1
|
Dronkers J, van Veldhuisen DJ, van der Meer P, Meems LMG. Heart Failure and Obesity: Unraveling Molecular Mechanisms of Excess Adipose Tissue. J Am Coll Cardiol 2024; 84:1666-1677. [PMID: 39415402 DOI: 10.1016/j.jacc.2024.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/01/2024] [Accepted: 07/11/2024] [Indexed: 10/18/2024]
Abstract
Obesity is an ongoing pandemic and is associated with the development of heart failure (HF), and especially HF with preserved ejection fraction. The definition of obesity is currently based on anthropometric measurements but neglects the location and molecular properties of excess fat. Important depots associated with HF development are subcutaneous adipose tissue and visceral adipose tissue, both located in the abdominal region, and epicardial adipose tissue (EAT) surrounding the myocardium. However, mechanisms linking these different adipose tissue depots to HF development are incompletely understood. EAT in particular is of great interest because of its close proximity to the heart. In this review, we therefore focus on the characteristics of different adipose tissue depots and their response to obesity. In addition, we evaluate how different mechanisms associated with EAT expansion potentially contribute to HF and in particular HF with preserved ejection fraction development.
Collapse
Affiliation(s)
- Just Dronkers
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Dirk J van Veldhuisen
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Peter van der Meer
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Laura M G Meems
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands.
| |
Collapse
|
2
|
Kucsera D, Ruppert M, Sayour NV, Tóth VE, Kovács T, Hegedűs ZI, Onódi Z, Fábián A, Kovács A, Radovits T, Merkely B, Pacher P, Ferdinandy P, Varga ZV. NASH triggers cardiometabolic HFpEF in aging mice. GeroScience 2024; 46:4517-4531. [PMID: 38630423 PMCID: PMC11336017 DOI: 10.1007/s11357-024-01153-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 03/20/2024] [Indexed: 08/22/2024] Open
Abstract
Both heart failure with preserved ejection fraction (HFpEF) and non-alcoholic fatty liver disease (NAFLD) develop due to metabolic dysregulation, has similar risk factors (e.g., insulin resistance, systemic inflammation) and are unresolved clinical challenges. Therefore, the potential link between the two disease is important to study. We aimed to evaluate whether NASH is an independent factor of cardiac dysfunction and to investigate the age dependent effects of NASH on cardiac function. C57Bl/6 J middle aged (10 months old) and aged mice (24 months old) were fed either control or choline deficient (CDAA) diet for 8 weeks. Before termination, echocardiography was performed. Upon termination, organ samples were isolated for histological and molecular analysis. CDAA diet led to the development of NASH in both age groups, without inducing weight gain, allowing to study the direct effect of NASH on cardiac function. Mice with NASH developed hepatomegaly, fibrosis, and inflammation. Aged animals had increased heart weight. Conventional echocardiography revealed normal systolic function in all cohorts, while increased left ventricular volumes in aged mice. Two-dimensional speckle tracking echocardiography showed subtle systolic and diastolic deterioration in aged mice with NASH. Histologic analyses of cardiac samples showed increased cross-sectional area, pronounced fibrosis and Col1a1 gene expression, and elevated intracardiac CD68+ macrophage count with increased Il1b expression. Conventional echocardiography failed to reveal subtle change in myocardial function; however, 2D speckle tracking echocardiography was able to identify diastolic deterioration. NASH had greater impact on aged animals resulting in cardiac hypertrophy, fibrosis, and inflammation.
Collapse
Affiliation(s)
- Dániel Kucsera
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Mihály Ruppert
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Department of Surgical Research and Techniques, Semmelweis University, Budapest, Hungary
| | - Nabil V Sayour
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Viktória E Tóth
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Tamás Kovács
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Zsombor I Hegedűs
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Zsófia Onódi
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Alexandra Fábián
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Attila Kovács
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Department of Surgical Research and Techniques, Semmelweis University, Budapest, Hungary
| | - Tamás Radovits
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Department of Surgical Research and Techniques, Semmelweis University, Budapest, Hungary
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Pál Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health/National Institute On Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary.
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary.
| |
Collapse
|
3
|
Peh ZH, Dihoum A, Hutton D, Arthur JSC, Rena G, Khan F, Lang CC, Mordi IR. Inflammation as a therapeutic target in heart failure with preserved ejection fraction. Front Cardiovasc Med 2023; 10:1125687. [PMID: 37456816 PMCID: PMC10339321 DOI: 10.3389/fcvm.2023.1125687] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 06/15/2023] [Indexed: 07/18/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) accounts for around half of all cases of heart failure and may become the dominant type of heart failure in the near future. Unlike HF with reduced ejection fraction there are few evidence-based treatment strategies available. There is a significant unmet need for new strategies to improve clinical outcomes in HFpEF patients. Inflammation is widely thought to play a key role in HFpEF pathophysiology and may represent a viable treatment target. In this review focusing predominantly on clinical studies, we will summarise the role of inflammation in HFpEF and discuss potential therapeutic strategies targeting inflammation.
Collapse
Affiliation(s)
- Zhen Hui Peh
- School of Medicine, University of Dundee, Ninewells Hospital, Dundee, United Kingdom
| | - Adel Dihoum
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Dana Hutton
- School of Medicine, University of Dundee, Ninewells Hospital, Dundee, United Kingdom
| | - J. Simon C. Arthur
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Graham Rena
- Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Faisel Khan
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Chim C. Lang
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Ify R. Mordi
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
4
|
Wang Y, Wang J, Zheng W, Zhang J, Wang J, Jin T, Tao P, Wang Y, Liu C, Huang J, Lee PY, Yu X, Zhou Q. Identification of an IL-1 receptor mutation driving autoinflammation directs IL-1-targeted drug design. Immunity 2023:S1074-7613(23)00231-5. [PMID: 37315560 DOI: 10.1016/j.immuni.2023.05.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 03/27/2023] [Accepted: 05/17/2023] [Indexed: 06/16/2023]
Abstract
The interleukin 1 (IL-1) pathway signals through IL-1 receptor type 1 (IL-1R1) and emerges as a central mediator for systemic inflammation. Aberrant IL-1 signaling leads to a range of autoinflammatory diseases. Here, we identified a de novo missense variant in IL-1R1 (p.Lys131Glu) in a patient with chronic recurrent multifocal osteomyelitis (CRMO). Patient PBMCs showed strong inflammatory signatures, particularly in monocytes and neutrophils. The p.Lys131Glu substitution affected a critical positively charged amino acid, which disrupted the binding of the antagonist ligand, IL-1Ra, but not IL-1α or IL-1β. This resulted in unopposed IL-1 signaling. Mice with a homologous mutation exhibited similar hyperinflammation and greater susceptibility to collagen antibody-induced arthritis, accompanied with pathological osteoclastogenesis. Leveraging the biology of the mutation, we designed an IL-1 therapeutic, which traps IL-1β and IL-1α, but not IL-1Ra. Collectively, this work provides molecular insights and a potential drug for improved potency and specificity in treating IL-1-driven diseases.
Collapse
Affiliation(s)
- Yusha Wang
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, Zhejiang, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, Zhejiang, China
| | - Jun Wang
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Wenjie Zheng
- Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Jiahui Zhang
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Jinbo Wang
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Taijie Jin
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Panfeng Tao
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, Zhejiang, China
| | - Yibo Wang
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Chenlu Liu
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Jiqian Huang
- Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Pui Y Lee
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xiaomin Yu
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, Zhejiang, China; Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University, Hangzhou 311121, Zhejiang, China; Kidney Disease Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China.
| | - Qing Zhou
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, Zhejiang, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, Zhejiang, China.
| |
Collapse
|
5
|
Manilall A, Mokotedi L, Gunter S, Le Roux R, Fourie S, Flanagan CA, Millen AME. Tumor Necrosis Factor-α Mediates Inflammation-induced Early-Stage Left Ventricular Systolic Dysfunction. J Cardiovasc Pharmacol 2023; 81:411-422. [PMID: 37078863 DOI: 10.1097/fjc.0000000000001428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 03/28/2023] [Indexed: 04/21/2023]
Abstract
ABSTRACT Elevated systemic inflammation contributes to pathogenesis of heart failure with preserved ejection fraction (HFpEF), but molecular mechanisms are poorly understood. Although left ventricular (LV) diastolic dysfunction is the main cause of HFpEF, subclinical systolic dysfunction also contributes. We have previously shown that rats with collagen-induced arthritis (CIA) have systemic inflammation, LV diastolic dysfunction, and that increased circulating TNF-α contributes to inflammation-induced HFpEF pathogenesis, but does not mediate LV diastolic dysfunction in CIA rats. Contribution of systemic inflammation to dysfunction of the active process of LV diastolic and systolic function are unknown. In the present study, we used the CIA rat model to investigate the effects of systemic inflammation and TNF-α blockade on systolic function, and mRNA expression of genes involved in active diastolic relaxation and of myosin heavy chain (MyHC) isoforms. Collagen inoculation and TNF-α blockade did not affect LV mRNA expression of genes that mediate active LV diastolic function. Collagen-induced inflammation impaired LV global longitudinal strain ( P = 0.03) and velocity ( P = 0.04). This impairment of systolic function was prevented by TNF-α blockade. Collagen inoculation decreased mRNA expression of α-MyHC ( Myh6, P = 0.03) and increased expression of β-MyHC ( Myh7, P = 0.0002), a marker, which is upregulated in failing hearts. TNF-α blockade prevented this MyHC isoform-switch. These results show that increased circulating TNF-α changes the relative expression of MyHC isoforms, favoring β-MyHC, which may underlie changes in contractile function that impair systolic function. Our results indicate that TNF-α initiates early-stage LV systolic, rather than LV diastolic dysfunction.
Collapse
Affiliation(s)
- Ashmeetha Manilall
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | | | | | | | | | | | |
Collapse
|
6
|
Fir(e)ing the Rhythm. JACC Basic Transl Sci 2023. [DOI: 10.1016/j.jacbts.2022.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
|
7
|
IL-1β neutralization prevents diastolic dysfunction development, but lacks hepatoprotective effect in an aged mouse model of NASH. Sci Rep 2023; 13:356. [PMID: 36611037 PMCID: PMC9825403 DOI: 10.1038/s41598-022-26896-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 12/21/2022] [Indexed: 01/09/2023] Open
Abstract
Interleukin-1β (IL-1β) is a key mediator of non-alcoholic steatohepatitis (NASH), a chronic liver disease, and of systemic inflammation-driven aging. IL-1β contributes to cardio-metabolic decline, and may promote hepatic oncogenic transformation. Therefore, IL-1β is a potential therapeutic target in these pathologies. We aimed to investigate the hepatic and cardiac effects of an IL-1β targeting monoclonal antibody in an aged mouse model of NASH. 24 months old male C57Bl/6J mice were fed with control or choline deficient (CDAA) diet and were treated with isotype control or anti-IL-1β Mab for 8 weeks. Cardiac functions were assessed by conventional-and 2D speckle tracking echocardiography. Liver samples were analyzed by immunohistochemistry and qRT-PCR. Echocardiography revealed improved cardiac diastolic function in anti-IL-1β treated mice with NASH. Marked hepatic fibrosis developed in CDAA-fed group, but IL-1β inhibition affected fibrosis only at transcriptomic level. Hepatic inflammation was not affected by the IL-1β inhibitor. PCNA staining revealed intensive hepatocyte proliferation in CDAA-fed animals, which was not influenced by neutralization of IL-1β. IL-1β inhibition increased hepatic expression of Pd-1 and Ctla4, while Pd-l1 expression increased in NASH. In conclusion, IL-1β inhibition improved cardiac diastolic function, but did not ameliorate features of NASH; moreover, even promoted hepatic immune checkpoint expression, with concomitant NASH-related hepatocellular proliferation.
