocytic capacity increased in response to VAD support, while inflammatory cytokines remain elevated for up to 2 weeks postoperatively. These findings may indicate that VAD implantation elicits circulating neutrophils to an abnormal preemptive phagocytotic phenotype.


Subject(s)
Cytokines , Heart-Assist Devices , Neutrophils , Phagocytosis , Humans , Neutrophils/immunology , Male , Female , Middle Aged , Adult , Cytokines/blood , Neutrophil Activation , Aged , Heart Failure/blood , Heart Failure/immunology , Heart Failure/surgery , Heart Failure/physiopathology
4.
Annu Rev Physiol ; 82: 1-20, 2020 02 10.
Article in English | MEDLINE | ID: mdl-31658002

ABSTRACT

Despite mounting evidence implicating inflammation in cardiovascular diseases, attempts at clinical translation have shown mixed results. Recent preclinical studies have reenergized this field and provided new insights into how to favorably modulate cardiac macrophage function in the context of acute myocardial injury and chronic disease. In this review, we discuss the origins and roles of cardiac macrophage populations in the steady-state and diseased heart, focusing on the human heart and mouse models of ischemia, hypertensive heart disease, and aortic stenosis. Specific attention is given to delineating the roles of tissue-resident and recruited monocyte-derived macrophage subsets. We also highlight emerging concepts of monocyte plasticity and heterogeneity among monocyte-derived macrophages, describe possible mechanisms by which infiltrating monocytes acquire unique macrophage fates, and discuss the putative impact of these populations on cardiac remodeling. Finally, we discuss strategies to target inflammatory macrophage populations.


Subject(s)
Heart Failure/immunology , Heart Failure/therapy , Immunity, Cellular/immunology , Inflammation/immunology , Inflammation/therapy , Animals , Humans , Macrophages/immunology , Monocytes/immunology , Myocardium/immunology , Myocardium/pathology
5.
Int J Mol Sci ; 25(10)2024 May 14.
Article in English | MEDLINE | ID: mdl-38791409

ABSTRACT

Heart failure (HF) poses a significant world health challenge due to the increase in the aging population and advancements in cardiac care. In the pathophysiology of HF, the inflammasome has been correlated with the development, progression, and complications of HF disease. Discovering biomarkers linked to inflammasomes enhances understanding of HF diagnosis and prognosis. Directing inflammasome signaling emerges as an innovative therapeutic strategy for managing HF. The present review aims to delve into this inflammatory cascade, understanding its role in the development of HF, its potential role as biomarker, as well as the prospects of modulating inflammasomes as a therapeutic approach for HF.


Subject(s)
Biomarkers , Heart Failure , Inflammasomes , Humans , Inflammasomes/metabolism , Heart Failure/metabolism , Heart Failure/immunology , Animals , Signal Transduction , Inflammation/metabolism , Inflammation/immunology
6.
Circ Res ; 128(11): 1766-1779, 2021 05 28.
Article in English | MEDLINE | ID: mdl-34043424

ABSTRACT

Cardiac injury remains a major cause of morbidity and mortality worldwide. Despite significant advances, a full understanding of why the heart fails to fully recover function after acute injury, and why progressive heart failure frequently ensues, remains elusive. No therapeutics, short of heart transplantation, have emerged to reliably halt or reverse the inexorable progression of heart failure in the majority of patients once it has become clinically evident. To date, most pharmacological interventions have focused on modifying hemodynamics (reducing afterload, controlling blood pressure and blood volume) or on modifying cardiac myocyte function. However, important contributions of the immune system to normal cardiac function and the response to injury have recently emerged as exciting areas of investigation. Therapeutic interventions aimed at harnessing the power of immune cells hold promise for new treatment avenues for cardiac disease. Here, we review the immune response to heart injury, its contribution to cardiac fibrosis, and the potential of immune modifying therapies to affect cardiac repair.


