Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 883
Filter
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
3.
Clin Appl Thromb Hemost ; 30: 10760296241259784, 2024.
Article in English | MEDLINE | ID: mdl-38825589

ABSTRACT

BACKGROUND: Acute heart failure (AHF) carries a grave prognosis, marked by high readmission and mortality rates within 90 days post-discharge. This underscores the urgent need for enhanced care transitions, early monitoring, and precise interventions for at-risk individuals during this critical period. OBJECTIVE: Our study aims to develop and validate an interpretable machine learning (ML) model that integrates peripheral immune cell data with conventional clinical markers. Our goal is to accurately predict 90-day readmission or mortality in patients AHF. METHODS: In our study, we conducted a retrospective analysis on 1210 AHF patients, segregating them into training and external validation cohorts. Patients were categorized based on their 90-day outcomes post-discharge into groups of 'with readmission/mortality' and 'without readmission/mortality'. We developed various ML models using data from peripheral immune cells, traditional clinical indicators, or both, which were then internally validated. The feature importance of the most promising model was examined through the Shapley Additive Explanations (SHAP) method, culminating in external validation. RESULTS: In our cohort of 1210 patients, 28.4% (344) faced readmission or mortality within 90 days post-discharge. Our study pinpointed 10 significant indicators-spanning peripheral immune cells and traditional clinical metrics-that predict these outcomes, with the support vector machine (SVM) model showing superior performance. SHAP analysis further distilled these predictors to five key determinants, including three clinical indicators and two immune cell types, essential for assessing 90-day readmission or mortality risks. CONCLUSION: Our analysis identified the SVM model, which merges traditional clinical indicators and peripheral immune cells, as the most effective for predicting 90-day readmission or mortality in AHF patients. This innovative approach promises to refine risk assessment and enable more targeted interventions for at-risk individuals through continuous improvement.


Subject(s)
Heart Failure , Machine Learning , Patient Readmission , Humans , Heart Failure/mortality , Heart Failure/immunology , Patient Readmission/statistics & numerical data , Male , Female , Aged , Acute Disease , Retrospective Studies , Middle Aged , Prognosis
4.
Sci Immunol ; 9(95): eade3814, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38787963

ABSTRACT

Patients with heart failure (HF) often experience repeated acute decompensation and develop comorbidities such as chronic kidney disease and frailty syndrome. Although this suggests pathological interaction among comorbidities, the mechanisms linking them are poorly understood. Here, we identified alterations in hematopoietic stem cells (HSCs) as a critical driver of recurrent HF and associated comorbidities. Bone marrow transplantation from HF-experienced mice resulted in spontaneous cardiac dysfunction and fibrosis in recipient mice, as well as increased vulnerability to kidney and skeletal muscle insults. HF enhanced the capacity of HSCs to generate proinflammatory macrophages. In HF mice, global chromatin accessibility analysis and single-cell RNA-seq showed that transforming growth factor-ß (TGF-ß) signaling was suppressed in HSCs, which corresponded with repressed sympathetic nervous activity in bone marrow. Transplantation of bone marrow from mice in which TGF-ß signaling was inhibited similarly exacerbated cardiac dysfunction. Collectively, these results suggest that cardiac stress modulates the epigenome of HSCs, which in turn alters their capacity to generate cardiac macrophage subpopulations. This change in HSCs may be a common driver of repeated HF events and comorbidity by serving as a key carrier of "stress memory."


Subject(s)
Heart Failure , Immunity, Innate , Immunologic Memory , Mice, Inbred C57BL , Animals , Heart Failure/immunology , Mice , Male , Multimorbidity , Transforming Growth Factor beta/metabolism , Hematopoietic Stem Cells/immunology , Signal Transduction/immunology , Macrophages/immunology , Trained Immunity
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.
JAMA Cardiol ; 9(6): 574-581, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38583091