Collapse
|
8
|
Larson KF, Malik A, Brozovich FV. Aging and Heart Failure with Preserved Ejection Fraction. Compr Physiol 2022; 12:3813-3822. [PMID: 35950652 DOI: 10.1002/cphy.c210035] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Heart failure is a clinical syndrome characterized by the inability of the cardiovascular system to provide adequate cardiac output at normal filling pressures. This results in a clinical syndrome characterized by dyspnea, edema, and decreased exertional tolerance. Heart failure with preserved ejection fraction (HFpEF) is an increasingly common disease, and the incidence of HFpEF increases with age. There are a variety of factors which contribute to the development of HFpEF, including the presence of hypertension, diabetes, obesity, and other pro-inflammatory states. These comorbid conditions result in changes at the biochemical and cell signaling level which ultimately lead to a disease with a great deal of phenotypic heterogeneity. In general, the physiologic dysfunction of HFpEF is characterized by vascular stiffness, increased cardiac filling pressures, pulmonary hypertension, and impaired volume management. The normal and abnormal processes associated with aging serve as an accelerant in this process, resulting in the hypothesis that HFpEF represents a form of presbycardia. In this article, we aim to review the processes importance of aging in the development of HFpEF by examining the disease and its causes from the biochemical to physiologic level. © 2022 American Physiological Society. Compr Physiol 12: 1-10, 2022.
Collapse
Affiliation(s)
- Kathryn F Larson
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Awais Malik
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Frank V Brozovich
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Department of Physiology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
9
|
Vitiello A, Ferrara F. Pharmacological agents modifying the renin angiotensin and natriuretic peptide systems in COVID-19 patients. Wien Klin Wochenschr 2021; 133:983-988. [PMID: 33877436 PMCID: PMC8055751 DOI: 10.1007/s00508-021-01855-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 03/18/2021] [Indexed: 12/17/2022]
Affiliation(s)
- Antonio Vitiello
- Pharmaceutical Department, Usl Umbria 1, A. Migliorati street, 06132 Perugia, Italy
| | - Francesco Ferrara
- Pharmaceutical Department, Usl Umbria 1, A. Migliorati street, 06132 Perugia, Italy
| |
Collapse
|
10
|
De Luca G, Cavalli G, Campochiaro C, Bruni C, Tomelleri A, Dagna L, Matucci-Cerinic M. Interleukin-1 and Systemic Sclerosis: Getting to the Heart of Cardiac Involvement. Front Immunol 2021; 12:653950. [PMID: 33833766 PMCID: PMC8021854 DOI: 10.3389/fimmu.2021.653950] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/02/2021] [Indexed: 12/12/2022] Open
Abstract
Systemic sclerosis (SSc) is rare, severe connective tissue disease characterized by endothelial and vascular damage, immune activation, and resulting in inflammation and fibrosis of skin and internal organs, including the heart. SSc is associated with high morbidity and mortality. Cardiac involvement is frequent in SSc patients, even though often asymptomatic at early stages, and represents one of the major causes of SSc-related mortality. Heart involvement has a variable clinical presentation, and its pathogenesis is not completely understood. Myocardial fibrosis is traditionally considered the immunopathologic hallmark of heart involvement in SSc. This unique histological feature is paralleled by distinctive clinical and prognostic features. The so-called "vascular hypothesis" represents the most credited hypothesis to explain myocardial fibrosis. More recently, the prominent role of an inflammatory myocardial process has been identified as a cardinal event in the evolution to fibrosis, thus also delineating an "inflammation-driven pathway to fibrosis". The pro-inflammatory cytokine interleukin (IL)-1 has an apical and cardinal role in the myocardial inflammatory cascade and in cardiac dysfunction. The primary aim of this perspective article is: to present the emerging evidence on the role of IL-1 and inflammasome in both SSc and heart inflammation, to review the complex interplay between cellular metabolism and inflammasome activation, and to discuss the rationale for targeted inhibition of IL-1 for the treatment of SSc-heart involvement, providing preliminary experimental and clinical data to support this hypothesis.
Collapse
Affiliation(s)
- Giacomo De Luca
- Unit of Immunology, Rheumatology, Allergy and Rare diseases (UnIRAR), IRCCS San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Giulio Cavalli
- Unit of Immunology, Rheumatology, Allergy and Rare diseases (UnIRAR), IRCCS San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Corrado Campochiaro
- Unit of Immunology, Rheumatology, Allergy and Rare diseases (UnIRAR), IRCCS San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Cosimo Bruni
- Department of Experimental and Clinical Medicine, University of Florence, and Division of Rheumatology AOUC, Florence, Italy
| | - Alessandro Tomelleri
- Unit of Immunology, Rheumatology, Allergy and Rare diseases (UnIRAR), IRCCS San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare diseases (UnIRAR), IRCCS San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, University of Florence, and Division of Rheumatology AOUC, Florence, Italy
| |
Collapse
|
11
|
Dariolli R, Campana C, Gutierrez A, Sobie EA. In vitro and In silico Models to Study SARS-CoV-2 Infection: Integrating Experimental and Computational Tools to Mimic "COVID-19 Cardiomyocyte". Front Physiol 2021; 12:624185. [PMID: 33679437 PMCID: PMC7925402 DOI: 10.3389/fphys.2021.624185] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/25/2021] [Indexed: 01/08/2023] Open
Abstract
The rapid dissemination of SARS-CoV-2 has made COVID-19 a tremendous social, economic, and health burden. Despite the efforts to understand the virus and treat the disease, many questions remain unanswered about COVID-19 mechanisms of infection and progression. Severe Acute Respiratory Syndrome (SARS) infection can affect several organs in the body including the heart, which can result in thromboembolism, myocardial injury, acute coronary syndromes, and arrhythmias. Numerous cardiac adverse events, from cardiomyocyte death to secondary effects caused by exaggerated immunological response against the virus, have been clinically reported. In addition to the disease itself, repurposing of treatments by using "off label" drugs can also contribute to cardiotoxicity. Over the past several decades, animal models and more recently, stem cell-derived cardiomyocytes have been proposed for studying diseases and testing treatments in vitro. In addition, mechanistic in silico models have been widely used for disease and drug studies. In these models, several characteristics such as gender, electrolyte imbalance, and comorbidities can be implemented to study pathophysiology of cardiac diseases and to predict cardiotoxicity of drug treatments. In this Mini Review, we (1) present the state of the art of in vitro and in silico cardiomyocyte modeling currently in use to study COVID-19, (2) review in vitro and in silico models that can be adopted to mimic the effects of SARS-CoV-2 infection on cardiac function, and (3) provide a perspective on how to combine some of these models to mimic "COVID-19 cardiomyocytes environment.".
Collapse
Affiliation(s)
- Rafael Dariolli
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | | | | |
Collapse
|
12
|
Vitiello A, La Porta R, Ferrara F. Scientific hypothesis and rational pharmacological for the use of sacubitril/valsartan in cardiac damage caused by COVID-19. Med Hypotheses 2021; 147:110486. [PMID: 33460992 PMCID: PMC7788318 DOI: 10.1016/j.mehy.2021.110486] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/30/2020] [Accepted: 01/02/2021] [Indexed: 12/17/2022]
Abstract
On March 11, 2020 the World Health Organization (WHO) declared the state of global pandemic caused by the new SARS-CoV-2 (COVID-19). To date, no antivirals directed against SARS-CoV-2 or effective vaccines to combat the viral infection are available. Severe acute respiratory syndrome caused by SARS-CoV-2 is treated empirically with antivirals, anti-inflammatory, anticoagulants. The approval of an effective vaccine still takes time. In this state, it may be useful to find new therapeutic solutions from drugs already on the market. Recent hypotheses suggest that the use of AT-1 receptor antagonists (ARB) in combination with neprilisin inhibitors (NEPi) could indirectly provide clinical benefits to patients with SARS-CoV-2 and cardiac involvement. In this article we investigate and describe a possible innovative pharmacological approach for the treatment of the most severe stages of COVID-19 infection.
Collapse
Affiliation(s)
- Antonio Vitiello
- Pharmaceutical Department, Usl Umbria 1, XIV Settembre Street, 06132 Perugia, Italy.
| | - Raffaele La Porta
- Clinical Pathology, Asur Marche, Viale Guido Da Montefeltro, Urbino, Italy.
| | - Francesco Ferrara
- Pharmaceutical Department, Usl Umbria 1, A. Migliorati Street, 06132 Perugia, Italy.
| |
Collapse
|
13
|
Cavalli G, Colafrancesco S, Emmi G, Imazio M, Lopalco G, Maggio MC, Sota J, Dinarello CA. Interleukin 1α: a comprehensive review on the role of IL-1α in the pathogenesis and treatment of autoimmune and inflammatory diseases. Autoimmun Rev 2021; 20:102763. [PMID: 33482337 DOI: 10.1016/j.autrev.2021.102763] [Citation(s) in RCA: 143] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 12/08/2020] [Indexed: 12/17/2022]
Abstract
The interleukin (IL)-1 family member IL-1α is a ubiquitous and pivotal pro-inflammatory cytokine. The IL-1α precursor is constitutively present in nearly all cell types in health, but is released upon necrotic cell death as a bioactive mediator. IL-1α is also expressed by infiltrating myeloid cells within injured tissues. The cytokine binds the IL-1 receptor 1 (IL-1R1), as does IL-1β, and induces the same pro-inflammatory effects. Being a bioactive precursor released upon tissue damage and necrotic cell death, IL-1α is central to the pathogenesis of numerous conditions characterized by organ or tissue inflammation. These include conditions affecting the lung and respiratory tract, dermatoses and inflammatory skin disorders, systemic sclerosis, myocarditis, pericarditis, myocardial infarction, coronary artery disease, inflammatory thrombosis, as well as complex multifactorial conditions such as COVID-19, vasculitis and Kawasaki disease, Behcet's syndrome, Sjogren Syndrome, and cancer. This review illustrates the clinical relevance of IL-1α to the pathogenesis of inflammatory diseases, as well as the rationale for the targeted inhibition of this cytokine for treatment of these conditions. Three biologics are available to reduce the activities of IL-1α; the monoclonal antibody bermekimab, the IL-1 soluble receptor rilonacept, and the IL-1 receptor antagonist anakinra. These advances in mechanistic understanding and therapeutic management make it incumbent on physicians to be aware of IL-1α and of the opportunity for therapeutic inhibition of this cytokine in a broad spectrum of diseases.