Subject(s)
Heart Failure/therapy , Heart Injuries/therapy , Immunotherapy/methods , Adaptive Immunity , B-Lymphocytes/physiology , Bioengineering , Cytokines/metabolism , Disease Progression , Eosinophils/physiology , Fibroblasts/physiology , Fibrosis , Heart Failure/etiology , Heart Failure/immunology , Heart Injuries/immunology , Humans , Immunotherapy, Adoptive , Macrophages/physiology , Mast Cells/physiology , Monocytes/physiology , Myocardium/pathology , Myocytes, Cardiac/physiology , Neutrophils/physiology , Receptors, Chimeric Antigen , T-Lymphocytes/physiology , T-Lymphocytes/transplantation
7.
Circ Res ; 128(2): 216-228, 2021 01 22.
Article in English | MEDLINE | ID: mdl-33155517

ABSTRACT

RATIONALE: Clonal hematopoiesis driven by mutations of DNMT3A (DNA methyltransferase 3a) is associated with increased incidence of cardiovascular disease and poor prognosis of patients with chronic heart failure (HF) and aortic stenosis. Although experimental studies suggest that DNMT3A clonal hematopoiesis-driver mutations may enhance inflammation, specific signatures of inflammatory cells in humans are missing. OBJECTIVE: To define subsets of immune cells mediating inflammation in humans using single-cell RNA sequencing. METHODS AND RESULTS: Transcriptomic profiles of peripheral blood mononuclear cells were analyzed in n=6 patients with HF harboring DNMT3A clonal hematopoiesis-driver mutations and n=4 patients with HF and no DNMT3A mutations by single-cell RNA sequencing. Monocytes of patients with HF carrying DNMT3A mutations demonstrated a significantly increased expression of inflammatory genes compared with monocytes derived from patients with HF without DNMT3A mutations. Among the specific upregulated genes were the prototypic inflammatory IL (interleukin) IL1B (interleukin 1B), IL6, IL8, the inflammasome NLRP3, and the macrophage inflammatory proteins CCL3 and CCL4 as well as resistin, which augments monocyte-endothelial adhesion. Silencing of DNMT3A in monocytes induced a paracrine proinflammatory activation and increased adhesion to endothelial cells. Furthermore, the classical monocyte subset of DNMT3A mutation carriers showed increased expression of T-cell stimulating immunoglobulin superfamily members CD300LB, CD83, SIGLEC12, as well as the CD2 ligand and cell adhesion molecule CD58, all of which may be involved in monocyte-T-cell interactions. DNMT3A mutation carriers were further characterized by increased expression of the T-cell alpha receptor constant chain and changes in T helper cell 1, T helper cell 2, T helper cell 17, CD8+ effector, CD4+ memory, and regulatory T-cell-specific signatures. CONCLUSIONS: This study demonstrates that circulating monocytes and T cells of patients with HF harboring clonal hematopoiesis-driver mutations in DNMT3A exhibit a highly inflamed transcriptome, which may contribute to the aggravation of chronic HF.


Subject(s)
Clonal Hematopoiesis/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , Heart Failure/genetics , Immunogenetic Phenomena , Monocytes/enzymology , Mutation , T-Lymphocytes/enzymology , Aged , Cell Adhesion , Chronic Disease , Coculture Techniques , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Female , Heart Failure/enzymology , Heart Failure/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation Mediators/metabolism , Male , Middle Aged , Monocytes/immunology , Paracrine Communication , RNA-Seq , Single-Cell Analysis , T-Lymphocytes/immunology , THP-1 Cells , Transcriptome
9.
Pacing Clin Electrophysiol ; 45(2): 188-195, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34978742