ABSTRACT

Importance: High-dose trivalent compared with standard-dose quadrivalent influenza vaccine did not significantly reduce all-cause mortality or cardiopulmonary hospitalizations in patients with high-risk cardiovascular disease in the INVESTED trial. Whether humoral immune response to influenza vaccine is associated with clinical outcomes is unknown. Objective: To examine the antibody response to high-dose trivalent compared with standard-dose quadrivalent inactivated influenza vaccine and its associations with clinical outcomes. Design, Setting, and Participants: This secondary analysis is a prespecified analysis of the immune response substudy of the randomized, double-blind, active-controlled INVESTED trial, which was conducted at 157 sites in the United States and Canada over 3 influenza seasons between September 2016 and January 2019. Antibody titers were determined by hemagglutination inhibition assays at randomization and 4 weeks during the 2017-2018 and 2018-2019 seasons. Eligibility criteria included recent acute myocardial infarction or heart failure hospitalization and at least 1 additional risk factor. Data were analyzed from February 2023 to June 2023. Main Outcomes and Measures: Mean antibody titer change, seroprotection (antibody titer level ≥1:40) and seroconversion (≥4-fold increase in titer) at 4 weeks, and the association between seroconversion status and the risk for adverse clinical outcomes. Interventions: High-dose trivalent or standard-dose quadrivalent inactivated influenza vaccine, with revaccination up to 3 seasons. Results: Antibody data were available for 658 of 5260 randomized participants (12.5%; mean [SD] age, 66.2 [11.4] years; 507 male [77.1%], 151 female [22.9%]; 348 with heart failure [52.9%]). High-dose vaccine was associated with an increased magnitude in antibody titers for A/H1N1, A/H3N2, and B-type antigens compared with standard dose. More than 92% of all participants achieved seroprotection for each of the contained antigens, while seroconversion rates were higher in participants who received high-dose vaccine. Seroconversion for any antigen was not associated with the risk for cardiopulmonary hospitalizations or all-cause mortality (hazard ratio, 1.09; 95% CI, 0.79-1.53; P = .59), irrespective of randomized treatment (P = .38 for interaction). Conclusions and Relevance: High-dose vaccine elicited a more robust humoral response in patients with heart failure or prior myocardial infarction enrolled in the INVESTED trial, with no association between seroconversion status and the risk for cardiopulmonary hospitalizations or all-cause mortality. Vaccination to prevent influenza remains critical in high-risk populations. Trial Registration: ClinicalTrials.gov Identifier: NCT02787044.


Subject(s)
Influenza Vaccines , Influenza, Human , Humans , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Male , Female , Aged , Influenza, Human/prevention & control , Influenza, Human/immunology , Double-Blind Method , Middle Aged , Cardiovascular Diseases/immunology , Cardiovascular Diseases/prevention & control , Cardiovascular Diseases/mortality , Antibodies, Viral/blood , Hospitalization/statistics & numerical data , Hemagglutination Inhibition Tests/methods , Myocardial Infarction/immunology , Heart Failure/immunology
7.
Free Radic Biol Med ; 218: 149-165, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38570171

ABSTRACT

Proper protein degradation is required for cellular protein homeostasis and organ function. Particularly, in post-mitotic cells, such as cardiomyocytes, unbalanced proteolysis due to inflammatory stimuli and oxidative stress contributes to organ dysfunction. To ensure appropriate protein turnover, eukaryotic cells exert two main degradation systems, the ubiquitin-proteasome-system and the autophagy-lysosome-pathway. It has been shown that proteasome activity affects the development of cardiac dysfunction differently, depending on the type of heart failure. Studies analyzing the inducible subtype of the proteasome, the immunoproteasome (i20S), demonstrated that the i20S plays a double role in diseased hearts. While i20S subunits are increased in cardiac hypertrophy, atrial fibrillation and partly in myocarditis, the opposite applies to diabetic cardiomyopathy and ischemia/reperfusion injury. In addition, the i20S appears to play a role in autophagy modulation depending on heart failure phenotype. This review summarizes the current literature on the i20S in different heart failure phenotypes, emphasizing the two faces of i20S in injured hearts. A selection of established i20S inhibitors is introduced and signaling pathways linking the i20S to autophagy are highlighted. Mapping the interplay of the i20S and autophagy in different types of heart failure offers potential approaches for developing treatment strategies against heart failure.