Collapse
Affiliation(s)
- Giulio Cavalli
- Unit of Immunology, Rheumatology, Allergy, and Rare Diseases, IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University, Milan, Italy.
| | - Serena Colafrancesco
- Dipartimento of Clinical Sciences (Internal Medicine, Anesthesia and Resuscitation, and Cardiology), Rheumatology Unit, Sapienza University of Rome, Rome, Italy
| | - Giacomo Emmi
- Department of Experimental and Clinical Medicine, Careggi University Hospital, Firenze, Italy
| | - Massimo Imazio
- University Division of Cardiology, Cardiovascular and Throracic Department, AOU Città della Salute e della Scienza di Torino, Torino, Italy
| | - Giuseppe Lopalco
- Department of Emergency and Organ Transplantation, Rheumatology Unit, University of Bari, Bari, Italy
| | - Maria Cristina Maggio
- Department of Health Promotion, Maternal and Infantile Care, Department of Internal Medicine and Medical Specialties "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Jurgen Sota
- Research Center of Systemic Autoinflammatory Diseases and Behçet's Disease Clinic, Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Charles A Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA.
| |
Collapse
|
14
|
Murphy SP, Kakkar R, McCarthy CP, Januzzi JL. Inflammation in Heart Failure: JACC State-of-the-Art Review. J Am Coll Cardiol 2020; 75:1324-1340. [PMID: 32192660 DOI: 10.1016/j.jacc.2020.01.014] [Citation(s) in RCA: 290] [Impact Index Per Article: 72.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 12/08/2019] [Accepted: 01/06/2020] [Indexed: 02/07/2023]
Abstract
It has long been observed that heart failure (HF) is associated with measures of systemic inflammation. In recent years, there have been significant advancements in our understanding of how inflammation contributes to the pathogenesis and progression of HF. However, although numerous studies have validated the association between measures of inflammation and HF severity and prognosis, clinical trials of anti-inflammatory therapies have proven mostly unsuccessful. On this backdrop emerges the yet unmet goal of targeting precise phenotypes within the syndrome of HF; if such precise definitions can be realized, and with better understanding of the roles played by specific inflammatory mediators, the expectation is that targeted anti-inflammatory therapies may improve prognosis in patients whose HF is driven by inflammatory pathobiology. Here, the authors describe mechanistic links between inflammation and HF, discuss traditional and novel inflammatory biomarkers, and summarize the latest evidence from clinical trials of anti-inflammatory therapies.
Collapse
Affiliation(s)
- Sean P Murphy
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Rahul Kakkar
- Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Cian P McCarthy
- Division of Cardiology, Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - James L Januzzi
- Division of Cardiology, Department of Medicine, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
15
|
Wang M, Scott SR, Koniaris LG, Zimmers TA. Pathological Responses of Cardiac Mitochondria to Burn Trauma. Int J Mol Sci 2020; 21:ijms21186655. [PMID: 32932869 PMCID: PMC7554938 DOI: 10.3390/ijms21186655] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 12/18/2022] Open
Abstract
Despite advances in treatment and care, burn trauma remains the fourth most common type of traumatic injury. Burn-induced cardiac failure is a key factor for patient mortality, especially during the initial post-burn period (the first 24 to 48 h). Mitochondria, among the most important subcellular organelles in cardiomyocytes, are a central player in determining the severity of myocardial damage. Defects in mitochondrial function and structure are involved in pathogenesis of numerous myocardial injuries and cardiovascular diseases. In this article, we comprehensively review the current findings on cardiac mitochondrial pathological changes and summarize burn-impaired mitochondrial respiration capacity and energy supply, induced mitochondrial oxidative stress, and increased cell death. The molecular mechanisms underlying these alterations are discussed, along with the possible influence of other biological variables. We hope this review will provide useful information to explore potential therapeutic approaches that target mitochondria for cardiac protection following burn injury.
Collapse
Affiliation(s)
- Meijing Wang
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (S.R.S.); (L.G.K.); (T.A.Z.)
- Correspondence:
| | - Susan R. Scott
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (S.R.S.); (L.G.K.); (T.A.Z.)
| | - Leonidas G. Koniaris
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (S.R.S.); (L.G.K.); (T.A.Z.)
- Simon Cancer Center, Indiana University, Indianapolis, IN 46202, USA
- Indiana Center for Musculoskeletal Health, Indianopolis, IN 46202, USA
- Center for Cachexia Research Innovation and Therapy, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Teresa A. Zimmers
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (S.R.S.); (L.G.K.); (T.A.Z.)
- Simon Cancer Center, Indiana University, Indianapolis, IN 46202, USA
- Indiana Center for Musculoskeletal Health, Indianopolis, IN 46202, USA
- Center for Cachexia Research Innovation and Therapy, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
| |
Collapse
|
16
|
Tan Y, Zhang Z, Zheng C, Wintergerst KA, Keller BB, Cai L. Mechanisms of diabetic cardiomyopathy and potential therapeutic strategies: preclinical and clinical evidence. Nat Rev Cardiol 2020; 17:585-607. [PMID: 32080423 PMCID: PMC7849055 DOI: 10.1038/s41569-020-0339-2] [Citation(s) in RCA: 383] [Impact Index Per Article: 95.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/16/2020] [Indexed: 02/07/2023]
Abstract
The pathogenesis and clinical features of diabetic cardiomyopathy have been well-studied in the past decade, but effective approaches to prevent and treat this disease are limited. Diabetic cardiomyopathy occurs as a result of the dysregulated glucose and lipid metabolism associated with diabetes mellitus, which leads to increased oxidative stress and the activation of multiple inflammatory pathways that mediate cellular and extracellular injury, pathological cardiac remodelling, and diastolic and systolic dysfunction. Preclinical studies in animal models of diabetes have identified multiple intracellular pathways involved in the pathogenesis of diabetic cardiomyopathy and potential cardioprotective strategies to prevent and treat the disease, including antifibrotic agents, anti-inflammatory agents and antioxidants. Some of these interventions have been tested in clinical trials and have shown favourable initial results. In this Review, we discuss the mechanisms underlying the development of diabetic cardiomyopathy and heart failure in type 1 and type 2 diabetes mellitus, and we summarize the evidence from preclinical and clinical studies that might provide guidance for the development of targeted strategies. We also highlight some of the novel pharmacological therapeutic strategies for the treatment and prevention of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Yi Tan
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA.
- Wendy Novak Diabetes Center, University of Louisville, Norton Children's Hospital, Louisville, KY, USA.
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA.
| | - Zhiguo Zhang
- Department of Cardiology, The First Hospital of Jilin University, Changchun, China
| | - Chao Zheng
- The Second Affiliated Hospital Center of Chinese-American Research Institute for Diabetic Complications, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kupper A Wintergerst
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
- Wendy Novak Diabetes Center, University of Louisville, Norton Children's Hospital, Louisville, KY, USA
- Division of Endocrinology, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Bradley B Keller
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA.
- Wendy Novak Diabetes Center, University of Louisville, Norton Children's Hospital, Louisville, KY, USA.
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA.
- Department of Radiation Oncology, University of Louisville School of Medicine, Louisville, KY, USA.
| |
Collapse
|
17
|
Heijman J, Muna AP, Veleva T, Molina CE, Sutanto H, Tekook M, Wang Q, Abu-Taha IH, Gorka M, Künzel S, El-Armouche A, Reichenspurner H, Kamler M, Nikolaev V, Ravens U, Li N, Nattel S, Wehrens XHT, Dobrev D. Atrial Myocyte NLRP3/CaMKII Nexus Forms a Substrate for Postoperative Atrial Fibrillation. Circ Res 2020; 127:1036-1055. [PMID: 32762493 DOI: 10.1161/circresaha.120.316710] [Citation(s) in RCA: 164] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
RATIONALE Postoperative atrial fibrillation (POAF) is a common and troublesome complication of cardiac surgery. POAF is generally believed to occur when postoperative triggers act on a preexisting vulnerable substrate, but the underlying cellular and molecular mechanisms are largely unknown. OBJECTIVE To identify cellular POAF mechanisms in right atrial samples from patients without a history of atrial fibrillation undergoing open-heart surgery. METHODS AND RESULTS Multicellular action potentials, membrane ion-currents (perforated patch-clamp), or simultaneous membrane-current (ruptured patch-clamp) and [Ca2+]i-recordings in atrial cardiomyocytes, along with protein-expression levels in tissue homogenates or cardiomyocytes, were assessed in 265 atrial samples from patients without or with POAF. No indices of electrical, profibrotic, or connexin remodeling were noted in POAF, but Ca2+-transient amplitude was smaller, although spontaneous sarcoplasmic reticulum (SR) Ca2+-release events and L-type Ca2+-current alternans occurred more frequently. CaMKII (Ca2+/calmodulin-dependent protein kinase-II) protein-expression, CaMKII-dependent phosphorylation of the cardiac RyR2 (ryanodine-receptor channel type-2), and RyR2 single-channel open-probability were significantly increased in POAF. SR Ca2+-content was unchanged in POAF despite greater SR Ca2+-leak, with a trend towards increased SR Ca2+-ATPase activity. Patients with POAF also showed stronger expression of activated components of the NLRP3 (NACHT, LRR, and PYD domains-containing protein-3)-inflammasome system in atrial whole-tissue homogenates and cardiomyocytes. Acute application of interleukin-1β caused NLRP3-signaling activation and CaMKII-dependent RyR2/phospholamban hyperphosphorylation in an immortalized mouse atrial cardiomyocyte cell-line (HL-1-cardiomyocytes) and enhanced spontaneous SR Ca2+-release events in both POAF cardiomyocytes and HL-1-cardiomyocytes. Computational modeling showed that RyR2 dysfunction and increased SR Ca2+-uptake are sufficient to reproduce the Ca2+-handling phenotype and indicated an increased risk of proarrhythmic delayed afterdepolarizations in POAF subjects in response to interleukin-1β. CONCLUSIONS Preexisting Ca2+-handling abnormalities and activation of NLRP3-inflammasome/CaMKII signaling are evident in atrial cardiomyocytes from patients who subsequently develop POAF. These molecular substrates sensitize cardiomyocytes to spontaneous Ca2+-releases and arrhythmogenic afterdepolarizations, particularly upon exposure to inflammatory mediators. Our data reveal a potential cellular and molecular substrate for this important clinical problem.
Collapse
Affiliation(s)
- Jordi Heijman
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (J.H., A.P.M., T.V., C.E.M., M.T., I.H.A.-T., M.G., S.N., D.D.).,Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Maastricht, The Netherlands (J.H., H.S.,)
| | - Azinwi Phina Muna
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (J.H., A.P.M., T.V., C.E.M., M.T., I.H.A.-T., M.G., S.N., D.D.)
| | - Tina Veleva
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (J.H., A.P.M., T.V., C.E.M., M.T., I.H.A.-T., M.G., S.N., D.D.)
| | - Cristina E Molina
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (J.H., A.P.M., T.V., C.E.M., M.T., I.H.A.-T., M.G., S.N., D.D.).,Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf and DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (C.E.M., V.N.)
| | - Henry Sutanto
- Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Maastricht, The Netherlands (J.H., H.S.,)
| | - Marcel Tekook
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (J.H., A.P.M., T.V., C.E.M., M.T., I.H.A.-T., M.G., S.N., D.D.)
| | - Qiongling Wang
- Cardiovascular Research Institute (Q.W., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Molecular Physiology and Biophysics, Medicine, Pediatrics, Neuroscience, and Center for Space Medicine (Q.W., X.H.T.W.), Baylor College of Medicine, Houston, TX
| | - Issam H Abu-Taha
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (J.H., A.P.M., T.V., C.E.M., M.T., I.H.A.-T., M.G., S.N., D.D.)
| | - Marcel Gorka
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (J.H., A.P.M., T.V., C.E.M., M.T., I.H.A.-T., M.G., S.N., D.D.)
| | - Stephan Künzel
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (J.H., A.P.M., T.V., C.E.M., M.T., I.H.A.-T., M.G., S.N., D.D.).,Pharmacology and Toxicology, Medical Faculty, Technische Universität Dresden, Dresden, Germany (S.K., A.E.-A.)
| | - Ali El-Armouche
- Pharmacology and Toxicology, Medical Faculty, Technische Universität Dresden, Dresden, Germany (S.K., A.E.-A.)
| | - Hermann Reichenspurner
- Cardiovascular Surgery, University Heart Center Hamburg and DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (H.R.)
| | - Markus Kamler
- Thoracic and Cardiovascular Surgery, West German Heart and Vascular Center Essen, University Hospital Essen, Essen, Germany (M.K.)
| | - Viacheslav Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf and DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (C.E.M., V.N.)
| | - Ursula Ravens
- Institute of Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany (U.R.).,Institute of Physiology, Medical Faculty Carl Gustav Carus, Technical University of Dresden, Dresden, Germany (U.R.)