ABSTRACT

BACKGROUND: Pro-inflammatory pathways play an important role in the follow-ups of patients with intracardiac defibrillators (ICDs) for heart failure (HF) reduced with ejection fraction (HFrEF). A newly defined index - the systemic immune-inflammation index (SII)-has recently been reported to have prognostic value in patients with cardiovascular disease. This study's aim is to evaluate the SII value regarding its association with long-term mortality and appropriate ICD therapy during a 10-year follow-up. METHODS: This retrospective study included 1011 patients with ICD for HFrEF. The SII was calculated as the neutrophil-to-lymphocyte ratio × total platelet count in the peripheral blood. The study population was divided into two groups according to the SII's optimal cut-off value to predict long-term mortality. The long-term prognostic impact of SII on these patients was evaluated regarding mortality and appropriate ICD therapy. RESULTS: The patients with a higher SII (≥1119) had significantly higher long-term mortality and appropriate ICD therapy rates. After adjustment for all confounding factors, the long-term mortality rate was 5.1 for a higher SII. (95% CI: 2.9-8.1). The long-term appropriate ICD therapy rate was 2.0 for a higher SII (95% CI: 1.4-3.0). CONCLUSION: SII may be an independent predictive marker for both long-term mortality and appropriate ICD therapy in patients with HFrEF.


Subject(s)
Defibrillators, Implantable , Heart Failure/immunology , Heart Failure/therapy , Inflammation/immunology , Stroke Volume , Aged , Female , Heart Failure/mortality , Humans , Male , Middle Aged , Predictive Value of Tests , Retrospective Studies
10.
Int J Mol Sci ; 23(5)2022 Feb 23.
Article in English | MEDLINE | ID: mdl-35269577

ABSTRACT

Macrophage activation syndrome (MAS) is one of the few entities in rheumatology with the potential to quickly cause multiple organ failure and loss of life, and as such, requires urgent clinical intervention. It has a broad symptomatology, depending on the organs it affects. One especially dangerous aspect of MAS's course of illness is myocarditis leading to acute heart failure and possibly death. Research in recent years has proved that macrophages settled in different organs are not a homogenous group, with particular populations differing in both structure and function. Within the heart, we can determine two major groups, based on the presence of the C-C 2 chemokine receptor (CCR2): CCR2+ and CCR2-. There are a number of studies describing their function and the changes in the population makeup between normal conditions and different illnesses; however, to our knowledge, there has not been one touching on the matter of changes occurring in the populations of heart macrophages during MAS and their possible consequences. This review summarizes the most recent knowledge on heart macrophages, the influence of select cytokines (those particularly significant in the development of MAS) on their activity, and both the immediate and long-term consequences of changes in the makeup of specific macrophage populations-especially the loss of CCR2- cells that are responsible for regenerative processes, as well as the substitution of tissue macrophages by the highly proinflammatory CCR2+ macrophages originating from circulating monocytes. Understanding the significance of these processes may lead to new discoveries that could improve the therapeutic methods in the treatment of MAS.


Subject(s)
Heart Failure/immunology , Macrophage Activation Syndrome/complications , Macrophages/immunology , Receptors, CCR2/metabolism , Disease Progression , Gene Expression Regulation , Heart Failure/etiology , Humans , Macrophage Activation Syndrome/immunology , Myocardium/immunology
11.
Inflammopharmacology ; 30(5): 1617-1622, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35876931

ABSTRACT

Inflammation has a major role in the pathogenesis of heart failure (HF). It triggers a cascade that leads to the release of pro-inflammatory cytokines which in turn cause cardiac hypertrophy, fibrosis, apoptosis, negative inotorpy and leukocyte recruitment which worsen the condition. Neopterin is an inflammatory biomarker which is released as a response to macrophage activation. Levels of neopterin are elevated in conditions which has an immunological component such as autoimmune disease, viral and bacterial infections and malignancy. Neopterin levels were found to be elevated in patients with HF. This is due to the fact that inflammation takes place during the development of the condition. Studies demonstrated that neopterin can be used as a biomarker for diagnosing HF, determining severity of the disease and monitoring its progression. Neopterin levels were higher in patients with New York Heart Association classification (NYHA) III-IV more than class I-II. Moreover, neopterin levels correlated well with morbidity and mortality. It has been suggested that neopterin be monitored levels to determine effectiveness of HF treatment options.