Subject(s)
Autophagy , Heart Failure , Proteasome Endopeptidase Complex , Heart Failure/pathology , Heart Failure/metabolism , Heart Failure/genetics , Heart Failure/immunology , Humans , Proteasome Endopeptidase Complex/metabolism , Animals , Myocytes, Cardiac/pathology , Myocytes, Cardiac/metabolism , Phenotype , Signal Transduction , Proteolysis , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/genetics , Myocarditis/pathology , Myocarditis/metabolism , Myocarditis/immunology , Myocarditis/genetics , Cardiomegaly/pathology , Cardiomegaly/metabolism , Cardiomegaly/genetics
8.
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
10.
Nat Rev Cardiol ; 21(7): 443-462, 2024 07.
Article in English | MEDLINE | ID: mdl-38279046

ABSTRACT

Immune checkpoint molecules are physiological regulators of the adaptive immune response. Immune checkpoint inhibitors (ICIs), such as monoclonal antibodies targeting programmed cell death protein 1 or cytotoxic T lymphocyte-associated protein 4, have revolutionized cancer treatment and their clinical use is increasing. However, ICIs can cause various immune-related adverse events, including acute and chronic cardiotoxicity. Of these cardiovascular complications, ICI-induced acute fulminant myocarditis is the most studied, although emerging clinical and preclinical data are uncovering the importance of other ICI-related chronic cardiovascular complications, such as accelerated atherosclerosis and non-myocarditis-related heart failure. These complications could be more difficult to diagnose, given that they might only be present alongside other comorbidities. The occurrence of these complications suggests a potential role of immune checkpoint molecules in maintaining cardiovascular homeostasis, and disruption of physiological immune checkpoint signalling might thus lead to cardiac pathologies, including heart failure. Although inflammation is a long-known contributor to the development of heart failure, the therapeutic targeting of pro-inflammatory pathways has not been successful thus far. The increasingly recognized role of immune checkpoint molecules in the failing heart highlights their potential use as immunotherapeutic targets for heart failure. In this Review, we summarize the available data on ICI-induced cardiac dysfunction and heart failure, and discuss how immune checkpoint signalling is altered in the failing heart. Furthermore, we describe how pharmacological targeting of immune checkpoints could be used to treat heart failure.


Subject(s)
Heart Failure , Immune Checkpoint Inhibitors , Humans , Heart Failure/drug therapy , Heart Failure/physiopathology , Heart Failure/immunology , Immune Checkpoint Inhibitors/adverse effects , Immune Checkpoint Inhibitors/therapeutic use , Animals , Signal Transduction , Cardiotoxicity
11.
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
12.
Curr Probl Cardiol ; 48(3): 101539, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36528207

ABSTRACT

Heart failure (HF) and chronic obstructive pulmonary disease (COPD) are closely related in clinical practice. This study aimed to investigate the co-genetic characteristics and potential molecular mechanisms of HF and COPD. HF and COPD datasets were downloaded from gene expression omnibus database. After identifying common differentially expressed genes (DEGs), the functional analysis highlighted the critical role of extracellular matrix and ribosomal signaling pathways in both diseases. In addition, GeneMANIA's results suggested that the 2 diseases were related to immune infiltration, and CIBERSORT suggested the role of macrophages. We also discovered 4 TFs and 1408 miRNAs linked to both diseases, and salbutamol may positively affect them.


Subject(s)
Heart Failure , MicroRNAs , Pulmonary Disease, Chronic Obstructive , Humans , Albuterol/therapeutic use , Computational Biology , Heart Failure/drug therapy , Heart Failure/genetics , Heart Failure/immunology , MicroRNAs/genetics , MicroRNAs/immunology , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/immunology
13.
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
14.
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
15.
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
16.
Cardiovasc Res ; 118(1): 169-183, 2022 01 07.
Article in English | MEDLINE | ID: mdl-33560342

ABSTRACT

AIMS: Myocardial infarction (MI) is the most common cause of heart failure (HF) worldwide. G protein-coupled receptor kinase 5 (GRK5) is upregulated in failing human myocardium and promotes maladaptive cardiac hypertrophy in animal models. However, the role of GRK5 in ischemic heart disease is still unknown. In this study, we evaluated whether myocardial GRK5 plays a critical role post-MI in mice and included the examination of specific cardiac immune and inflammatory responses. METHODS AND RESULTS: Cardiomyocyte-specific GRK5 overexpressing transgenic mice (TgGRK5) and non-transgenic littermate control (NLC) mice as well as cardiomyocyte-specific GRK5 knockout mice (GRK5cKO) and wild type (WT) were subjected to MI and, functional as well as structural changes together with outcomes were studied. TgGRK5 post-MI mice showed decreased cardiac function, augmented left ventricular dimension and decreased survival rate compared to NLC post-MI mice. Cardiac hypertrophy and fibrosis as well as fetal gene expression were increased post-MI in TgGRK5 compared to NLC mice. In TgGRK5 mice, GRK5 elevation produced immuno-regulators that contributed to the elevated and long-lasting leukocyte recruitment into the injured heart and ultimately to chronic cardiac inflammation. We found an increased presence of pro-inflammatory neutrophils and macrophages as well as neutrophils, macrophages and T-lymphocytes at 4-days and 8-weeks respectively post-MI in TgGRK5 hearts. Conversely, GRK5cKO mice were protected from ischemic injury and showed reduced early immune cell recruitment (predominantly monocytes) to the heart, improved contractility and reduced mortality compared to WT post-MI mice. Interestingly, cardiomyocyte-specific GRK2 transgenic mice did not share the same phenotype of TgGRK5 mice and did not have increased cardiac leukocyte migration and cytokine or chemokine production post-MI. CONCLUSIONS: Our study shows that myocyte GRK5 has a crucial and GRK-selective role on the regulation of leucocyte infiltration into the heart, cardiac function and survival in a murine model of post-ischemic HF, supporting GRK5 inhibition as a therapeutic target for HF.


Subject(s)
Chemotaxis, Leukocyte , G-Protein-Coupled Receptor Kinase 5/metabolism , Heart Failure/enzymology , Leukocytes/metabolism , Myocardial Infarction/enzymology , Myocytes, Cardiac/enzymology , Ventricular Function, Left , Animals , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , G-Protein-Coupled Receptor Kinase 5/genetics , Heart Failure/immunology , Heart Failure/pathology , Heart Failure/physiopathology , Inflammation Mediators/metabolism , Leukocytes/immunology , Mice, Knockout , Myocardial Contraction , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/pathology , Signal Transduction , Stroke Volume , Transcriptome , Ventricular Pressure
17.
Cardiovasc Res ; 118(1): 37-52, 2022 01 07.
Article in English | MEDLINE | ID: mdl-33537710

ABSTRACT

The interplay between the cardiovascular system, metabolism, and inflammation plays a central role in the pathophysiology of a wide spectrum of cardiovascular diseases, including heart failure. Here, we provide an overview of the fundamental aspects of the interrelation between inflammation and metabolism, ranging from the role of metabolism in immune cell function to the processes how inflammation modulates systemic and cardiac metabolism. Furthermore, we discuss how disruption of this immuno-metabolic interface is involved in the development and progression of cardiovascular disease, with a special focus on heart failure. Finally, we present new technologies and therapeutic approaches that have recently emerged and hold promise for the future of cardiovascular medicine.


Subject(s)
Energy Metabolism , Heart Failure/metabolism , Heart/physiopathology , Immune System/metabolism , Inflammation/metabolism , Myocardium/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Energy Metabolism/drug effects , Heart/drug effects , Heart Failure/drug therapy , Heart Failure/immunology , Heart Failure/physiopathology , Humans , Immune System/drug effects , Immune System/immunology , Immune System/physiopathology , Inflammation/drug therapy , Inflammation/immunology , Inflammation/physiopathology , Inflammation Mediators , Myocardium/immunology , Signal Transduction
18.
J Clin Pathol ; 75(1): 30-33, 2022 Jan.
Article in English | MEDLINE | ID: mdl-33785545

ABSTRACT

AIMS: So far, little has been known on whether myocardial inflammatory infiltration influences heart failure (HF) progression. Thus, the aim of this study was to test the impact of intramyocardial infiltration on clinical outcomes. METHODS: Biopsy samples from 358 patients with stable HF secondary to dilated cardiomyopathy were studied. Immunohistochemistry for lymphocyte (CD3) and macrophage (CD68) markers was performed and counted. After a 1-year follow-up, patients were classified as improved based on the predefined definition of improvement. The clinical data were collected from 324 patients (90.5%). RESULTS: According to the predefined definition of improvement, 133 patients improved (41.0%) but 191 remained unchanged or deteriorated (58.9%). After a 12-month follow-up, the OR with 95% CI of counts of myocardial inflammatory CD68-positive ≥4 cell/high power field (HPF) compared with CD68-positive <4 cell/HPF for lack of improvement was 1.91 (1.65-2.54). However, the number of CD3 positive cell infiltration had no impact on clinical outcome after a 1-year follow-up. In the baseline study, a reasonably negative correlation was found between the number of CD68 positive cells and troponin T (r=-0.39; p<0.001 by Spearman's r). This was corroborated with a low negative correlation between these cells and myocardial form of creatine kinase (CK-MB) fraction (r=-0.27; p=0.006). There was no correlation between CD3 and CD68 positive cells (Spearman's r; r=-0.17, p=0.16). CONCLUSIONS: The current results provide evidence that high macrophage counts may be a predisposing factor for HF progression.


Subject(s)
Cardiomyopathy, Dilated/diagnosis , Cardiovascular Diseases/diagnosis , Heart Failure/diagnosis , Adult , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Biomarkers/metabolism , Biopsy , CD3 Complex/metabolism , Cardiomyopathy, Dilated/immunology , Cardiomyopathy, Dilated/pathology , Cardiovascular Diseases/immunology , Cardiovascular Diseases/pathology , Female , Heart Failure/immunology , Heart Failure/pathology , Humans , Immunohistochemistry , Inflammation , Lymphocytes/immunology , Lymphocytes/pathology , Macrophages/immunology , Macrophages/pathology , Male , Middle Aged , Myocardium/immunology , Myocardium/pathology , Prognosis
19.
Cardiovasc Res ; 118(8): 1964-1977, 2022 06 29.
Article in English | MEDLINE | ID: mdl-34264317

ABSTRACT

AIMS: The exploration of novel immunomodulatory interventions to improve outcome in heart failure (HF) is hampered by the complexity/redundancies of inflammatory pathways, which remain poorly understood. We thus aimed to investigate the associations between the activation of diverse immune processes and outcomes in patients with HF. METHODS AND RESULTS: We measured 355 biomarkers in 2022 patients with worsening HF and an independent validation cohort (n = 1691) (BIOSTAT-CHF index and validation cohorts), and classified them according to their functions into biological processes based on the gene ontology classification. Principal component analyses were used to extract weighted scores per process. We investigated the association of these processes with all-cause mortality at 2-year follow-up. The contribution of each biomarker to the weighted score(s) of the processes was used to identify potential therapeutic targets. Mean age was 69 (±12.0) years and 537 (27%) patients were women. We identified 64 unique overrepresented immune-related processes representing 188 of 355 biomarkers. Of these processes, 19 were associated with all-cause mortality (10 positively and 9 negatively). Increased activation of 'T-cell costimulation' and 'response to interferon-gamma/positive regulation of interferon-gamma production' showed the most consistent positive and negative associations with all-cause mortality, respectively, after external validation. Within T-cell costimulation, inducible costimulator ligand, CD28, CD70, and tumour necrosis factor superfamily member-14 were identified as potential therapeutic targets. CONCLUSIONS: We demonstrate the divergent protective and harmful effects of different immune processes in HF and suggest novel therapeutic targets. These findings constitute a rich knowledge base for informing future studies of inflammation in HF.


Subject(s)
Heart Failure , Aged , Biomarkers , Female , Forecasting , Heart Failure/diagnosis , Heart Failure/immunology , Humans , Immunity , Interferon-gamma , Male , Middle Aged
20.
Front Immunol ; 12: 763647, 2021.
Article in English | MEDLINE | ID: mdl-34745139

ABSTRACT

Heart failure is a complex clinical syndrome characterized by insufficient cardiac function. Heart-resident and infiltrated macrophages have been shown to play important roles in the cardiac remodeling that occurs in response to cardiac pressure overload. However, the possible roles of T cells in this process, have not been well characterized. Here we show that T cell depletion conferred late-stage heart protection but induced cardioprotective hypertrophy at an early stage of heart failure caused by cardiac pressure overload. Single-cell RNA sequencing analysis revealed that CD8+T cell depletion induced cardioprotective hypertrophy characterized with the expression of mitochondrial genes and growth factor receptor genes. CD8+T cells regulated the conversion of both cardiac-resident macrophages and infiltrated macrophages into cardioprotective macrophages expressing growth factor genes such as Areg, Osm, and Igf1, which have been shown to be essential for the myocardial adaptive response after cardiac pressure overload. Our results demonstrate a dynamic interplay between cardiac CD8+T cells and macrophages that is necessary for adaptation to cardiac stress, highlighting the homeostatic functions of resident and infiltrated macrophages in the heart.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Heart Failure/immunology , Macrophages/physiology , Single-Cell Analysis/methods , Animals , Cardiomegaly/etiology , Cell Differentiation , Disease Models, Animal , Macrophages/cytology , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...