| | - Na Li
- Cardiovascular Research Institute (Q.W., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Medicine (Section of Cardiovascular Research) (N.L.), Baylor College of Medicine, Houston, TX
| | - Stanley Nattel
- Medicine, Montreal Heart Institute and Université de Montréal & Department of Pharmacology and Therapeutics, McGill University Montreal, Canada (S.N.).,IHU LIRYC and Fondation Bordeaux Université, Bordeaux, France (S.N.)
| | - Xander H T Wehrens
- Cardiovascular Research Institute (Q.W., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Molecular Physiology and Biophysics, Medicine, Pediatrics, Neuroscience, and Center for Space Medicine (Q.W., X.H.T.W.), Baylor College of Medicine, Houston, TX
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (J.H., A.P.M., T.V., C.E.M., M.T., I.H.A.-T., M.G., S.N., D.D.)
| |
Collapse
|
18
|
Mokotedi L, Michel FS, Mogane C, Gomes M, Woodiwiss AJ, Norton GR, Millen AME. Associations of inflammatory markers with impaired left ventricular diastolic and systolic function in collagen-induced arthritis. PLoS One 2020; 15:e0230657. [PMID: 32208438 PMCID: PMC7092986 DOI: 10.1371/journal.pone.0230657] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/22/2020] [Indexed: 12/19/2022] Open
Abstract
Background High-grade inflammation may play a pivotal role in the pathogenesis of left ventricular (LV) dysfunction. Evidence to support a role of systemic inflammation in mediating impaired LV function in experimental models of rheumatoid arthritis (RA) remains limited. The aim of the present study was to determine the effects of high-grade systemic inflammation on LV diastolic and systolic function in collagen-induced arthritis (CIA). Methods To induce CIA, bovine type-II collagen emulsified in incomplete Freund’s adjuvant was injected at the base of the tail into 21 three-month old Sprague Dawley rats. Nine-weeks after the first immunisation, LV function was assessed by pulsed Doppler, tissue Doppler imaging and Speckle tracking echocardiography. Cardiac collagen content was determined by picrosirius red staining; circulating inflammatory markers were measured using ELISA. Results Compared to controls (n = 12), CIA rats had reduced myocardial relaxation as indexed by lateral e’ (early diastolic mitral annular velocity) and e’/a’ (early-to-late diastolic mitral annular velocity) and increased filling pressures as indexed by E/e’. No differences in ejection fraction and LV endocardial fractional shortening between the groups were recorded. LV global radial and circumferential strain and strain rate were reduced in CIA rats compared to controls. Higher concentrations of circulating inflammatory markers were associated with reduced lateral e’, e’/a’, radial and circumferential strain and strain rate. Greater collagen content was associated with increased concentrations of circulating inflammatory markers and E/e’. Conclusion High-grade inflammation is associated with impaired LV diastolic function and greater myocardial deformation independent of haemodynamic load in CIA rats.
Collapse
Affiliation(s)
- Lebogang Mokotedi
- Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- * E-mail:
| | - Frederic S. Michel
- Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Conrad Mogane
- Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Monica Gomes
- Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Angela J. Woodiwiss
- Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Gavin R. Norton
- Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Aletta M. E. Millen
- Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
19
|
Buckley LF, Abbate A. Interleukin-1 blockade in cardiovascular diseases: a clinical update. Eur Heart J 2019; 39:2063-2069. [PMID: 29584915 DOI: 10.1093/eurheartj/ehy128] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 02/23/2018] [Indexed: 11/13/2022] Open
Abstract
Interleukin-1 (IL-1) is the prototypical pro-inflammatory cytokine. IL-1 was implicated as a cardiodepressant factor in septic shock, and subsequent pre-clinical and clinical research has defined important roles for IL-1 in atherosclerosis, acute myocardial infarction (AMI), and heart failure (HF). IL-1 promotes the formation of the atherosclerotic plaque and facilitates its progression and complication. In a large phase III clinical trial of stable patients with prior AMI, blocking IL-1 activity using a monoclonal antibody prevented recurrent atherothrombotic cardiovascular events. IL-1 also contributes to adverse remodelling and left ventricular dysfunction after AMI, and in phase II studies, IL-1 blockade quenched the inflammatory response associated with ST-segment elevation AMI and prevented HF. In patients with established HF, IL-1 is thought to impair beta-adrenergic receptor signalling and intracellular calcium handling. Phase II studies in patients with HF show improved exercise capacity with IL-1 blockade. Thus, IL-1 blockade is poised to enter the clinical arena as an additional strategy to reduce the residual cardiovascular risk and/or address inflammatory cardiovascular conditions refractory to standard treatments. There are several IL-1 blockers available for clinical use, which differ in mechanism of action, and potentially also efficacy and safety. While IL-1 blockade is not immunosuppressive and not associated with opportunistic infections or an increased risk of cancer, fatal infections may occur more frequently while on treatment with IL-1 blockers likely due to a blunting of the inflammatory signs of infection leading to delayed presentation and diagnosis. We discuss the practical use of IL-1 blockade, including considerations for patient selection and safety monitoring.
Collapse
Affiliation(s)
- Leo F Buckley
- Division of Cardiovascular Medicine and Department of Pharmacy Services, Brigham and Women's Hospital, 45 Francis Street, PBB-AB-314, Boston, MA 02120, USA
| | - Antonio Abbate
- Department of Cardiology, VCU Pauley Heart Center, Virginia Commonwealth University, 1200 E Broad St, Box 980204 Richmond, VA 23298, USA
| |
Collapse
|
20
|
Affiliation(s)
- Jake M. Kieserman
- Division of CardiologyThe Department of MedicineLewis Katz School of Medicine at Temple UniversityPhiladelphiaPA
| | - Valerie D. Myers
- Division of CardiologyThe Department of MedicineLewis Katz School of Medicine at Temple UniversityPhiladelphiaPA
| | - Praveen Dubey
- Division of CardiologyThe Department of MedicineLewis Katz School of Medicine at Temple UniversityPhiladelphiaPA
| | - Joseph Y. Cheung
- Division of CardiologyThe Department of MedicineLewis Katz School of Medicine at Temple UniversityPhiladelphiaPA
| | - Arthur M. Feldman
- Division of CardiologyThe Department of MedicineLewis Katz School of Medicine at Temple UniversityPhiladelphiaPA
| |
Collapse
|
21
|
Broderick TL, Sennott JM, Gutkowska J, Jankowski M. Anti-inflammatory and angiogenic effects of exercise training in cardiac muscle of diabetic mice. Diabetes Metab Syndr Obes 2019; 12:565-573. [PMID: 31118719 PMCID: PMC6499146 DOI: 10.2147/dmso.s197127] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 03/12/2019] [Indexed: 12/27/2022] Open
Abstract
Background: Improved glycemic control and cardiovascular function are major benefits of regular exercise training (ET) in type 2 diabetes. Recent work has demonstrated that ET improves cardiac and vascular functions independent of obesity, inflammation, and glucose control in the diabetic db/db mouse. In this study, we determined whether ET can overcome the effects of elevated inflammatory cytokines and hyperglycemia on markers of cardiac angiogenesis and inflammation in the diabetic mouse. Methods: Male diabetic db/db mice were assigned to a sedentary and exercise-trained group. Sedentary lean control littermates were used as controls. ET was performed at moderate intensity on a treadmill 5 days a week for a period of 8 weeks. After ET, blood was collected for assay of glucose, hemoglobin (HB and HB1AC), C-reactive protein (CRP), and IL-6. Markers of inflammation and insulin resistance (IL-6, IL-1β, and tumor necrosis factor-alpha [TNF-α]) and angiogenesis (endothelial nitric oxide synthase [eNOS], vascular endothelial growth factor-A [VEGF-A], and hypoxia-inducible factor-1α [HIF-1α]) were measured in hearts. Results: Diabetic db/db mice remained obese and hyperglycemic after ET. Percent total HB and HB1AC were significantly higher in ET db/db mice compared to sedentary db/db mice, indicating further deterioration of glucose control with ET. Plasma levels of CRP and IL-6 were higher in sedentary db/db mice compared to control mice and were unaffected by ET. However, in the presence of hyperglycemia and elevated plasma cytokines, protein expression of eNOS, mRNA expression of VEGF-A, and HIF-1α was increased in db/db hearts after ET. On the other hand, protein expression of TNF-α and mRNA expression IL-6 and IL-1β was significantly decreased by ET in hearts of db/db mice. Conclusion: Our results indicate that ET improves cardiac markers of angiogenesis, insulin resistance, and endothelial dysfunction in the db/db mouse. This was observed independently of obesity, hyperglycemia, and the systemic inflammatory state.
Collapse
Affiliation(s)
- Tom L Broderick
- Laboratory of Diabetes and Exercise Metabolism, Department of Physiology, College of Graduate Studies, Midwestern University, Glendale, AZ, USA
- Correspondence: Tom L BroderickLaboratory of Diabetes and Exercise Metabolism, Department of Physiology, College of Graduate Studies, Midwestern University, 19555 North 59 Avenue, Glendale, AZ85308, USATel +1 623 572 3664Fax +1 623 572 3673Email
| | - Jacqueline M Sennott
- Department of Cardiology, Medical Education H23, Saint-Joseph Mercy Health System, Pontiac, MI, USA
| | - Jolanta Gutkowska
- Cardiovascular Biochemistry Laboratory, CRCHUM (7-134), Department of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Marek Jankowski
- Cardiovascular Biochemistry Laboratory, CRCHUM (7-134), Department of Medicine, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
22
|
Li W, Chen M, Xu L, Lv Z, Chen L, Li Y, He W. Morroniside alleviates coxsackievirus B3-induced myocardial damage apoptosis via restraining NLRP3 inflammasome activation. RSC Adv 2019; 9:1222-1229. [PMID: 35518014 PMCID: PMC9059572 DOI: 10.1039/c8ra08662a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 12/19/2018] [Indexed: 02/05/2023] Open
Abstract
Coxsackievirus B3 (CVB3)-induced myocardial damage always leads to serious heart failure by inducing cardiac injury. NLRP3 inflammasome activation has been identified as a central player in the pathogenesis of CVB3-induced viral myocarditis. Therefore, restraining NLRP3 inflammasome activation has been supposed to significantly alleviate the severity of myocardial damage and improve cardiac function. Morroniside (MR), one of the main iridoid glycosides, has the ability to depress the production of reactive oxygen species (ROS) and restrain the expression of caspase-3 and -9. Of importance, ROS and caspase are essential for NLRP3 inflammasome activation in response to CVB3 infection. Therefore, in the present study, MR was selected as a model drug to alleviate CVB3-induced myocardial damage. The results of cardiac function index determination showed that abnormal indexes including mean arterial pressure, heart rate, and left ventricular systolic pressure of myocardial damage rats could be recovered by treating with MR. Such results can be further verified by histopathological evaluation, with the heart tissues of CVB3-infected rats displaying the most amount of H&E and TUNEL positive cells. The underlying mechanism by which MR improves the cardiac function was subsequently investigated. The detection of various gene levels indicated that NLRP3 inflammasome activation was inhibited by MR through down-regulating the expression of pro-inflammatory cytokines: interleukin (IL)-β and IL-18, the pivotal factors that lead to inflammatory responses. More importantly, the related genes, cardiac function indexes, and various myocardial damage markers of normal rats treated with MR did not exhibit any obvious changes compared with the control group, indicating a satisfactory biocompatibility of MR. In summary, MR holds a great potential in the alleviation of CVB3-induced myocardial damage with a negligible cytotoxicity to normal heart tissues. Coxsackievirus B3 (CVB3)-induced myocardial damage always leads to serious heart failure by inducing cardiac injury.![]()
Collapse
Affiliation(s)
- Weidong Li
- Department of Cardiology
- Sichuan University
- West China Hospital
- Chengdu
- China
| | - Mao Chen
- Department of Cardiology
- Sichuan University
- West China Hospital
- Chengdu
- China
| | - Lishuai Xu
- Department of Ophthalmology
- Affiliated Hospital of North Sichuan Medical College
- China
| | - Zhan Lv
- Department of Cardiology
- Affiliated Hospital of North Sichuan Medical College
- China
| | - Li Chen
- Department of Cardiology
- Affiliated Hospital of North Sichuan Medical College
- China
| | - Yiling Li
- Department of Cardiology
- Second Affiliated Hospital of North Sichuan Medical College
- China
| | - WenFen He
- Department of Cardiology
- Affiliated Hospital of North Sichuan Medical College
- China
| |
Collapse
|
23
|
Abstract
Interleukin-1 (IL-1) is the prototypical pro-inflammatory cytokine that occupies an apical place in the inflammatory cascade and also modulates cardiac function, functioning as a soluble cardiodepressant factor. Preclinical research over the past 4 decades has shown that blocking IL-1 processing or activity favorably affects cardiomyocyte survival and cardiac function in experimental animal models, paving the way for clinical studies in patients with heart disease. The promising results of phase II clinical trials of IL-1 blockade in patients with acute myocardial infarction and heart failure have been followed by a successful phase III trial in patients with prior acute myocardial infarction. Three IL-1 blockers with different mechanism of action are currently available for clinical use, although currently none have an indication for heart disease. We herein review the bench-to-bedside clinical translation of IL-1 targeting strategies and discuss the potential use of IL-1 blockade in patients with heart disease.
Collapse
|
24
|
De Luca G, Cavalli G, Campochiaro C, Tresoldi M, Dagna L. Myocarditis: An Interleukin-1-Mediated Disease? Front Immunol 2018; 9:1335. [PMID: 29951067 PMCID: PMC6008311 DOI: 10.3389/fimmu.2018.01335] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 05/29/2018] [Indexed: 12/20/2022] Open
Affiliation(s)
- Giacomo De Luca
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Giulio Cavalli
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy.,Department of Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Corrado Campochiaro
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Moreno Tresoldi
- Unit of General Medicine and Advanced Care, IRCCS San Raffaele Hospital and Scientific Institute, Milan, Italy
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
25
|
Sun XQ, Abbate A, Bogaard HJ. Role of cardiac inflammation in right ventricular failure. Cardiovasc Res 2018; 113:1441-1452. [PMID: 28957536 DOI: 10.1093/cvr/cvx159] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 08/09/2017] [Indexed: 12/18/2022] Open
Abstract
Right ventricular failure (RVF) is the main determinant of mortality in patients with pulmonary arterial hypertension (PAH). Although the exact pathophysiology underlying RVF remains unclear, inflammation may play an important role, as it does in left heart failure. Perivascular pulmonary artery and systemic inflammation is relatively well studied and known to contribute to the initiation and maintenance of the pulmonary vascular insult in PAH. However, less attention has been paid to the role of cardiac inflammation in RVF and PAH. Consistent with many other types of heart failure, cardiac inflammation, triggered by systemic and local stressors, has been shown in RVF patients as well as in RVF animal models. RV inflammation likely contributes to impaired RV contractility, maladaptive remodelling and a vicious circle between RV and pulmonary vascular injury. Although the potential to improve RV function through anti-inflammatory therapy has not been tested, this approach has been applied clinically in left ventricular failure patients, with variable success. Because inflammation plays a dual role in the development of both pulmonary vascular pathology and RVF, anti-inflammatory therapies may have a potential double benefit in patients with PAH and associated RVF.
Collapse
Affiliation(s)
- Xiao-Qing Sun
- Department of Pulmonology, VU University Medical Center/Institute for Cardiovascular Research, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Antonio Abbate
- Department of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Harm-Jan Bogaard
- Department of Pulmonology, VU University Medical Center/Institute for Cardiovascular Research, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
26
|
Miteva K, Pappritz K, Sosnowski M, El-Shafeey M, Müller I, Dong F, Savvatis K, Ringe J, Tschöpe C, Van Linthout S. Mesenchymal stromal cells inhibit NLRP3 inflammasome activation in a model of Coxsackievirus B3-induced inflammatory cardiomyopathy. Sci Rep 2018; 8:2820. [PMID: 29434214 PMCID: PMC5809634 DOI: 10.1038/s41598-018-20686-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 01/16/2018] [Indexed: 12/20/2022] Open
Abstract
Inflammation in myocarditis induces cardiac injury and triggers disease progression to heart failure. NLRP3 inflammasome activation is a newly identified amplifying step in the pathogenesis of myocarditis. We previously have demonstrated that mesenchymal stromal cells (MSC) are cardioprotective in Coxsackievirus B3 (CVB3)-induced myocarditis. In this study, MSC markedly inhibited left ventricular (LV) NOD2, NLRP3, ASC, caspase-1, IL-1β, and IL-18 mRNA expression in CVB3-infected mice. ASC protein expression, essential for NLRP3 inflammasome assembly, increased upon CVB3 infection and was abrogated in MSC-treated mice. Concomitantly, CVB3 infection in vitro induced NOD2 expression, NLRP3 inflammasome activation and IL-1β secretion in HL-1 cells, which was abolished after MSC supplementation. The inhibitory effect of MSC on NLRP3 inflammasome activity in HL-1 cells was partly mediated via secretion of the anti-oxidative protein stanniocalcin-1. Furthermore, MSC application in CVB3-infected mice reduced the percentage of NOD2-, ASC-, p10- and/or IL-1β-positive splenic macrophages, natural killer cells, and dendritic cells. The suppressive effect of MSC on inflammasome activation was associated with normalized expression of prominent regulators of myocardial contractility and fibrosis to levels comparable to control mice. In conclusion, MSC treatment in myocarditis could be a promising strategy limiting the adverse consequences of cardiac and systemic NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Kapka Miteva
- Berlin-Brandenburg Center for Regenerative Therapies, Charité, University Medicine Berlin, Campus Virchow, Berlin, Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Kathleen Pappritz
- Berlin-Brandenburg Center for Regenerative Therapies, Charité, University Medicine Berlin, Campus Virchow, Berlin, Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Marzena Sosnowski
- Berlin-Brandenburg Center for Regenerative Therapies, Charité, University Medicine Berlin, Campus Virchow, Berlin, Germany
| | - Muhammad El-Shafeey
- Berlin-Brandenburg Center for Regenerative Therapies, Charité, University Medicine Berlin, Campus Virchow, Berlin, Germany.,Medical Biotechnology Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications, Alexandria, Egypt
| | - Irene Müller
- Berlin-Brandenburg Center for Regenerative Therapies, Charité, University Medicine Berlin, Campus Virchow, Berlin, Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Fengquan Dong
- Berlin-Brandenburg Center for Regenerative Therapies, Charité, University Medicine Berlin, Campus Virchow, Berlin, Germany
| | - Konstantinos Savvatis
- Berlin-Brandenburg Center for Regenerative Therapies, Charité, University Medicine Berlin, Campus Virchow, Berlin, Germany
| | - Jochen Ringe
- Berlin-Brandenburg Center for Regenerative Therapies, Charité, University Medicine Berlin, Campus Virchow, Berlin, Germany.,Laboratory for Tissue Engineering, Charité, University Medicine Berlin, Berlin, Germany
| | - Carsten Tschöpe
- Berlin-Brandenburg Center for Regenerative Therapies, Charité, University Medicine Berlin, Campus Virchow, Berlin, Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany.,Charité-University-Medicine Berlin, Campus Rudolf Virchow, Department of Cardiology, Berlin, Germany
| | - Sophie Van Linthout
- Berlin-Brandenburg Center for Regenerative Therapies, Charité, University Medicine Berlin, Campus Virchow, Berlin, Germany. .,DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany. .,Charité-University-Medicine Berlin, Campus Rudolf Virchow, Department of Cardiology, Berlin, Germany.
| |
Collapse
|
27
|
Szekely Y, Arbel Y. A Review of Interleukin-1 in Heart Disease: Where Do We Stand Today? Cardiol Ther 2018; 7:25-44. [PMID: 29417406 PMCID: PMC5986669 DOI: 10.1007/s40119-018-0104-3] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Indexed: 12/16/2022] Open
Abstract
Cardiovascular diseases are the leading cause of death worldwide. Research in the last two decades has emphasized the inflammatory process as a key component in the pathogenesis of many of them. The Interleukin-1 family is a pivotal element of inflammation and has been well studied as a therapeutic target in various inflammatory states. Recent trials have explored the effect of Interleukin-1 blockade in cardiovascular diseases and initial evidence of the relevance of such treatment in this field of medicine accumulate. This review will describe the role of Interleukin-1 in heart diseases and the potential therapeutic effect of its blockade in such diseases.
Collapse
Affiliation(s)
- Yishay Szekely
- Department of Cardiology, Tel Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Yaron Arbel
- Department of Cardiology, Tel Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
28
|
Peiró C, Lorenzo Ó, Carraro R, Sánchez-Ferrer CF. IL-1β Inhibition in Cardiovascular Complications Associated to Diabetes Mellitus. Front Pharmacol 2017; 8:363. [PMID: 28659798 PMCID: PMC5468794 DOI: 10.3389/fphar.2017.00363] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 05/26/2017] [Indexed: 01/15/2023] Open
Abstract
Diabetes mellitus (DM) is a chronic disease that affects nowadays millions of people worldwide. In adults, type 2 diabetes mellitus (T2DM) accounts for the majority of all diagnosed cases of diabetes. The course of the T2DM is characterized by insulin resistance and a progressive loss of β-cell mass. DM is associated with a number of related complications, among which cardiovascular complications and atherosclerosis are the main cause of morbidity and mortality in patients suffering from the disease. DM is acknowledged as a low-grade chronic inflammatory state characterized by the over-secretion of pro-inflammatory cytokines, including interleukin (IL)-1β, which reinforce inflammatory signals thus contributing to the development of complications. In this context, the pharmacological approaches to treat diabetes should not only correct hyperglycaemia, but also attenuate inflammation and prevent the development of metabolic and cardiovascular complications. Over the last years, novel biological drugs have been developed to antagonize the pathophysiological actions of IL-1β. The drugs currently used in clinical practice are anakinra, a recombinant form of the naturally occurring IL-1 receptor antagonist, the soluble decoy receptor rilonacept and the monoclonal antibodies canakinumab and gevokizumab. This review will summarize the main experimental and clinical findings obtained with pharmacological IL-1β inhibitors in the context of the cardiovascular complications of DM, and discuss the perspectives of IL-1β inhibitors as novel therapeutic tools for treating these patients.
Collapse
Affiliation(s)
- Concepción Peiró
- Department of Pharmacology, School of Medicine, Universidad Autónoma de MadridMadrid, Spain.,Instituto de Investigación Sanitaria Hospital Universitario de La Paz (IdiPAZ)Madrid, Spain
| | - Óscar Lorenzo
- Department of Medicine, School of Medicine, Universidad Autónoma de MadridMadrid, Spain.,Instituto de Investigación Sanitaria Fundación Jiménez DíazMadrid, Spain
| | - Raffaele Carraro
- Department of Medicine, School of Medicine, Universidad Autónoma de MadridMadrid, Spain.,Service of Endocrinology, Hospital de La PrincesaMadrid, Spain.,Instituto de Investigación Sanitaria Hospital de La PrincesaMadrid, Spain
| | - Carlos F Sánchez-Ferrer
- Department of Pharmacology, School of Medicine, Universidad Autónoma de MadridMadrid, Spain.,Instituto de Investigación Sanitaria Hospital Universitario de La Paz (IdiPAZ)Madrid, Spain
| |
Collapse
|
29
|
De Jesus NM, Wang L, Lai J, Rigor RR, Francis Stuart SD, Bers DM, Lindsey ML, Ripplinger CM. Antiarrhythmic effects of interleukin 1 inhibition after myocardial infarction. Heart Rhythm 2017; 14:727-736. [PMID: 28111350 DOI: 10.1016/j.hrthm.2017.01.027] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Indexed: 01/21/2023]
Abstract
BACKGROUND Interleukin 1β (IL-1β) is a key regulator of the inflammatory response after myocardial infarction (MI) by modulating immune cell recruitment, cytokine production, and extracellular matrix turnover. Elevated levels of IL-1β are associated with adverse remodeling, and inhibition of IL-1 signaling after MI results in improved contractile function. OBJECTIVE The goal of this study was to determine whether IL-1 signaling also contributes to post-MI arrhythmogenesis. METHODS MI was created in 2 murine models of elevated inflammation: atherosclerotic on the Western diet or wild-type with a subseptic dose of lipopolysaccharide. The role of IL-1β was assessed with the IL-1 receptor antagonist anakinra (10 mg/(kg·d), starting 24 hours post-MI). RESULTS In vivo and ex vivo molecular imaging showed reduced myocardial inflammation after a 4-day course of anakinra treatment, despite no change in infarct size. At day 5 post-MI, high-speed optical mapping of transmembrane potential and intracellular Ca2+ in isolated hearts revealed that IL-1β inhibition improved conduction velocity, reduced action potential duration dispersion, improved intracellular Ca2+ handling, decreased transmembrane potential and Ca2+ alternans magnitude, and reduced spontaneous and inducible ventricular arrhythmias. These functional improvements were linked to increased expression of connexin 43 and sarcoplasmic reticulum Ca2+-ATPase. CONCLUSION This study revealed a novel mechanism for IL-1β in contributing to defective excitation-contraction coupling and arrhythmogenesis in the post-MI heart. Our results suggest that inhibition of IL-1 signaling post-MI may represent a novel antiarrhythmic therapy.
Collapse
Affiliation(s)
| | - Lianguo Wang
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, California
| | - Johnny Lai
- Department of Biomedical Engineering, School of Engineering, and
| | - Robert R Rigor
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, California
| | | | - Donald M Bers
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, California
| | - Merry L Lindsey
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi
| | - Crystal M Ripplinger
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, California.
| |
Collapse
|
30
|
Westphal E, Pilowski C, Koch S, Ebelt H, Müller-Werdan U, Werdan K, Loppnow H. Endotoxin-activated cultured neonatal rat cardiomyocytes express functional surface-associated interleukin-1α. ACTA ACUST UNITED AC 2016; 13:25-34. [PMID: 17621543 DOI: 10.1177/0968051907078609] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Interleukin-1 (IL-1) is a potent regulator of cardiovascular proliferation, apoptosis, contraction or production of inflammatory mediators. Thus, we investigated expression and function of IL-1 in cultured neonatal rat heart cells upon endotoxin stimulation. We show that cultured neonatal rat cardiomyocytes expressed IL—1α and IL—1β mRNA. The cells expressed functional cell-associated IL—1 activity and a specific anti-IL—1α-antibody inhibited the activity. Biologically active IL—1α was present at the cell surface of the cardiomyocytes, as indicated in co-culture experiments. Immunohistochemistry showed IL—1α-staining of the neonatal cardiomyocytes. Although the cells also expressed IL—1β mRNA, we did not detect IL—1β in the supernatants of cultured cardiomyocytes by ELISA or in immunohistochemical staining. Furthermore, neonatal and adult rat heart tissues expressed IL—1α mRNA, whereas fetal, but not adult, human cardiac tissues expressed detectable IL—1α mRNA. In contrast, IL-1β mRNA was present in rat and human fetal and adult samples. Furthermore, in patients with dilated or ischemic cardiomyopathy, we measured IL—1β, but not IL—1α, mRNA. These results provide evidence for the presence of functionally active IL—1α on the cell surface of neonatal rat cardiomyocytes and may suggest a differential role of IL—1α in regulation of cellular functions during development, aging and disease in rat and human heart cells.
Collapse
Affiliation(s)
- Elena Westphal
- Martin-Luther-Universität Halle-Wittenberg, Universitätsklinik und Poliklinik für Innere Medizin III, Halle, Germany
| | | | | | | | | | | | | |
Collapse
|
31
|
The crossroads of inflammation, fibrosis, and arrhythmia following myocardial infarction. J Mol Cell Cardiol 2015; 91:114-22. [PMID: 26739214 DOI: 10.1016/j.yjmcc.2015.12.024] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 12/23/2015] [Accepted: 12/26/2015] [Indexed: 01/08/2023]
Abstract
Optimal healing of damaged tissue following myocardial infarction (MI) requires a coordinated cellular response that can be divided into three phases: inflammatory, proliferative/reparative, and maturation. The inflammatory phase, characterized by rapid influx of cytokines, chemokines, and immune cells, is critical to the removal of damaged tissue. The onset of the proliferative/reparative phase is marked by increased proliferation of myofibroblasts and secretion of collagen to replace dead tissue. Lastly, crosslinking of collagen fibers and apoptosis of immune cells marks the maturation phase. Excessive inflammation or fibrosis has been linked to increased incidence of arrhythmia and other MI-related pathologies. This review describes the roles of inflammation and fibrosis in arrhythmogenesis and prospective therapies for anti-arrhythmic treatment.
Collapse
|
32
|
Holloway TL, Rani M, Cap AP, Stewart RM, Schwacha MG. The association between the Th-17 immune response and pulmonary complications in a trauma ICU population. Cytokine 2015; 76:328-333. [PMID: 26364992 DOI: 10.1016/j.cyto.2015.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 08/03/2015] [Accepted: 09/02/2015] [Indexed: 01/03/2023]
Abstract
BACKGROUND The overall immunopathology of the T-helper cell (Th)-17 immune response has been implicated in various inflammatory diseases including pulmonary inflammation; however its potential role in acute respiratory distress syndrome (ARDS) is not defined. This study aimed to evaluate the Th-17 response in bronchoalveolar lavage fluid (BALF) and blood and from trauma patients with pulmonary complications. METHODS A total of 21 severely injured intensive care unit (ICU) subjects, who were mechanically ventilated and undergoing bronchoscopy, were enrolled. BALF and blood were collected and analyzed for Th-1 (interferon [IFN]γ), Th-2 (interleukin [IL]-4, -10), Th-17 (IL-17A, -17F, -22, 23) and pro-inflammatory (IL-1β, IL-6, tumor necrosis factor [TNF]α) cytokine levels. RESULTS Significant levels of the Th-17 cytokines IL-17A, -17F and -21 and IL-6 (which can be classified as a Th-17 cytokine) were observed in the BALF of all subjects. There were no significant differences in Th-17 cytokines between those subjects with ARDS and those without, with the exception of plasma and BALF IL-6, which was markedly greater in ARDS subjects, as compared with controls and non-ARDS subjects. CONCLUSIONS Trauma patients with pulmonary complications exhibited a significant Th-17 response in the lung and blood, suggesting that this pro-inflammatory milieu may be a contributing factor to such complications.
Collapse
Affiliation(s)
- Travis L Holloway
- Department of Surgery, The University of Texas Health Science Center at San Antonio, TX 78229, United States
| | - Meenakshi Rani
- Department of Surgery, The University of Texas Health Science Center at San Antonio, TX 78229, United States
| | - Andrew P Cap
- US Army Institute of Surgical Research, Fort Sam Houston, TX 78234, United States
| | - Ronald M Stewart
- Department of Surgery, The University of Texas Health Science Center at San Antonio, TX 78229, United States
| | - Martin G Schwacha
- Department of Surgery, The University of Texas Health Science Center at San Antonio, TX 78229, United States; US Army Institute of Surgical Research, Fort Sam Houston, TX 78234, United States.
| |
Collapse
|
33
|
Arabacilar P, Marber M. The case for inhibiting p38 mitogen-activated protein kinase in heart failure. Front Pharmacol 2015; 6:102. [PMID: 26029107 PMCID: PMC4428223 DOI: 10.3389/fphar.2015.00102] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 04/24/2015] [Indexed: 11/30/2022] Open
Abstract
This minireview discusses the evidence that the inhibition of p38 mitogen-activated protein kinases (p38 MAPKs) maybe of therapeutic value in heart failure. Most previous experimental studies, as well as past and ongoing clinical trials, have focussed on the role of p38 MAPKs in myocardial infarction and acute coronary syndromes. There is now growing evidence that these kinases are activated within the myocardium of the failing human heart and in the heart and blood vessels of animal models of heart failure. Furthermore, from a philosophical viewpoint the chronic activation of the adaptive stress pathways that lead to the activation of p38 MAPKs in heart failure is analogous to the chronic activation of the sympathetic, renin-aldosterone-angiotensin and neprilysin systems. These have provided some of the most effective therapies for heart failure. This minireview questions whether similar and synergistic advantages would follow the inhibition of p38 MAPKs.
Collapse
Affiliation(s)
- Pelin Arabacilar
- Cardiovascular Division, Department of Cardiology, King's College London British Heart Foundation Centre, The Rayne Institute, St Thomas' Hospital London, UK
| | - Michael Marber
- Cardiovascular Division, Department of Cardiology, King's College London British Heart Foundation Centre, The Rayne Institute, St Thomas' Hospital London, UK
| |
Collapse
|
34
|
Pätilä T, Miyagawa S, Imanishi Y, Fukushima S, Siltanen A, Mervaala E, Kankuri E, Harjula A, Sawa Y. Comparison of arrhythmogenicity and proinflammatory activity induced by intramyocardial or epicardial myoblast sheet delivery in a rat model of ischemic heart failure. PLoS One 2015; 10:e0123963. [PMID: 25860790 PMCID: PMC4393220 DOI: 10.1371/journal.pone.0123963] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 03/08/2015] [Indexed: 01/14/2023] Open
Abstract
Although cell therapy of the failing heart by intramyocardial injections of myoblasts to results in regenerative benefit, it has also been associated with undesired and prospectively fatal arrhythmias. We hypothesized that intramyocardial injections of myoblasts could enhance inflammatory reactivity and facilitate electrical cardiac abnormalities that can be reduced by epicardial myoblast sheet delivery. In a rat model of ischemic heart failure, myoblast therapy either by intramyocardial injections or epicardial cell sheets was given 2 weeks after occlusion of the coronary artery. Ventricular premature contractions (VPCs) were assessed, using an implanted three-lead electrocardiograph at 1, 7, and 14 days after therapy, and 16-point epicardial electropotential mapping (EEPM) was used to evaluate ventricular arrhythmogenicity under isoproterenol stress. Cardiac functioning was assessed by echocardiography. Both transplantation groups showed therapeutic benefit over sham therapy. However, VPCs were more frequent in the Injection group on day 1 and day 14 after therapy than in animals receiving epicardial or sham therapy (p < 0.05 and p < 0.01, respectively). EEPM under isoproterenol stress showed macroreentry at the infarct border area, leading to ventricular tachycardias in the Injection group, but not in the myoblast sheet- or sham-treated groups (p = 0.045). Both transplantation types modified the myocardial cytokine expression profile. In animals receiving epicardial myoblast therapy, selective reductions in the expressions of interferon gamma, interleukin (IL)-1β and IL12 were observed, accompanied by reduced infiltration of inflammatory CD11b- and CD68-positive leukocytes, compared with animals receiving myoblasts as intramyocardial injections. Intramyocardial myoblast delivery was associated with enhanced inflammatory and immunomodulatory reactivity and increased frequency of VPCs. In comparison to intramyocardial injection, the epicardial route may serve as the preferred method of skeletal myoblast transplantation to treat heart failure.
Collapse
Affiliation(s)
- Tommi Pätilä
- Department of Cardiothoracic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Pediatric Cardiac Surgery, Hospital for Children and Adolescents, University of Helsinki, Helsinki, Finland
- * E-mail:
| | - Shigeru Miyagawa
- Department of Cardiothoracic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yukiko Imanishi
- Department of Cardiothoracic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Satsuki Fukushima
- Department of Cardiothoracic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | | | - Eero Mervaala
- Pharmacology, University of Helsinki, Helsinki, Finland
| | - Esko Kankuri
- Pharmacology, University of Helsinki, Helsinki, Finland
| | - Ari Harjula
- Department of Cardiothoracic Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Yoshiki Sawa
- Department of Cardiothoracic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
35
|
|
36
|
Interleukin-1β Blockade Improves Left Ventricular Systolic/Diastolic Function and Restores Contractility Reserve in Severe Ischemic Cardiomyopathy in the Mouse. J Cardiovasc Pharmacol 2014; 64:1-6. [DOI: 10.1097/fjc.0000000000000106] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
37
|
El Khoury N, Mathieu S, Fiset C. Interleukin-1β reduces L-type Ca2+ current through protein kinase Cϵ activation in mouse heart. J Biol Chem 2014; 289:21896-908. [PMID: 24936064 DOI: 10.1074/jbc.m114.549642] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Inflammation is now widely recognized as a key component of heart disease. Patients suffering from arrhythmias and heart failure have increased levels of tumor necrosis factor-α (TNFα) and interleukin-1β (IL-1β). Evidence suggests that these cytokines are important mediators of cardiac remodeling; however, their effects on ion channels and arrhythmogenesis remain incompletely understood. The L-type Ca(2+) current (ICaL) is a major determinant of the plateau phase of cardiac action potential and has a critical excitation-contraction coupling role. Thus, altering its properties could have detrimental effects on cardiac electrical and contractile functions. Accordingly, the objective of this study was to elucidate the effect of TNFα and IL-1β on ICaL, while exploring the underlying regulatory mechanisms. Neonatal mouse ventricular myocytes were treated with a pathophysiological concentration (30 pg/ml) of TNFα and IL-1β for 24 h. Voltage-clamp recordings showed that TNFα had no effect on ICaL, whereas IL-1β decreased the current density by 36%. Although both IL-1β- and TNFα-treated myocytes showed significant increase in reactive oxidative species (ROS), Western blot experiments revealed that only IL-1β increased PKCϵ membrane translocation. The antioxidant N-acetyl-L-cysteine normalized ROS levels and restored ICaL density. Furthermore, the PKCϵ translocation inhibitor ϵ-V1-2 blocked the effect of IL-1β on ICaL. The reduction of ICaL by IL-1β was also seen in cultured adult ventricular myocytes. Overall, chronic IL-1β treatment decreased ICaL density in cardiomyocytes. These effects implicated ROS signaling and PKCϵ activation. These findings could contribute to explain the role of IL-1β in the development of arrhythmia and heart failure.
Collapse
Affiliation(s)
- Nabil El Khoury
- From the Research Center, Montreal Heart Institute, 5000 Bélanger, Montréal, Québec H1T 1C8, the Department of Physiology, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Sophie Mathieu
- From the Research Center, Montreal Heart Institute, 5000 Bélanger, Montréal, Québec H1T 1C8, the Faculty of Pharmacy, Université de Montréal, Montréal, Québec, and
| | - Céline Fiset
- From the Research Center, Montreal Heart Institute, 5000 Bélanger, Montréal, Québec H1T 1C8, the Faculty of Pharmacy, Université de Montréal, Montréal, Québec, and
| |
Collapse
|
38
|
Toldo S, Mezzaroma E, O'Brien L, Marchetti C, Seropian IM, Voelkel NF, Van Tassell BW, Dinarello CA, Abbate A. Interleukin-18 mediates interleukin-1-induced cardiac dysfunction. Am J Physiol Heart Circ Physiol 2014; 306:H1025-31. [PMID: 24531812 PMCID: PMC3962640 DOI: 10.1152/ajpheart.00795.2013] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 02/12/2014] [Indexed: 12/15/2022]
Abstract
Patients with heart failure (HF) have enhanced systemic IL-1 activity, and, in the experimental mouse model, IL-1 induces left ventricular (LV) systolic dysfunction. Whether the effects of IL-1 are direct or mediated by an inducible cytokine, such as IL-18, is unknown. Recombinant human IL-18-binding protein (IL-18BP) or an IL-18-blocking antibody (IL-18AB) was used to neutralize endogenous IL-18 after challenge with the plasma of patients with HF or with recombinant murine IL-1β in adult male mice. Plasma levels of IL-18 and IL-6 (a key mediator of IL-1-induced systemic effects) and LV fractional shortening were measured in mice sedated with pentobarbital sodium (30-50 mg/kg). Mice with genetic deletion of IL-18 or IL-18 receptors were compared with matching wild-type mice. A group of mice received murine IL-18 to evaluate the effects on LV fractional shortening. Plasma from HF patients and IL-1β induced LV systolic dysfunction that was prevented by pretreatment with IL-18AB or IL-18BP. IL-1β failed to induce LV systolic dysfunction in mice with genetic deletion of IL-18 signaling. IL-1β induced a significant increase in plasma IL-18 and IL-6 levels. Genetic or pharmacological inhibition of IL-18 signaling failed to block the induction of IL-6 by IL-1β. In conclusion, IL-1 induces a release of active IL-18 in the mouse that mediates the LV systolic dysfunction but not the induction of IL-6. IL-18 blockade may therefore represent a novel and more targeted therapeutic approach to treat HF.
Collapse
Affiliation(s)
- Stefano Toldo
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Affiliation(s)
- Benjamin W Van Tassell
- VCU Pauley Heart Center (B.W.V.T., S.T., E.M., A.A.), Victoria Johnson Research Laboratory (B.W.V.T., S.T., E.M., A.A.), and School of Pharmacy (B.W.V.T., E.M.), Virginia Commonwealth University, Richmond, VA
| | | | | | | |
Collapse
|
40
|
Le DE, Pascotto M, Leong-Poi H, Sari I, Micari A, Kaul S. Anti-inflammatory and pro-angiogenic effects of beta blockers in a canine model of chronic ischemic cardiomyopathy: comparison between carvedilol and metoprolol. Basic Res Cardiol 2013; 108:384. [PMID: 24072434 DOI: 10.1007/s00395-013-0384-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 08/26/2013] [Accepted: 08/29/2013] [Indexed: 11/30/2022]
Abstract
There is controversy regarding the superiority of carvedilol (C) over metoprolol (M) in congestive heart failure. We hypothesized that C is superior to M in chronic ischemic cardiomyopathy because of its better anti-inflammatory and pro-angiogenic effects. In order to test our hypothesis we used a chronic canine model of multivessel ischemic cardiomyopathy where myocardial microcatheters were placed from which interstitial fluid was collected over time to measure leukocyte count and cytokine levels. After development of left ventricular dysfunction, the animals were randomized into four groups: sham (n = 7), placebo (n = 8), M (n = 11), and C (n = 10), and followed for 3 months after treatment initiation. Tissue was examined for immunohistochemistry, oxidative stress, and capillary density. At 3 months both rest and stress wall thickening were better in C compared to the other groups. At the end of 3 months of treatment end-systolic wall stress also decreased the most in C. Similarly resting myocardial blood flow (MBF) improved the most in C as did the stress endocardial/epicardial MBF. Myocardial interstitial fluid showed greater attenuation of leukocytosis with C compared to M, which was associated with less fibrosis and oxidative stress. C also had higher IL-10 level and capillary density. In conclusion, in a chronic canine model of multivessel ischemic cardiomyopathy we found 3 months of C treatment resulted in better resting global and regional function as well as better regional function at stress compared to M. These changes were associated with higher myocardial levels of the anti-inflammatory cytokine IL-10 and less myocardial oxidative stress, leukocytosis, and fibrosis. Capillary density and MBF were almost normalized. Thus in the doses used in this study, C appears to be superior to M in a chronic canine model of ischemic cardiomyopathy from beneficial effects on inflammation and angiogenesis. Further studies are required for comparing additional doses of these drugs.
Collapse
Affiliation(s)
- D Elizabeth Le
- Hospital and Specialty Medicine - Cardiology, Portland VA Medical Center and Knight Cardiovascular Institute, Portland, OR, USA
| | | | | | | | | | | |
Collapse
|
41
|
Activation of invariant natural killer T cells by α-galactosylceramide ameliorates myocardial ischemia/reperfusion injury in mice. J Mol Cell Cardiol 2013; 62:179-88. [PMID: 23774048 DOI: 10.1016/j.yjmcc.2013.06.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 05/21/2013] [Accepted: 06/05/2013] [Indexed: 11/22/2022]
Abstract
Invariant natural killer T (iNKT) cells orchestrate tissue inflammation via regulating various cytokine productions. However the role of iNKT cells has not been determined in myocardial ischemia/reperfusion (I/R) injury. The purpose of this study was to examine whether the activation of iNKT cells by α-galactosylceramide (α-GC), which specifically activates iNKT cells, could affect myocardial I/R injury. I/R or sham operation was performed in male C57BL/6J mice. I/R mice received the injection of either αGC (I/R+αGC, n=48) or vehicle (I/R+vehicle, n=49) 30 min before reperfusion. After 24h, infarct size/area at risk was smaller in I/R+αGC than in I/R+vehicle (37.8 ± 2.7% vs. 47.1 ± 2.5%, P<0.05), with no significant changes in area at risk. The numbers of infiltrating myeloperoxidase- and CD3-positive cells were lower in I/R+αGC. Apoptosis evaluated by TUNEL staining and caspase-3 protein was also attenuated in I/R+αGC. Myocardial gene expression of tumor necrosis factor-α and interleukin (IL)-1β in I/R+αGC was lower to 46% and 80% of that in I/R+vehicle, respectively, whereas IL-10, IL-4, and interferon (IFN)-γ were higher in I/R+αGC than I/R+vehicle by 2.0, 4.1, and 9.6 folds, respectively. The administration of anti-IL-10 receptor antibody into I/R+αGC abolished the protective effects of αGC on I/R injury (infarct size/area at risk: 53.1 ± 5.2% vs. 37.4 ± 3.5%, P<0.05). In contrast, anti-IL-4 and anti-IFN-γ antibodies did not exert such effects. In conclusion, activated iNKT cells by αGC play a protective role against myocardial I/R injury through the enhanced expression of IL-10. Therapies designed to activate iNKT cells might be beneficial to protect the heart from I/R injury.
Collapse
|
42
|
Fujiu K, Nagai R. Contributions of cardiomyocyte–cardiac fibroblast–immune cell interactions in heart failure development. Basic Res Cardiol 2013; 108:357. [DOI: 10.1007/s00395-013-0357-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 03/13/2013] [Accepted: 03/21/2013] [Indexed: 12/20/2022]
|
43
|
Cardiac intercellular communication: are myocytes and fibroblasts fair-weather friends? J Cardiovasc Transl Res 2012; 5:768-82. [PMID: 23015462 DOI: 10.1007/s12265-012-9404-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2012] [Accepted: 08/20/2012] [Indexed: 10/27/2022]
Abstract
The cardiac fibroblast (CF) has historically been thought of as a quiescent cell of the heart, passively maintaining the extracellular environment for the cardiomyocytes (CM), the functional cardiac cell type. The increasingly appreciated role of the CF, however, extends well beyond matrix production, governing many aspects of cardiac function including cardiac electrophysiology and contractility. Importantly, its contributions to cardiac pathophysiology and pathologic remodeling have created a shift in the field's focus from the CM to the CF as a therapeutic target in the treatment of cardiac diseases. In response to cardiac injury, the CF undergoes a pathologic phenotypic transition into a myofibroblast, characterized by contractile smooth muscle proteins and upregulation of collagens, matrix proteins, and adhesion molecules. Further, the myofibroblast upregulates expression and secretion of a variety of pro-inflammatory, profibrotic mediators, including cytokines, chemokines, and growth factors. These mediators act in both an autocrine fashion to further activate CFs, as well as in a paracrine manner on both CMs and circulating inflammatory cells to induce myocyte dysfunction and chronic inflammation, respectively. Together, cell-specific cytokine-induced effects exacerbate pathologic remodeling and progression to HF. A better understanding of this dynamic intercellular communication will lead to novel targets for the attenuation of cardiac remodeling. Current strategies aimed at targeting cytokines have been largely unsuccessful in clinical trials, lending insights into ways that such intercellular cross talk can be more effectively attenuated. This review will summarize the current knowledge regarding CF functions in the heart and will discuss the regulation and signaling behind CF-mediated cytokine production and function. We will then highlight clinical trials that have exploited cytokine cross talk in the treatment of heart failure and provide novel strategies currently under investigation that may more effectively target pathologic CF-CM communication for the treatment of cardiac disease. This review explores novel mechanisms to directly attenuate heart failure progression through inhibition of signaling downstream of pro-inflammatory cytokines that are elevated after cardiac injury.
Collapse
|
44
|
Abstract
The innate immune system, mediated via toll-like receptors (TLRs), represents the first line of defensive mechanisms that protects hosts from invading microbial pathogens. TLRs are a family of pattern recognition receptors (PRRs), and are pathologically activated by a set of pathogen-associated microbial patterns (PAMPs) and damage-associated molecular patterns (DAMPs). TLRs deliver signals via a specific intracellular signaling pathway involving distinctive adaptor proteins and protein kinases, and ultimately initiate transcriptional factors resulting in inflammatory responses. TLR4 is a paramount type of TLRs, located in the heart, and plays an important role in mediating myocardial ischemic reperfusion (I/R) injury. Loss-of-function experiments and animal models using genetic techniques have found that the MyD88-independent and the MyD88-dependent pathways together participate in the pathological process of myocardial I/R injury. Some other distinctive signaling pathways, such as the PI3K/AKt and AMPK/ERK pathways, interacting with the TLR4 signaling pathway, were also found to be causes of myocardial I/R injury. These different pathways activate a series of downstream transcriptional factors, produced a great quantity of inflammatory cytokines, such as IL, TNF, and initiate inflammatory response. This results in cardiac injury and dysfunction, such as myocardial stunning, no reflow phenomenon, reperfusion arrhythmias and lethal reperfusion injury, and other related complication such as ventricular remodeling. In the future, blockades aimed at blocking the signaling pathway could benefit developments in pharmacology.
Collapse
Affiliation(s)
- Yu Fang
- Department of Cardiovascular and Thoracic Surgery, 2nd Xiangya Hospital, Central South University, Changsha, China
| | | |
Collapse
|
45
|
Lu Y, Li L, Zhao X, Huang W, Wen W. Beta blocker metoprolol protects against contractile dysfunction in rats after coronary microembolization by regulating expression of myocardial inflammatory cytokines. Life Sci 2011; 88:1009-15. [PMID: 21443890 DOI: 10.1016/j.lfs.2011.03.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Revised: 01/15/2011] [Accepted: 03/10/2011] [Indexed: 10/18/2022]
Abstract
AIMS To examine the effects of metoprolol on expression of myocardial inflammatory cytokines and myocardial function in rats following coronary microembolization (CME). MAIN METHODS Male rats were randomly assigned to receive either sham-operation (control group), CME plus saline (CME group), or CME plus metoprolol (metoprolol group). CME was induced by injecting 3000 polyethylene microspheres (42 μm) into the left ventricle during a 10-second occlusion of the ascending aorta. Metoprolol (2.5mg/kg) or saline was administered as three intravenous bolus injections after CME. At 3h, 6h, 12h, 24h and 4 weeks after CME, myocardial function was measured with echocardiography; and the mRNA and protein levels of tumor necrosis factor-α (TNF-α), interleukin-10 (IL-10) and interleukin 1-β (IL-1β) were determined. KEY FINDINGS Induced CME led to markedly higher mRNA and protein levels of TNF-α, IL-1β and IL-10 at 3, 6, 12, and 24h, as well as reduced left ventricular function, compared to the control group. Metoprolol administration reduced TNF-α and IL-1β levels, but increased IL-10 levels at 3, 6, 12, and 24h compared to the CME group. Moreover, metoprolol treatment resulted in significantly improved left ventricular function at 12h, 24h and 4 weeks, but afforded no cardiac protection at 3h and 12h, compared to the CME group. SIGNIFICANCE Higher levels of TNF-α and IL-1β in rats following CME are associated with the development of myocardial contractile dysfunction. Metoprolol-conferred protection against progressive contractile dysfunction following CME may be mediated by its anti-inflammation potential.
Collapse
Affiliation(s)
- Yongguang Lu
- Department of Cardiology, the First Affiliated Hospital of Guangxi Medical University, Nanning city, Guangxi province, China
| | | | | | | | | |
Collapse
|
46
|
Hedayat M, Mahmoudi MJ, Rose NR, Rezaei N. Proinflammatory cytokines in heart failure: double-edged swords. Heart Fail Rev 2011; 15:543-62. [PMID: 20405319 DOI: 10.1007/s10741-010-9168-4] [Citation(s) in RCA: 150] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Increased circulating and intracardiac levels of proinflammatory cytokines have been associated with chronic heart failure. Following an initial insult, the increased production of proinflammatory cytokines, including TNF-α, IL-6, IL-1, and IL-18, jeopardizes the surrounding tissue through propagation of the inflammatory response and direct effects on the cardiac myocyte structure and function. Cardiac myocyte hypertrophy, contractile dysfunction, cardiac myocyte apoptosis, and extracellular matrix remodeling contribute enormously to the development and progression of chronic heart failure. Despite the identification of efficacious pharmacological regimens and introduction of mechanical interventions, chronic heart failure remains among the leading causes of mortality worldwide. To introduce novel therapeutic strategies that modulate the inflammatory response in the context of the failing heart, it is of prime importance to determine the contributions of TNF-α, IL-6, IL-1, and IL-18 in mediating cardiac adaptive and maladaptive responses, as well as delineating their downstream intracellular signaling pathways and their potential therapeutic implications.
Collapse
Affiliation(s)
- Mona Hedayat
- Department of Internal Medicine, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | |
Collapse
|
47
|
Amlodipine inhibits TNF-α production and attenuates cardiac dysfunction induced by lipopolysaccharide involving PI3K/Akt pathway. Int Immunopharmacol 2009; 9:1032-41. [DOI: 10.1016/j.intimp.2009.04.010] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2009] [Revised: 04/12/2009] [Accepted: 04/20/2009] [Indexed: 01/13/2023]
|
48
|
Abstract
Clinical consequences of heart failure are fatigue, dyspnea, and progressive impairment of exercise tolerance. Regular exercise training is associated with health-improving effects. In patients with stable heart failure, exercise training can relieve symptoms, improve exercise capacity and quality of life, as well as reduce hospitalization and, to some extent, risk of mortality. Progressive exercise training is associated with pulmonary, cardiovascular, and skeletal muscle metabolic adaptations that increase oxygen delivery and energy production. This Review focuses on current knowledge of mechanisms by which progressive and moderate exercise training can have sustained beneficial effects on patients with heart failure.
Collapse
|
49
|
Haghighi K, Chen G, Sato Y, Fan GC, He S, Kolokathis F, Pater L, Paraskevaidis I, Jones WK, Dorn GW, Kremastinos DT, Kranias EG. A human phospholamban promoter polymorphism in dilated cardiomyopathy alters transcriptional regulation by glucocorticoids. Hum Mutat 2008; 29:640-7. [PMID: 18241046 DOI: 10.1002/humu.20692] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Depressed calcium handling by the sarcoplasmic reticulum (SR) Ca-ATPase and its regulator phospholamban (PLN) is a key characteristic of human and experimental heart failure. Accumulating evidence indicates that increases in the relative levels of PLN to Ca-ATPase in failing hearts and resulting inhibition of Ca sequestration during diastole, impairs contractility. Here, we identified a genetic variant in the PLN promoter region, which increases its expression and may serve as a genetic modifier in dilated cardiomyopathy (DCM). The variant AF177763.1:g.203A>C (at position -36 bp relative to the PLN transcriptional start site) was found only in the heterozygous form in 1 out of 296 normal subjects and in 22 out of 381 cardiomyopathy patients (heart failure at age of 18-44 years, ejection fraction=22+/-9%). In vitro analysis, using luciferase as a reporter gene in rat neonatal cardiomyocytes, indicated that the PLN-variant increased activity by 24% compared to the wild type. Furthermore, the g.203A>C substitution altered the specific sequence of the steroid receptor for the glucocorticoid nuclear receptor (GR)/transcription factor in the PLN promoter, resulting in enhanced binding to the mutated DNA site. These findings suggest that the g.203A>C genetic variant in the human PLN promoter may contribute to depressed contractility and accelerate functional deterioration in heart failure.
Collapse
Affiliation(s)
- Kobra Haghighi
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, Ohio 45267-0575, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Volonte D, McTiernan CF, Drab M, Kasper M, Galbiati F. Caveolin-1 and caveolin-3 form heterooligomeric complexes in atrial cardiac myocytes that are required for doxorubicin-induced apoptosis. Am J Physiol Heart Circ Physiol 2007; 294:H392-401. [PMID: 17982011 DOI: 10.1152/ajpheart.01039.2007] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Caveolae are 50- to 100-nm invaginations of the plasma membrane. Caveolins are the structural protein components of caveolar membranes. The caveolin gene family is composed of three members: caveolin-1, caveolin-2, and caveolin-3. Caveolin-1 and caveolin-2 are coexpressed in many cell types, including adipocytes, endothelial cells, epithelial cells, and fibroblasts. In contrast, caveolin-3 expression is essentially restricted to skeletal and smooth muscle cells as well as cardiac myocytes. While the interaction between caveolin-1 and caveolin-2 has been documented previously, the reciprocal interaction between endogenous caveolin-1 and caveolin-3 and their functional role in cell types expressing both isoforms have yet to be identified. Here we demonstrate for the first time that caveolin-1 and caveolin-3 are coexpressed in mouse and rat cardiac myocytes of the atria but not ventricles. We also found that caveolin-1 and caveolin-3 can interact and form heterooligomeric complexes in this cell type. Doxorubicin is an effective anticancer agent, but its use is limited by the possible development of cardiotoxicity. Using caveolin-1- and caveolin-3-null mice, we show that both caveolin-1 and caveolin-3 expression are required for doxorubicin-induced apoptosis in the atria through activation of caspase 3. Together, these results bring new insight into the functional role of caveolae and suggest that caveolin-1/caveolin-3 heterooligomeric complexes may play a key role in chemotherapy-induced cardiotoxicity in the atria.
Collapse
Affiliation(s)
- Daniela Volonte
- Department of Pharmacology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | | | | | |
Collapse
|