Subject(s)
Heart Failure , Inflammation , Neopterin , Biomarkers/blood , Cytokines , Heart Failure/drug therapy , Heart Failure/immunology , Humans , Inflammation/drug therapy , Inflammation/immunology , Neopterin/blood , Neopterin/immunology
12.
BMC Immunol ; 22(1): 51, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34344299

ABSTRACT

BACKGROUND: Neutrophils induce the synthesis and release of angiopoietin 1 (Ang1), a cytosolic growth factor involved in angiogenesis and capable of inducing several pro-inflammatory activities in neutrophils. Neutrophils also synthesize and release neutrophil extracellular traps (NETs), comprised from decondensed nuclear DNA filaments carrying proteins such as neutrophil elastase (NE), myeloperoxidase (MPO), proteinase 3 (PR3) and calprotectin (S100A8/S100A9), which together, contribute to the innate immune response against pathogens (e.g., bacteria). NETs are involved in various pathological conditions through pro-inflammatory, pro-thrombotic and endothelial dysfunction effects and have recently been found in heart failure (HF) and type 2 diabetes (T2DM) patients. The aim of the present study was to investigate the role of NETs on the synthesis and release of Ang1 by the neutrophils in patients with T2DM and HF with preserved ejection fraction (HFpEF) (stable or acute decompensated; ADHFpEF) with or without T2DM. RESULTS: Our data show that at basal level (PBS) and upon treatment with LPS, levels of NETs are slightly increased in patients suffering from T2DM, HFpEF ± T2DM and ADHF without (w/o) T2DM, whereas this increase was significant in ADHFpEF + T2DM patients compared to healthy control (HC) volunteers and ADHFpEF w/o T2DM. We also observed that treatments with PMA or A23187 increase the synthesis of Ang1 (from 150 to 250%) in HC and this effect is amplified in T2DM and in all cohorts of HF patients. Ang1 is completely released (100%) by neutrophils of all groups and does not bind to NETs as opposed to calprotectin. CONCLUSIONS: Our study suggests that severely ill patients with HFpEF and diabetes synthesize and release a greater abundance of NETs while Ang1 exocytosis is independent of NETs synthesis.


Subject(s)
Angiopoietin-1/metabolism , Diabetes Mellitus, Type 2/immunology , Extracellular Traps/metabolism , Heart Failure/immunology , Neutrophils/immunology , Aged , Aged, 80 and over , Cells, Cultured , Exocytosis , Female , Humans , Immunity, Innate , Leukocyte L1 Antigen Complex/metabolism , Male
13.
Am J Physiol Heart Circ Physiol ; 321(4): H633-H649, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34415184

ABSTRACT

ß-Adrenergic receptors (ßARs) regulate normal and pathophysiological heart function through their impact on contractility. ßARs are also regulators of immune function where they play a unique role depending on the disease condition and immune cell type. Emerging evidence suggests an important role for the ß2AR subtype in regulating remodeling in the pathological heart; however, the importance of these responses has never been examined. In heart failure, catecholamines are elevated, leading to chronic ßAR activation and contributing to the detrimental effects in the heart. We hypothesized that immune cell ß2AR plays a critical role in the development of heart failure in response to chronic catecholamine elevations through their regulation of immune cell infiltration. To test this, chimeric mice were generated by performing bone marrow transplant (BMT) experiments using wild-type (WT) or ß2AR knockout (KO) donors. WT and ß2ARKO BMT mice were chronically administered the ßAR agonist isoproterenol. Immune cell recruitment to the heart was examined by histology and flow cytometry. Numerous changes in immune cell recruitment were observed with isoproterenol administration in WT BMT mice including proinflammatory myeloid populations and lymphocytes with macrophages made up the majority of immune cells in the heart and which were absent in ß2ARKO BMT animal. ß2ARKO BMT mice had decreased cardiomyocyte death, hypertrophy, and interstitial fibrosis following isoproterenol treatment, culminating in improved function. These findings demonstrate an important role for immune cell ß2AR expression in the heart's response to chronically elevated catecholamines.NEW & NOTEWORTHY Immune cell ß2-adrenergic receptors (ß2ARs) are important for proinflammatory macrophage infiltration to the heart in a chronic isoproterenol administration model of heart failure. Mice lacking immune cell ß2AR have decreased immune cell infiltration to their heart, primarily proinflammatory macrophage populations. This decrease culminated to decreased cardiac injury with lessened cardiomyocyte death, decreased interstitial fibrosis and hypertrophy, and improved function demonstrating that ß2AR regulation of immune responses plays an important role in the heart's response to persistent ßAR stimulation.


Subject(s)
Heart Failure/metabolism , Macrophages/metabolism , Myocardium/metabolism , Receptors, Adrenergic, beta-2/metabolism , Adoptive Transfer , Animals , Bone Marrow Transplantation , Cell Death , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Female , Fibrosis , Heart Failure/immunology , Heart Failure/pathology , Heart Failure/physiopathology , Inflammation Mediators/metabolism , Isoproterenol , Lymphocytes/immunology , Lymphocytes/metabolism , Macrophage Activation , Macrophages/immunology , Macrophages/transplantation , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction , Myocardium/immunology , Myocardium/pathology , Phenotype , Receptors, Adrenergic, beta-2/genetics , Signal Transduction , Ventricular Remodeling
14.
Circ Res ; 125(3): 265-281, 2019 07 19.
Article in English | MEDLINE | ID: mdl-31318648

ABSTRACT

Advanced heart failure (HF) is a progressive disease characterized by recurrent hospitalizations and high risk of mortality. Indeed, outcomes in late stages of HF approximate those seen in patients with various aggressive malignancies. Clinical trials assessing beneficial outcomes of new treatments in patients with cancer have used innovative approaches to measure impact on total disease burden or surrogates to assess treatment efficacy. Although most cardiovascular outcomes trials continue to use time-to-first event analyses to assess the primary efficacy end point, such analyses do not adequately reflect the impact of new treatments on the totality of the chronic disease burden. Consequently, patient enrichment and other strategies for ongoing clinical trial design, as well as new statistical methodologies, are important considerations, particularly when studying a population with advanced chronic HF. The DREAM-HF trial (Double-Blind Randomized Assessment of Clinical Events With Allogeneic Mesenchymal Precursor Cells in Advanced Heart Failure) is an ongoing, randomized, sham-controlled phase 3 study of the efficacy and safety of mesenchymal precursor cells as immunotherapy in patients with advanced chronic HF with reduced ejection fraction. Mesenchymal precursor cells have a unique multimodal mechanism of action that is believed to result in polarization of proinflammatory type 1 macrophages in the heart to an anti-inflammatory type 2 macrophage state, inhibition of maladaptive adverse left ventricular remodeling, reversal of cardiac and peripheral endothelial dysfunction, and recovery of deranged vasculature. The objective of DREAM-HF is to confirm earlier phase 2 results and evaluate whether mesenchymal precursor cells will reduce the rate of nonfatal recurrent HF-related major adverse cardiac events while delaying or preventing progression of HF to terminal cardiac events. DREAM-HF is an example of an ongoing contemporary events-driven cardiovascular cell-based immunotherapy study that has utilized the concepts of baseline disease enrichment, prognostic enrichment, and predictive enrichment to improve its efficiency by using accumulating data from within as well as external to the trial. Adaptive enrichment designs and strategies are important components of a rational approach to achieve clinical research objectives in shorter clinical trial timelines and with increased cost-effectiveness without compromising ethical standards or the overall statistical integrity of the study. The DREAM-HF trial also presents an alternative approach to traditional composite time-to-first event primary efficacy end points. Statistical methodologies such as the joint frailty model provide opportunities to expand the scope of events-driven HF with reduced ejection fraction clinical trials to utilize time to recurrent nonfatal HF-related major adverse cardiac events as the primary efficacy end point without compromising the integrity of the statistical analyses for terminal cardiac events. In advanced chronic HF with reduced ejection fraction studies, the joint frailty model is utilized to reflect characteristics of the high-risk patient population with important unmet therapeutic needs. In some cases, use of the joint frailty model may substantially reduce sample size requirements. In addition, using an end point that is acceptable to the Food and Drug Administration and the European Medicines Agency, such as recurrent nonfatal HF-related major adverse cardiac events, enables generation of clinically relevant pharmacoeconomic data while providing comprehensive views of the patient's overall cardiovascular disease burden. The major goal of this review is to provide lessons learned from the ongoing DREAM-HF trial that relate to biologic plausibility and flexible clinical trial design and are potentially applicable to other development programs of innovative therapies for patients with advanced cardiovascular disease. Clinical Trial Registration: URL: https://www.clinicaltrials.gov. Unique identifier: NCT02032004.


Subject(s)
Clinical Trials, Phase III as Topic/methods , Heart Failure/therapy , Immunotherapy/methods , Mesenchymal Stem Cell Transplantation , Multicenter Studies as Topic/methods , Randomized Controlled Trials as Topic/methods , Cell Differentiation , Double-Blind Method , Endothelium, Vascular/physiopathology , Endpoint Determination , Health Services Needs and Demand , Heart Failure/economics , Heart Failure/immunology , Heart Failure/physiopathology , Humans , Inflammation , Macrophages/classification , Macrophages/immunology , Neovascularization, Pathologic/etiology , Research Design , Stroke Volume , Treatment Outcome , Ventricular Remodeling
15.
Circ J ; 85(6): 948-952, 2021 05 25.
Article in English | MEDLINE | ID: mdl-33980782

ABSTRACT

BACKGROUND: Multisystem inflammatory syndrome in children (MIS-C) is a rare syndrome temporally related to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). MIS-C shares similarities with Kawasaki disease, but left ventricular dysfunction is more common in MIS-C.Methods and Results:This study reports the case of a 16-year-old Japanese male patient with MIS-C. Although the initial presentation was severe with circulatory and respiratory failure, the patient recovered completely. Endomyocardial biopsy showed active myocarditis with fibrosis. Immunoglobulin treatment was useful for recovery. CONCLUSIONS: This is the first reported case of MIS-C in Japan. Cardiologists should be aware of MIS-C, a new disease, occurring during the global SARS-CoV-2 pandemic.


Subject(s)
COVID-19/immunology , Heart Failure/immunology , Systemic Inflammatory Response Syndrome/immunology , Acute Disease , Adolescent , COVID-19/diagnosis , COVID-19/therapy , Diagnosis, Differential , Heart Failure/diagnosis , Heart Failure/therapy , Humans , Male , Predictive Value of Tests , Recovery of Function , Systemic Inflammatory Response Syndrome/diagnosis , Systemic Inflammatory Response Syndrome/therapy , Treatment Outcome
16.
Cardiovasc Drugs Ther ; 35(2): 231-247, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33404925

ABSTRACT

The coronavirus disease 19 (COVID-19) pandemic poses a serious global threat to human health and the economy. Based on accumulating evidence, its continuous progression involves not only pulmonary injury but also damage to the cardiovascular system due to intertwined pathophysiological risks. As a point of convergence in the pathophysiologic process between COVID-19 and heart failure (HF), cytokine storm induces the progression of COVID-19 in patients presenting pre-existing or new onset myocardial damage and even HF. Cytokine storm, as a trigger of the progression of HF in patients with COVID-19, has become a novel focus to explore therapies for target populations. In this review, we briefly introduce the basis of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and illuminate the mechanism and links among COVID-19, cytokine storm, and HF. Furthermore, we discuss drugs and therapeutic targets for patients with COVID-19 and HF.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 , Cytokine Release Syndrome , Heart Failure , SARS-CoV-2 , COVID-19/immunology , COVID-19/physiopathology , COVID-19/therapy , Cytokine Release Syndrome/physiopathology , Cytokine Release Syndrome/therapy , Cytokine Release Syndrome/virology , Disease Progression , Heart Failure/diagnosis , Heart Failure/immunology , Heart Failure/therapy , Humans , Immunomodulation/immunology , SARS-CoV-2/pathogenicity , SARS-CoV-2/physiology
17.
BMC Cardiovasc Disord ; 21(1): 333, 2021 07 07.
Article in English | MEDLINE | ID: mdl-34233611

ABSTRACT

BACKGROUND: Inflammation is a key feature of heart failure including HFpEF. The leukocyte count is a marker of inflammation that is widely used in clinical practice. However, there is little available evidence for the relationship between leukocyte count and the outcomes of HFpEF. METHODS: We analyzed data from the TOPCAT (Treatment of Preserved Cardiac Function Heart Failure with an Aldosterone Antagonist) trial. The primary outcome was all-cause mortality, the secondary outcome was composite cardiovascular events and hospitalization for heart failure. Multivariable Cox proportional hazard models were used to compare the risk profiles of patients with leukocyte quartiles, subgroup study divided by sex was also analyzed. RESULTS: The present study included 2898 patients with HFpEF.429 deaths, 671 composite cardiovascular events and 386 hospitalization for heart failure occurred during a mean 3.4 years follow-up. The association between leukocyte count and adverse outcomes followed a U-shaped curve. After multivariable adjustment, the patients with the lowest leukocyte count (Q1) and the highest leukocyte count (Q4) faced higher risk of all-cause death(Q1 vs. Q2, adjusted HR: 1.439; 95% CI: 1.060-1.953, p = 0.020; Q4 vs. Q2, adjusted HR, 1.901; 95%CI: 1.424-2.539, p < 0.001). The subgroup analysis showed a consistent result in female but not male patients. CONCLUSIONS: The association between leukocyte count and risk of adverse outcomes followed a U-shaped curve. Both higher and lower leukocyte count are associated with worse outcomes in patients with HFpEF, which may be attributed to the two sides of inflammation in cardiac remodeling.


Subject(s)
Heart Failure/immunology , Inflammation/immunology , Leukocytes/immunology , Stroke Volume , Ventricular Function, Left , Aged , Female , Heart Failure/drug therapy , Heart Failure/mortality , Heart Failure/physiopathology , Hospitalization , Humans , Inflammation/mortality , Leukocyte Count , Male , Middle Aged , Mineralocorticoid Receptor Antagonists/therapeutic use , Randomized Controlled Trials as Topic , Risk Assessment , Risk Factors , Time Factors , Treatment Outcome
18.
Proc Natl Acad Sci U S A ; 115(20): E4661-E4669, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29712858

ABSTRACT

Nonischemic cardiomyopathy (NICM) resulting from long-standing hypertension, valvular disease, and genetic mutations is a major cause of heart failure worldwide. Recent observations suggest that myeloid cells can impact cardiac function, but the role of tissue-intrinsic vs. tissue-extrinsic myeloid cells in NICM remains poorly understood. Here, we show that cardiac resident macrophage proliferation occurs within the first week following pressure overload hypertrophy (POH; a model of heart failure) and is requisite for the heart's adaptive response. Mechanistically, we identify Kruppel-like factor 4 (KLF4) as a key transcription factor that regulates cardiac resident macrophage proliferation and angiogenic activities. Finally, we show that blood-borne macrophages recruited in late-phase POH are detrimental, and that blockade of their infiltration improves myocardial angiogenesis and preserves cardiac function. These observations demonstrate previously unappreciated temporal and spatial roles for resident and nonresident macrophages in the development of heart failure.


Subject(s)
Cardiomegaly/pathology , Cardiomyopathies/pathology , Heart Failure/pathology , Kruppel-Like Transcription Factors/metabolism , Macrophages/pathology , Myocardium/pathology , Animals , Cardiomegaly/immunology , Cardiomegaly/metabolism , Cardiomyopathies/immunology , Cardiomyopathies/metabolism , Cells, Cultured , Heart Failure/immunology , Heart Failure/metabolism , Kruppel-Like Factor 4 , Macrophages/immunology , Macrophages/metabolism , Mice , Myocardium/immunology , Myocardium/metabolism , Pressure
19.
Curr Heart Fail Rep ; 18(3): 99-106, 2021 06.
Article in English | MEDLINE | ID: mdl-33890193

ABSTRACT

PURPOSE OF THE REVIEW: Coronavirus Disease 2019 (COVID-19) and cardiovascular (CV) disease have a close relationship that emerged from the earliest reports. The aim of this review is to show the possible associations between COVID-19 and heart failure (HF) with preserved ejection fraction (HFpEF). RECENT FINDINGS: In hospitalized patients with COVID-19, the prevalence of HFpEF is high, ranging from 4 to 16%, probably due to the shared cardio-metabolic risk profile. Indeed, comorbidities including hypertension, diabetes, obesity and chronic kidney disease - known predictors of a severe course of COVID-19 - are major causes of HFpEF, too. COVID-19 may represent a precipitating factor leading to acute decompensation of HF in patients with known HFpEF and in those with subclinical diastolic dysfunction, which becomes overt. COVID-19 may also directly or indirectly affect the heart. In otherwise healthy patients, echocardiographic studies showed that the majority of COVID-19 patients present diastolic (rather than systolic) impairment, pulmonary hypertension and right ventricular dysfunction. Such abnormalities are observed both in the acute or subacute phase of COVID-19. Cardiac magnetic resonance reveals myocardial inflammation and fibrosis in up to the 78% of patients in the chronic phase of the disease. These findings suggest that COVID-19 might be a novel independent risk factor for the development of HFpEF, through the activation of a systemic pro-inflammatory state. Follow-up studies are urgently needed to better understand long-term sequelae of COVID-19 inflammatory cardiomyopathy.


Subject(s)
COVID-19 , Heart Failure , COVID-19/epidemiology , COVID-19/physiopathology , Comorbidity , Disease Progression , Heart Failure/epidemiology , Heart Failure/immunology , Heart Failure/virology , Humans , SARS-CoV-2 , Stroke Volume
20.
J Infect Dis ; 221(8): 1315-1320, 2020 03 28.
Article in English | MEDLINE | ID: mdl-31100122

ABSTRACT

Human immunodeficiency virus (HIV) imparts increased heart failure risk to women. Among women with HIV (WHIV), immune pathways relating to heart failure precursors may intimate targets for heart failure prevention strategies. Twenty asymptomatic, antiretroviral-treated WHIV and 14 non-HIV-infected women matched on age and body mass index underwent cardiac magnetic resonance imaging and immune phenotyping. WHIV (vs non-HIV-infected women) exhibited increased myocardial fibrosis (extracellular volume fraction, 0.34 ± 0.06 vs 0.29 ± 0.04; P = .002), reduced diastolic function (diastolic strain rate, 1.10 ± 0.23 s-1 vs 1.39 ± 0.27 s-1; P = .003), and heightened systemic monocyte activation. Among WHIV, soluble CD163 levels correlated with myocardial fibrosis (r = 0.53; P = .02), while circulating inflammatory CD14+CD16+ monocyte CCR2 expression related directly to myocardial fibrosis (r = 0.48; P = .04) and inversely to diastolic function (r = -0.49; P = .03). Clinical Trials Registration. NCT02874703.


Subject(s)
Aging/immunology , Fibrosis/etiology , Fibrosis/immunology , HIV Infections/complications , HIV Infections/immunology , HIV/immunology , Myocardium/immunology , Adult , Aged , Anti-Retroviral Agents/therapeutic use , Cardiomyopathies/etiology , Cardiomyopathies/immunology , Cardiomyopathies/virology , Female , Fibrosis/virology , HIV/drug effects , HIV Infections/drug therapy , Heart/virology , Heart Failure/etiology , Heart Failure/immunology , Heart Failure/virology , Humans , Magnetic Resonance Imaging/methods , Middle Aged , Prospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL