1
|
Wang C, Luo H. Crosstalk Between Innate Immunity and Autophagy in Viral Myocarditis Leading to Dilated Cardiomyopathy. Rev Med Virol 2024; 34:e2586. [PMID: 39349889 DOI: 10.1002/rmv.2586] [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: 06/05/2024] [Revised: 09/02/2024] [Accepted: 09/12/2024] [Indexed: 11/08/2024]
Abstract
Viral myocarditis, characterised by inflammation of the heart muscle, presents a significant challenge to global public health, particularly affecting younger individuals and often progressing to dilated cardiomyopathy (DCM), a leading cause of heart failure. Despite ongoing research efforts, viable treatments for this condition remain elusive. Recent studies have shed light on the complex interplay between the innate immune response and autophagy mechanisms, revealing their pivotal roles in the pathogenesis of viral myocarditis and subsequent DCM development. This review aims to delve into the recent advancements in understanding the molecular mechanisms and pathways that intersect innate immunity and autophagy in the context of viral myocarditis. Furthermore, it explores the potential therapeutic implications of these findings, offering insights into promising avenues for the management and treatment of this debilitating condition.
Collapse
Affiliation(s)
- Chen Wang
- Centre for Heart Lung Innovation, St. Paul's Hospital-University of British Columbia, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Honglin Luo
- Centre for Heart Lung Innovation, St. Paul's Hospital-University of British Columbia, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| |
Collapse
|
2
|
Dörr M, Böhm M, Erdmann E, Groß S, Mahabadi AA, Nauck M, Nickening G, Schultheiss HP, Staudt A, Werdan K, Waagstein F, Hjalmarson Å, Felix SB. Multicentre, randomized, double-blind, prospective study on the effects of ImmunoAdSorptiOn on cardiac function in patients with Dilated CardioMyopathy (IASO-DCM): Rationale and design. Eur J Heart Fail 2024. [PMID: 39359033 DOI: 10.1002/ejhf.3476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/11/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024] Open
Abstract
AIMS Pilot studies indicate that immunoadsorption with subsequent IgG substitution (IA/IgG) induces beneficial effects in patients with dilated cardiomyopathy (DCM) and heart failure. This placebo-controlled study investigates whether IA/IgG treatment enhances left ventricular (LV) systolic function as compared to a control group receiving pseudo-treatment. METHODS This multicentre, randomized, double-blind, parallel-group trial aims to include 200 patients with heart failure due to DCM (LV ejection fraction [LVEF] <40%) on optimized guideline-directed heart failure medication. Participants are randomly assigned in a 1:1 ratio to IA/IgG using protein-A columns, or to pseudo-immunoadsorption followed by an intravenous infusion without IgG. Follow-up visits take place by telephone after 1 and 3 months and at the study centres after 6, 12 and 24 months. The primary efficacy endpoint is the change in LVEF from baseline to 6 months determined by contrast echocardiography, analysed at a core lab. In addition, LV end-diastolic and end-systolic volumes will be analysed as secondary endpoints over the entire study period to assess whether IA/IgG affects LV remodelling. As main secondary outcome, a composite of all-cause death, cardiac resuscitation, hospitalization for heart failure, and need for cardiac surgery to improve myocardial pump function will be evaluated after 24 months. In addition, exploratory outcomes as well as safety endpoints related to the treatment will be assessed throughout the whole study period. CONCLUSION IASO-DCM is a randomized study which will provide comprehensive insights into the effects of immunoadsorption with subsequent IgG substitution in patients with DCM.
Collapse
Affiliation(s)
- Marcus Dörr
- Department of Internal Medicine B, University Medicine, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner site Greifswald, Greifswald, Germany
| | - Michael Böhm
- Department of Internal Medicine III, Saarland University, Homburg, Germany
| | - Erland Erdmann
- Department of Internal Medicine III, University of Cologne, Cologne, Germany
| | - Stefan Groß
- Department of Internal Medicine B, University Medicine, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner site Greifswald, Greifswald, Germany
| | - Amir-Abbas Mahabadi
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Hospital Essen, Essen, Germany
| | - Matthias Nauck
- DZHK (German Center for Cardiovascular Research), Partner site Greifswald, Greifswald, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine, Greifswald, Germany
| | - Georg Nickening
- Department of Internal Medicine II - Pneumology/Cardiology, University Hospital Bonn, Bonn, Germany
| | | | - Alexander Staudt
- Helios-Kliniken Schwerin, Department of Cardiology and Angiology, Schwerin, Germany
| | - Karl Werdan
- Department of Internal Medicine III - Cardiology, Angiology and Internal Intensive Care Medicine, Mid-German Heart Center, University Hospital Halle (Saale), Halle, Germany
| | - Finn Waagstein
- Wallenberg Laboratory, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Åke Hjalmarson
- Wallenberg Laboratory, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Stephan B Felix
- Department of Internal Medicine B, University Medicine, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner site Greifswald, Greifswald, Germany
| |
Collapse
|
3
|
McNamara DM, Cooper LT, Arbel Y, Bhimaraj A, Bocchi E, Friedrich MG, Kerneis M, Liu P, Parker AB, Smith ER, Tang WHW, Torre‐Amione G, Tschöpe C. Impact of cannabidiol on myocardial recovery in patients with acute myocarditis: Rationale & design of the ARCHER trial. ESC Heart Fail 2024; 11:3416-3424. [PMID: 38937900 PMCID: PMC11424368 DOI: 10.1002/ehf2.14889] [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: 04/10/2024] [Revised: 05/10/2024] [Accepted: 05/11/2024] [Indexed: 06/29/2024] Open
Abstract
AIMS Acute myocarditis, although a rare disease, can be associated with sudden cardiac death or the need for transplantation in both children and young adults. To date, there is no definitive evidence to support the routine use of immunosuppressive therapy or treatment targeting inflammation in patients with myocarditis. Animal models of cardiovascular (CV), as well as neurological diseases, have demonstrated that cannabidiol has significant anti-inflammatory properties and may represent a promising therapy in acute myocarditis. This efficacy has been shown in a murine model of autoimmune myocarditis as well as in in vitro and in vivo models of heart failure (HF). METHODS AND RESULTS We present the rationale and design of the ARCHER Trial, an international multicentre, double-blind, randomized, placebo-controlled, phase II study examining the safety and efficacy of a pharmaceutically produced cannabidiol formulation, in patients with mild to moderate acute myocarditis. Eligible patients are those with acute myocarditis, randomized within 10 days of the diagnostic cardiac MRI (CMR), which has met defined diagnostic criteria for myocarditis. Oral treatment (cannabidiol or placebo) is titrated from 2.5 mg/kg of body weight up to 10 mg/kg of body weight b.i.d. (or highest tolerated dose) and taken for 12 weeks in addition to standard of care therapy for HF. The primary endpoints are defined as changes in global longitudinal strain (GLS) and extra cellular volume (ECV), measured by CMR at 12 weeks. Assuming 80% power, a 5% alpha risk and 25% missing CMR follow-up data at Week 12, 100 patients are required to demonstrate the desired treatment effect of 18%. The change in left ventricular ejection fraction (LVEF) from baseline to Week 12 was selected as the secondary endpoint. Additional exploratory endpoints include changes in hs-troponin, NT-proBNP, markers of inflammation and endothelial function during the 12-week treatment period. The trial is ongoing but is now more than 50% recruited. As enrolment in the trial continues, no interim data are available for inclusion in this Design paper. CONCLUSIONS The ongoing ARCHER Trial is an international, multicentre, double-blind, randomized, placebo-controlled phase II study, designed to determine the effect of a pharmaceutically produced cannabidiol formulation on CMR parameters in patients presenting with acute myocarditis. Enrolment of 100 patients is expected to conclude in Q3 2024. Study results will be available in early 2025.
Collapse
Affiliation(s)
- Dennis M. McNamara
- Center for Heart FailureUniversity of Pittsburgh Medical CenterPittsburghPennsylvaniaUSA
| | - Leslie T. Cooper
- Department of Cardiovascular MedicineMayo Clinic College of Medicine and ScienceJacksonvilleFloridaUSA
| | - Yaron Arbel
- Sourasky Medical CenterTel Aviv UniversityTel AvivIsrael
| | - Arvind Bhimaraj
- Houston Methodist HospitalHoustonTexasUSA
- Weill Cornell Medical CollegeNew YorkNew YorkUSA
| | - Edimar Bocchi
- Instituto do Coração Hospital das Clinicas HCFMUSP, Faculdade de MedicinaUniversidade de São PauloSão PauloBrazil
| | - Matthias G. Friedrich
- Departments of Medicine and Diagnostic Radiology, Research Institute of the McGill University Health CentreMcGill UniversityMontrealCanada
- Department of CardiologyUniversitätsklinikum HeidelbergHeidelbergGermany
- Department of Cardiac Sciences and RadiologyUniversity of CalgaryCalgaryCanada
| | - Matthieu Kerneis
- Pitié Salpêtrière HospitalParisFrance
- Sorbonne UniversityParisFrance
- ACTION Study GroupParisFrance
| | - Peter Liu
- University of Ottawa Heart InstituteOttawaCanada
| | | | | | - W. H. Wilson Tang
- Heart Vascular and Thoracic InstituteCleveland ClinicClevelandOhioUSA
| | - Guillermo Torre‐Amione
- Instituto de Cardiologia, Hospital Zambrano‐HellionEscuela de Medicina y Ciencias de la Salud, Tecnologico de MonterreyMonterreyMexico
| | - Carsten Tschöpe
- Department of Cardiology, Angiology and Intensive Medicine, Deutsches Herzzentrum der Charité (DHZC)Campus Virchow (CVK)BerlinGermany
- Berlin Institute of Health (BIH) at Charité – Center for Regenerative Therapies (BCRT)BerlinGermany
- German Center for Cardiovascular Research (DZHK); Partner Site BerlinCharité UniversityBerlinGermany
| | | |
Collapse
|
4
|
Frustaci A, Letizia C, Alfarano M, Marchionni G, Verardo R, Chimenti C. Immunomodulating and Immunosuppressive Therapy for Virus-Negative Immune-Mediated Myocarditis. Biomedicines 2024; 12:1565. [PMID: 39062138 PMCID: PMC11274480 DOI: 10.3390/biomedicines12071565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/11/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Myocarditis is an inflammatory disease of the myocardium caused by infectious and noninfectious agents. Clinical manifestations range from mildly symptomatic forms to acute heart failure, cardiogenic shock, life-threatening arrhythmias and sudden death. Myocarditis is still a challenging diagnosis because of its wide variability in clinical presentation and unpredictable course. Moreover, a standardized, specific treatment in not yet available. Immunosuppressive treatment for virus-negative lymphocytic myocarditis is still controversial. Conversely, immunosuppression is well established in sarcoidosis, eosinophilic, giant-cell, drug hypersensitivity, and trauma-related myocarditis as well as lymphocytic myocarditis associated with connective tissue diseases or with the rejection of a transplanted heart. Recently, immunosuppressive therapy has been also recognized as an effective treatment in virus-negative inflammatory cardiomyopathy. The aim of this review is to underline the role of immunomodulating and immunosuppressive therapies in patients with immune-mediated myocarditis and illustrate the different treatment strategies depending on the etiology. An endomyocardial biopsy remains the gold standard for the diagnosis of myocarditis as well as for a tailored treatment.
Collapse
Affiliation(s)
- Andrea Frustaci
- Cellular and Molecular Cardiology Laboratory, IRCCS Lazzaro Spallanzani, 00149 Rome, Italy;
- IRCCS San Raffaele, 00163 Rome, Italy
| | - Claudio Letizia
- Department of Clinical, Internal, Anaesthesiology and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (C.L.); (M.A.); (C.C.)
| | - Maria Alfarano
- Department of Clinical, Internal, Anaesthesiology and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (C.L.); (M.A.); (C.C.)
| | - Giulia Marchionni
- Policlinico San Matteo Pavia IRCCS Foundation, University of Pavia, 27100 Pavia, Italy;
| | - Romina Verardo
- Cellular and Molecular Cardiology Laboratory, IRCCS Lazzaro Spallanzani, 00149 Rome, Italy;
| | - Cristina Chimenti
- Department of Clinical, Internal, Anaesthesiology and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (C.L.); (M.A.); (C.C.)
| |
Collapse
|
5
|
Goldberg JF, Spinner JA, Soslow JH. Myocarditis in children 2024, new themes and continued questions. Curr Opin Cardiol 2024; 39:315-322. [PMID: 38661130 DOI: 10.1097/hco.0000000000001151] [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] [Indexed: 04/26/2024]
Abstract
PURPOSE OF REVIEW While pediatric myocarditis incidence has increased since the coronavirus disease 2019 (COVID-19) pandemic, there remain questions regarding diagnosis, risk stratification, and optimal therapy. This review highlights recent publications and continued unanswered questions related to myocarditis in children. RECENT FINDINGS Emergence from the COVID-19 era has allowed more accurate description of the incidence and prognosis of myocarditis adjacent to COVID-19 infection and vaccine administration as well that of multi-system inflammatory disease in children (MIS-C). As cardiac magnetic resonance technology has shown increased availability and evidence in pediatric myocarditis, it is important to understand conclusions from adult imaging studies and define the use of this imaging biomarker in children. Precision medicine has begun to allow real-time molecular evaluations to help diagnose and risk-stratify cardiovascular diseases, with emerging evidence of these modalities in myocarditis. SUMMARY Recent information regarding COVID-19 associated myocarditis, cardiac magnetic resonance, and molecular biomarkers may help clinicians caring for children with myocarditis and identify needs for future investigations.
Collapse
|
6
|
Mann DL. The Emerging Field of Cardioimmunology: Past, Present and Foreseeable Future. Circ Res 2024; 134:1663-1680. [PMID: 38843286 PMCID: PMC11160976 DOI: 10.1161/circresaha.123.323656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/08/2024] [Indexed: 06/09/2024]
Abstract
Over the past 30 years, the field of cardioimmunology has moved from being dismissed as a field that was chasing an epiphenomenon of little biological consequence to a scientific discipline that is providing important new insights into the immunologic basis for hypertension, atherosclerosis, myocarditis, pericarditis, autoimmune heart disease, and heart failure. In this article, we will review the conceptual insights and technical breakthroughs that have allowed the field to move forward, as well as the clinical trials in the cardioimmunology space, to provide a historical context for the articles that will appear in the compendium that is focused on the interface between cardioimmunology, myocardial function, and disease.
Collapse
Affiliation(s)
- Douglas L Mann
- Cardiovascular Division, Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
7
|
Golino M, Harding D, Del Buono MG, Fanti S, Mohiddin S, Toldo S, Smyth J, Sanna T, Marelli-Berg F, Abbate A. Innate and adaptive immunity in acute myocarditis. Int J Cardiol 2024; 404:131901. [PMID: 38403204 PMCID: PMC11450758 DOI: 10.1016/j.ijcard.2024.131901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/12/2024] [Accepted: 02/21/2024] [Indexed: 02/27/2024]
Abstract
Acute myocarditis is an acute inflammatory cardiomyopathy associated with cardiac damage triggered by a virus or a pathological immune activation. It may present with a wide range of clinical presentations, ranging from mild symptoms to severe forms like fulminant myocarditis, characterized by hemodynamic compromise and cardiogenic shock. The immune system plays a central role in the pathogenesis of myocarditis. In fact, while its function is primarily protective, aberrant responses can be detrimental. In this context, both innate and adaptive immunity play pivotal roles; notably, the innate system offers a non-specific and immediate defense, while the adaptive provides specialized protection with immunological memory. However, dysregulation in these systems can misidentify cardiac tissue, triggering autoimmune reactions and possibly leading to significant cardiac tissue damage. This review highlights the importance of innate and adaptive immune responses in the progression and treatment of acute myocarditis.
Collapse
Affiliation(s)
- Michele Golino
- Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States of America; Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Daniel Harding
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Marco Giuseppe Del Buono
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Silvia Fanti
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Saidi Mohiddin
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom; Barts Heart Centre, London, United Kingdom
| | - Stefano Toldo
- Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States of America
| | - James Smyth
- Fralin Biomedical Research Institute at Virginia Tech Carillion, Roanoke, VA, United States of America; Virginia Tech Carilion School of Medicine, Roanoke, VA, United States of America; Department of Biological Sciences, College of Science, Virginia Tech, Blacksburg, VA, United States of America
| | - Tommaso Sanna
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Federica Marelli-Berg
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom.
| | - Antonio Abbate
- Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States of America.
| |
Collapse
|
8
|
Jiang J, Shu H, Wang DW, Hui R, Li C, Ran X, Wang H, Zhang J, Nie S, Cui G, Xiang D, Shao Q, Xu S, Zhou N, Li Y, Gao W, Chen Y, Bian Y, Wang G, Xia L, Wang Y, Zhao C, Zhang Z, Zhao Y, Wang J, Chen S, Jiang H, Chen J, Du X, Chen M, Sun Y, Li S, Ding H, Ma X, Zeng H, Lin L, Zhou S, Ma L, Tao L, Chen J, Zhou Y, Guo X. Chinese Society of Cardiology guidelines on the diagnosis and treatment of adult fulminant myocarditis. SCIENCE CHINA. LIFE SCIENCES 2024; 67:913-939. [PMID: 38332216 DOI: 10.1007/s11427-023-2421-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/25/2023] [Indexed: 02/10/2024]
Abstract
Fulminant myocarditis is an acute diffuse inflammatory disease of myocardium. It is characterized by acute onset, rapid progress and high risk of death. Its pathogenesis involves excessive immune activation of the innate immune system and formation of inflammatory storm. According to China's practical experience, the adoption of the "life support-based comprehensive treatment regimen" (with mechanical circulation support and immunomodulation therapy as the core) can significantly improve the survival rate and long-term prognosis. Special emphasis is placed on very early identification,very early diagnosis,very early prediction and very early treatment.
Collapse
Affiliation(s)
- Jiangang Jiang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hongyang Shu
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dao Wen Wang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Rutai Hui
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Chenze Li
- Zhongnan Hospital of Wuhan University, Wuhan, 430062, China
| | - Xiao Ran
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hong Wang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jing Zhang
- Fuwai Huazhong Cardiovascular Hospital, Zhengzhou, 450003, China
| | - Shaoping Nie
- Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Guanglin Cui
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dingcheng Xiang
- Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, 510010, China
| | - Qun Shao
- Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Shengyong Xu
- Union Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ning Zhou
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuming Li
- Taida Hospital, Tianjin, 300457, China
| | - Wei Gao
- Peking University Third Hospital, Beijing, 100191, China
| | - Yuguo Chen
- Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Yuan Bian
- Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Guoping Wang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Liming Xia
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yan Wang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chunxia Zhao
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhiren Zhang
- Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Yuhua Zhao
- Kanghua Hospital, Dongguan, Guangzhou, 523080, China
| | - Jianan Wang
- Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Shaoliang Chen
- Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Hong Jiang
- Renmin Hospital of Wuhan University, Wuhan, 430060, Wuhan, China
| | - Jing Chen
- Renmin Hospital of Wuhan University, Wuhan, 430060, Wuhan, China
| | - Xianjin Du
- Renmin Hospital of Wuhan University, Wuhan, 430060, Wuhan, China
| | - Mao Chen
- West China Hospital, Sichuan University, Chengdu, 610044, China
| | - Yinxian Sun
- First Hospital of China Medical University, Shenyang, 110002, China
| | - Sheng Li
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hu Ding
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xueping Ma
- General Hospital of Ningxia Medical University, Yinchuan, 750003, China
| | - Hesong Zeng
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li Lin
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shenghua Zhou
- The Second Xiangya Hospital, Central South University, Changsha, 410012, China
| | - Likun Ma
- The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230002, China
| | - Ling Tao
- The First Affiliated Hospital of Air Force Medical University, Xi'an, 710032, China
| | - Juan Chen
- Central Hospital of Wuhan City, Wuhan, 430014, China
| | - Yiwu Zhou
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaomei Guo
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
9
|
Weisleder H, Jacobson E, Frishman WH, Dhand A. Cardiac Manifestations of Viral Infections, Including COVID-19: A Review. Cardiol Rev 2024; 32:124-130. [PMID: 36730913 DOI: 10.1097/crd.0000000000000481] [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] [Indexed: 02/04/2023]
Abstract
Viral infections have been linked to a variety of cardiac pathology, which may include acute myocarditis, dilated cardiomyopathy, heart failure, cardiogenic shock, pericarditis, acute coronary syndromes, and arrhythmias. We performed a systematic review of literature focusing on the cardiovascular effects of various viral infections, as well as providing an update on the current understanding of the pathophysiology of Coronavirus disease-2019 (COVID-19). Cardiac manifestations of viral illnesses are usually self-limiting, have variable clinical presentations, and require sufficient clinical suspicion for diagnosis and optimal management.
Collapse
Affiliation(s)
| | | | | | - Abhay Dhand
- From the New York Medical College, Valhalla, NY
- Department of Medicine and Surgery, Westchester Medical Center, Valhalla, NY
| |
Collapse
|
10
|
Markousis-Mavrogenis G, Baumhove L, Al-Mubarak AA, Aboumsallem JP, Bomer N, Voors AA, van der Meer P. Immunomodulation and immunopharmacology in heart failure. Nat Rev Cardiol 2024; 21:119-149. [PMID: 37709934 DOI: 10.1038/s41569-023-00919-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/03/2023] [Indexed: 09/16/2023]
Abstract
The immune system is intimately involved in the pathophysiology of heart failure. However, it is currently underused as a therapeutic target in the clinical setting. Moreover, the development of novel immunomodulatory therapies and their investigation for the treatment of patients with heart failure are hampered by the fact that currently used, evidence-based treatments for heart failure exert multiple immunomodulatory effects. In this Review, we discuss current knowledge on how evidence-based treatments for heart failure affect the immune system in addition to their primary mechanism of action, both to inform practising physicians about these pleiotropic actions and to create a framework for the development and application of future immunomodulatory therapies. We also delineate which subpopulations of patients with heart failure might benefit from immunomodulatory treatments. Furthermore, we summarize completed and ongoing clinical trials that assess immunomodulatory treatments in heart failure and present several therapeutic targets that could be investigated in the future. Lastly, we provide future directions to leverage the immunomodulatory potential of existing treatments and to foster the investigation of novel immunomodulatory therapeutics.
Collapse
Affiliation(s)
- George Markousis-Mavrogenis
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Lukas Baumhove
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Ali A Al-Mubarak
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Joseph Pierre Aboumsallem
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Cardiology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Nils Bomer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Adriaan A Voors
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
| |
Collapse
|
11
|
Papamichail A, Kourek C, Briasoulis A, Xanthopoulos A, Tsougos E, Farmakis D, Paraskevaidis I. Targeting Key Inflammatory Mechanisms Underlying Heart Failure: A Comprehensive Review. Int J Mol Sci 2023; 25:510. [PMID: 38203681 PMCID: PMC10778956 DOI: 10.3390/ijms25010510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 12/27/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Inflammation is a major component of heart failure (HF), causing peripheral vasculopathy and cardiac remodeling. High levels of circulating inflammatory cytokines in HF patients have been well recognized. The hallmark of the inflammatory imbalance is the insufficient production of anti-inflammatory mediators, a condition that leads to dysregulated cytokine activity. The condition progresses because of the pathogenic consequences of the cytokine imbalance, including the impact of endothelial dysfunction and adrenergic responsiveness deterioration, and unfavorable inotropic effects on the myocardium. Hence, to develop possible anti-inflammatory treatment options that will enhance the outcomes of HF patients, it is essential to identify the potential pathophysiological mechanisms of inflammation in HF. Inflammatory mediators, such as cytokines, adhesion molecules, and acute-phase proteins, are elevated during this process, highlighting the complex association between inflammation and HF. Therefore, these inflammatory markers can be used in predicting prognosis of the syndrome. Various immune cells impact on myocardial remodeling and recovery. They lead to stimulation, release of alarmins and risk-related molecule patterns. Targeting key inflammatory mechanisms seems a quite promising therapy strategy in HF. Cytokine modulation is only one of several possible targets in the fight against inflammation, as the potential molecular targets for therapy in HF include immune activation, inflammation, oxidative stress, alterations in mitochondrial bioenergetics, and autophagy.
Collapse
Affiliation(s)
- Adamantia Papamichail
- Medical School of Athens, National and Kapodistrian University of Athens, 15772 Athens, Greece; (A.P.); (C.K.); (A.B.)
| | - Christos Kourek
- Medical School of Athens, National and Kapodistrian University of Athens, 15772 Athens, Greece; (A.P.); (C.K.); (A.B.)
| | - Alexandros Briasoulis
- Medical School of Athens, National and Kapodistrian University of Athens, 15772 Athens, Greece; (A.P.); (C.K.); (A.B.)
| | - Andrew Xanthopoulos
- Department of Cardiology, University Hospital of Larissa, 41110 Larissa, Greece;
| | - Elias Tsougos
- Department of Cardiology, Hygeia Hospital, 15123 Athens, Greece;
| | - Dimitrios Farmakis
- Attikon University Hospital, Medical School of Athens, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Ioannis Paraskevaidis
- Medical School of Athens, National and Kapodistrian University of Athens, 15772 Athens, Greece; (A.P.); (C.K.); (A.B.)
| |
Collapse
|
12
|
Uccello G, Bonacchi G, Rossi VA, Montrasio G, Beltrami M. Myocarditis and Chronic Inflammatory Cardiomyopathy, from Acute Inflammation to Chronic Inflammatory Damage: An Update on Pathophysiology and Diagnosis. J Clin Med 2023; 13:150. [PMID: 38202158 PMCID: PMC10780032 DOI: 10.3390/jcm13010150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Acute myocarditis covers a wide spectrum of clinical presentations, from uncomplicated myocarditis to severe forms complicated by hemodynamic instability and ventricular arrhythmias; however, all these forms are characterized by acute myocardial inflammation. The term "chronic inflammatory cardiomyopathy" describes a persistent/chronic inflammatory condition with a clinical phenotype of dilated and/or hypokinetic cardiomyopathy associated with symptoms of heart failure and increased risk for arrhythmias. A continuum can be identified between these two conditions. The importance of early diagnosis has grown markedly in the contemporary era with various diagnostic tools available. While cardiac magnetic resonance (CMR) is valid for diagnosis and follow-up, endomyocardial biopsy (EMB) should be considered as a first-line diagnostic modality in all unexplained acute cardiomyopathies complicated by hemodynamic instability and ventricular arrhythmias, considering the local expertise. Genetic counseling should be recommended in those cases where a genotype-phenotype association is suspected, as this has significant implications for patients' and their family members' prognoses. Recognition of the pathophysiological pathway and clinical "red flags" and an early diagnosis may help us understand mechanisms of progression, tailor long-term preventive and therapeutic strategies for this complex disease, and ultimately improve clinical outcomes.
Collapse
Affiliation(s)
- Giuseppe Uccello
- Division of Cardiology, Alessandro Manzoni Hospital—ASST Lecco, 23900 Lecco, Italy;
| | - Giacomo Bonacchi
- Division of Cardiology, Tor Vergata University Hospital, 00133 Rome, Italy;
| | | | - Giulia Montrasio
- Inherited Cardiovascular Diseases Unit, Barts Heart Centre, St. Bartholomew’s Hospital, London EC1A 7BS, UK;
| | - Matteo Beltrami
- Cardiomyopathy Unit, Careggi University Hospital, 50134 Florence, Italy
- Arrhythmia and Electrophysiology Unit, Careggi University Hospital, 50134 Florence, Italy
| |
Collapse
|
13
|
Tian X, Zhou G, Li H, Zhang X, Zhao L, Zhang K, Wang L, Liu M, Liu C, Yang P. RBM25 binds to and regulates alternative splicing levels of Slc38a9, Csf1, and Coro6 to affect immune and inflammatory processes in H9c2 cells. PeerJ 2023; 11:e16312. [PMID: 37953772 PMCID: PMC10637245 DOI: 10.7717/peerj.16312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 09/27/2023] [Indexed: 11/14/2023] Open
Abstract
Background Alternative splicing (AS) is a biological process that allows genes to be translated into diverse proteins. However, aberrant AS can predispose cells to aberrations in biological mechanisms. RNA binding proteins (RBPs), closely affiliated with AS, have gained increased attention in recent years. Among these RBPs, RBM25 has been reported to participate in the cardiac pathological mechanism through regulating AS; however, the involvement of RBM25 as a splicing factor in heart failure remains unclarified. Methods RBM25 was overexpressed in H9c2 cells to explore the target genes bound and regulated by RBM25 during heart failure. RNA sequencing (RNA-seq) was used to scrutinize the comprehensive transcriptional level before identifying AS events influenced by RBM25. Further, improved RNA immunoprecipitation sequencing (iRIP-seq) was employed to pinpoint RBM25-binding sites, and RT-qPCR was used to validate specific genes modulated by RBM25. Results RBM25 was found to upregulate the expression of genes pertinent to the inflammatory response and viral processes, as well as to mediate the AS of genes associated with cellular apoptosis and inflammation. Overlap analysis between RNA-seq and iRIP-seq suggested that RBM25 bound to and manipulated the AS of genes associated with inflammation in H9c2 cells. Moreover, qRT-PCR confirmed Slc38a9, Csf1, and Coro6 as the binding and AS regulatory targets of RBM25. Conclusion Our research implies that RBM25 plays a contributory role in cardiac inflammatory responses via its ability to bind to and regulate the AS of related genes. This study offers preliminary evidence of the influence of RBM25 on inflammation in H9c2 cells.
Collapse
Affiliation(s)
- Xin Tian
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Guangli Zhou
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hao Li
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xueting Zhang
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lingmin Zhao
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Keyi Zhang
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Luqiao Wang
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Mingwei Liu
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Chen Liu
- Department of Radiology, Affiliated Hospital of Yunnan University, Kunming, China
| | - Ping Yang
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
14
|
Luo Z, Cheng J, Wang Y. m6A regulator-mediated RNA methylation modification remodels immune microenvironment in dilated cardiomyopathy. J Cell Physiol 2023; 238:2282-2292. [PMID: 37475583 DOI: 10.1002/jcp.31085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 06/25/2023] [Accepted: 07/07/2023] [Indexed: 07/22/2023]
Abstract
The latest evidence suggested that the onset of dilated cardiomyopathy (DCM) is closely associated with immune microenvironment disturbance. Since N6 -methyladenosine (m6A) RNA methylation impacts on immunocyte function and antitumor immunity, it is predictable that m6A RNA methylation may result in immune microenvironment disorder. Here, we attempted to verify this hypothesis. We used single-sample gene set enrichment analysis (ssGSEA) to investigate the infiltration abundance of immunocytes, single-cell RNA-Seq to identify key m6A regulator, and a doxorubicin (Dox)-induced DCM mouse model to confirm our findings. ssGSEA revealed a higher infiltration abundance of CD8+ T lymphocytes, NK cells, monocytes, and B+ lymphocytes in DCM myocardium tissue. Single-cell RNA-Seq indicated a critical role of IGFBP2 in DCM. Cross-checking analysis hinted an interaction between IGFBP2 and NSUN5, ALYREF, RRP8, and ALKBH3. Mechanically, IGFBP2-mediated RNA methylation deteriorated the immune microenvironment and thus increased the risk of DCM by enhancing CD8+ T lymphocyte, NK cell, monocyte, B+ lymphocyte infiltration and activating check-point, MHC-I, and T cell co-stimulation signaling pathways. In the DCM mouse model, echocardiography indicated a significant reduction in ejection fraction (EF) and fractional shortening (FS) and an increase in left ventricular internal dimensions at systole (LVIDs) and diastole (LVIDd). MASSON staining indicated an increased fibrosis in myocardium tissue. qPCR and immunofluorescence staining indicated a significant increase in mRNA and protein levels of IGFBP2. The present study indicated that IGFBP2-mediated RNA methylation remodeled the immune microenvironment and increased the risk of DCM. IGFBP2 may serve as potential therapeutic target for DCM.
Collapse
Affiliation(s)
- Zhi Luo
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Jun Cheng
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yanggan Wang
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Medical Research Institute of Wuhan University, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| |
Collapse
|
15
|
Wang E, Zhou R, Li T, Hua Y, Zhou K, Li Y, Luo S, An Q. The Molecular Role of Immune Cells in Dilated Cardiomyopathy. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1246. [PMID: 37512058 PMCID: PMC10385992 DOI: 10.3390/medicina59071246] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023]
Abstract
Dilated cardiomyopathy (DCM) is a rare and severe condition characterized by chamber dilation and impaired contraction of the left ventricle. It constitutes a fundamental etiology for profound heart failure and abrupt cardiac demise, rendering it a prominent clinical indication for heart transplantation (HTx) among both adult and pediatric populations. DCM arises from various etiologies, including genetic variants, epigenetic disorders, infectious insults, autoimmune diseases, and cardiac conduction abnormalities. The maintenance of cardiac function involves two distinct types of immune cells: resident immune cells and recruited immune cells. Resident immune cells play a crucial role in establishing a harmonious microenvironment within the cardiac tissue. Nevertheless, in response to injury, cardiomyocytes initiate a cytokine cascade that attracts peripheral immune cells, thus perturbing this intricate equilibrium and actively participating in the initiation and pathological remodeling of dilated cardiomyopathy (DCM), particularly during the progression of myocardial fibrosis. Additionally, immune cells assume a pivotal role in orchestrating the inflammatory processes, which are intimately linked to the prognosis of DCM. Consequently, understanding the molecular role of various immune cells and their regulation mechanisms would provide an emerging era for managing DCM. In this review, we provide a summary of the most recent advancements in our understanding of the molecular mechanisms of immune cells in DCM. Additionally, we evaluate the effectiveness and limitations of immunotherapy approaches for the treatment of DCM, with the aim of optimizing future immunotherapeutic strategies for this condition.
Collapse
Affiliation(s)
- Enping Wang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Ruofan Zhou
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Tiange Li
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Yimin Hua
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Kaiyu Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Yifei Li
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shuhua Luo
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Qi An
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
16
|
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
|
17
|
Brociek E, Tymińska A, Giordani AS, Caforio ALP, Wojnicz R, Grabowski M, Ozierański K. Myocarditis: Etiology, Pathogenesis, and Their Implications in Clinical Practice. BIOLOGY 2023; 12:874. [PMID: 37372158 PMCID: PMC10295542 DOI: 10.3390/biology12060874] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/29/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023]
Abstract
Myocarditis is an inflammatory disease of the myocardium caused by infectious or non-infectious agents. It can lead to serious short-term and long-term sequalae, such as sudden cardiac death or dilated cardiomyopathy. Due to its heterogenous clinical presentation and disease course, challenging diagnosis and limited evidence for prognostic stratification, myocarditis poses a great challenge to clinicians. As it stands, the pathogenesis and etiology of myocarditis is only partially understood. Moreover, the impact of certain clinical features on risk assessment, patient outcomes and treatment options is not entirely clear. Such data, however, are essential in order to personalize patient care and implement novel therapeutic strategies. In this review, we discuss the possible etiologies of myocarditis, outline the key processes governing its pathogenesis and summarize best available evidence regarding patient outcomes and state-of-the-art therapeutic approaches.
Collapse
Affiliation(s)
- Emil Brociek
- First Department of Cardiology, Medical University of Warsaw, 02-097 Warsaw, Poland; (E.B.); (M.G.); (K.O.)
| | - Agata Tymińska
- First Department of Cardiology, Medical University of Warsaw, 02-097 Warsaw, Poland; (E.B.); (M.G.); (K.O.)
| | - Andrea Silvio Giordani
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, 35-100 Padova, Italy; (A.S.G.); (A.L.P.C.)
| | - Alida Linda Patrizia Caforio
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, 35-100 Padova, Italy; (A.S.G.); (A.L.P.C.)
| | - Romuald Wojnicz
- Department of Histology and Cell Pathology in Zabrze, School of Medicine with the Division of Dentistry, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Marcin Grabowski
- First Department of Cardiology, Medical University of Warsaw, 02-097 Warsaw, Poland; (E.B.); (M.G.); (K.O.)
| | - Krzysztof Ozierański
- First Department of Cardiology, Medical University of Warsaw, 02-097 Warsaw, Poland; (E.B.); (M.G.); (K.O.)
| |
Collapse
|
18
|
Matsumori A. Myocarditis and Autoimmunity. Expert Rev Cardiovasc Ther 2023. [PMID: 37243585 DOI: 10.1080/14779072.2023.2219895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 05/20/2023] [Accepted: 05/26/2023] [Indexed: 05/29/2023]
Abstract
INTRODUCTION Autoimmune myocarditis may develop due to heterogeneous causes. Myocarditis is often caused by viral infections, but it can also be caused by systemic autoimmune diseases. Immune checkpoint inhibitors and virus vaccines induce immune activation, and they can cause the development of myocarditis, as well as several immune-related adverse events. The development of myocarditis is dependent on the genetic factors of the host, and the major histocompatibility complex (MHC) may be an important determinant of the type and severity of the disease. However, non-MHC immunoregulatory genes may also play a role in determining susceptibility. AREA COVERED This review summarizes the current knowledge of the etiology, pathogenesis, diagnosis and treatment of autoimmune myocarditis with a particular focus on viral infection and autoimmunity, and biomarkers of myocarditis. EXPERT OPINION An endomyocardial biopsy may not be the gold standard for the diagnosis of myocarditis. Cardiac magnetic resonance imaging is useful in diagnosing autoimmune myocarditis. Recently identified biomarkers of inflammation and myocyte injury are promising for the diagnosis of myocarditis when measured simultaneously. Future treatments should focus on the appropriate diagnosis of the etiologic agent, as well as on the specific stage of the evolution of immune and inflammatory processes.
Collapse
Affiliation(s)
- Akira Matsumori
- Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto 612-8555, Japan
| |
Collapse
|
19
|
Smith S, Ascione R. Targeting neuro-immune systems to achieve cardiac tissue repair following myocardial infarction: A review of therapeutic approaches from in-vivo preclinical to clinical studies. Pharmacol Ther 2023; 245:108397. [PMID: 36996910 PMCID: PMC7616359 DOI: 10.1016/j.pharmthera.2023.108397] [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: 11/25/2022] [Revised: 03/12/2023] [Accepted: 03/27/2023] [Indexed: 03/30/2023]
Abstract
Myocardial healing following myocardial infarction (MI) toward either functional tissue repair or excessive scarring/heart failure, may depend on a complex interplay between nervous and immune system responses, myocardial ischemia/reperfusion injury factors, as well as genetic and epidemiological factors. Hence, enhancing cardiac repair post MI may require a more patient-specific approach targeting this complex interplay and not just the heart, bearing in mind that the dysregulation or modulation of just one of these systems or some of their mechanisms may determine the outcome either toward functional repair or toward heart failure. In this review we have elected to focus on existing preclinical and clinical in-vivo studies aimed at testing novel therapeutic approaches targeting the nervous and immune systems to trigger myocardial healing toward functional tissue repair. To this end, we have only selected clinical and preclinical in-vivo studies reporting on novel treatments targeting neuro-immune systems to ultimately treat MI. Next, we have grouped and reported treatments under each neuro-immune system. Finally, for each treatment we have assessed and reported the results of each clinical/preclinical study and then discussed their results collectively. This structured approach has been followed for each treatment discussed. To keep this review focused, we have deliberately omitted to cover other important and related research areas such as myocardial ischemia/reperfusion injury, cell and gene therapies as well as any ex-vivo and in-vitro studies. The review indicates that some of the treatments targeting the neuro-immune/inflammatory systems appear to induce beneficial effects remotely on the healing heart post MI, warranting further validation. These remote effects on the heart also indicates the presence of an overarching synergic response occurring across the nervous and immune systems in response to acute MI, which appear to influence cardiac tissue repair in different ways depending on age and timing of treatment delivery following MI. The cumulative evidence arising from this review allows also to make informed considerations on safe as opposed to detrimental treatments, and within the safe treatments to ascertain those associated with conflicting or supporting preclinical data, and those warranting further validation.
Collapse
Affiliation(s)
- Sarah Smith
- Bristol Heart Institute and Translational Biomedical Research Centre, Faculty of Health Science, University of Bristol, Bristol, UK
| | - Raimondo Ascione
- Bristol Heart Institute and Translational Biomedical Research Centre, Faculty of Health Science, University of Bristol, Bristol, UK.
| |
Collapse
|
20
|
Mitochondria Dysfunction at the Heart of Viral Myocarditis: Mechanistic Insights and Therapeutic Implications. Viruses 2023; 15:v15020351. [PMID: 36851568 PMCID: PMC9963085 DOI: 10.3390/v15020351] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
The myocardium/heart is the most mitochondria-rich tissue in the human body with mitochondria comprising approximately 30% of total cardiomyocyte volume. As the resident "powerhouse" of cells, mitochondria help to fuel the high energy demands of a continuously beating myocardium. It is no surprise that mitochondrial dysfunction underscores the pathogenesis of many cardiovascular ailments, including those of viral origin such as virus-induced myocarditis. Enteroviruses have been especially linked to injuries of the myocardium and its sequelae dilated cardiomyopathy for which no effective therapies currently exist. Intriguingly, recent mechanistic insights have demonstrated viral infections to directly damage mitochondria, impair the mitochondrial quality control processes of the cell, such as disrupting mitochondrial antiviral innate immune signaling, and promoting mitochondrial-dependent pathological inflammation of the infected myocardium. In this review, we briefly highlight recent insights on the virus-mitochondria crosstalk and discuss the therapeutic implications of targeting mitochondria to preserve heart function and ultimately combat viral myocarditis.
Collapse
|
21
|
Harding D, Chong MHA, Lahoti N, Bigogno CM, Prema R, Mohiddin SA, Marelli-Berg F. Dilated cardiomyopathy and chronic cardiac inflammation: Pathogenesis, diagnosis and therapy. J Intern Med 2023; 293:23-47. [PMID: 36030368 DOI: 10.1111/joim.13556] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Dilated cardiomyopathy (DCM) is typically defined by left ventricular dilation and systolic dysfunction in the absence of a clear precipitant. Idiopathic disease is common; up to 50% of patients with DCM have no cause found despite imaging, genetic and biopsy assessments. Treatment remains focused on managing symptoms, reducing the risk of sudden cardiac death and ameliorating the structural and electrical complications of disease progression. In the absence of aetiology-specific treatments, the condition remains associated with a poor prognosis; mortality is approximately 40% at 10 years. The role of immune-mediated inflammatory injury in the development and progression of DCM was first proposed over 30 years ago. Despite the subsequent failures of three large clinical trials of immunosuppressive treatment (ATTACH, RENEWAL and the Myocarditis Treatment Trial), evidence for an abnormal adaptive immune response in DCM remains significant. In this review, we summarise and discuss available evidence supporting immune dysfunction in DCM, with a specific focus on cellular immunity. We also highlight current clinical and experimental treatments. We propose that the success of future immunosuppressive treatment trials in DCM will be dependent on the deep immunophenotyping of patients, to identify those with active inflammation and/or an abnormal immune response who are most likely to respond to therapy.
Collapse
Affiliation(s)
- Daniel Harding
- Centre for Biochemical Pharmacology, William Harvey Research Institute, London, UK
| | - Ming H A Chong
- Barts and The London School of Medicine and Dentistry, London, UK
| | - Nishant Lahoti
- Conquest Hospital, East Sussex Healthcare NHS Trust, St Leonards-on-Sea, UK
| | - Carola M Bigogno
- Barts and The London School of Medicine and Dentistry, London, UK
| | - Roshni Prema
- University Hospital, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK
| | | | | |
Collapse
|
22
|
Ikeda M, Ide T, Matsushima S, Ikeda S, Okabe K, Ishikita A, Tadokoro T, Sada M, Abe K, Sato M, Hanada A, Arai S, Ohtani K, Nonami A, Mizuno S, Morimoto S, Motohashi S, Akashi K, Taniguchi M, Tsutsui H. Immunomodulatory Cell Therapy Using αGalCer-Pulsed Dendritic Cells Ameliorates Heart Failure in a Murine Dilated Cardiomyopathy Model. Circ Heart Fail 2022; 15:e009366. [PMID: 36268712 PMCID: PMC9760469 DOI: 10.1161/circheartfailure.122.009366] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is a life-threatening disease, resulting in refractory heart failure. An immune disorder underlies the pathophysiology associated with heart failure progression. Invariant natural killer T (iNKT) cell activation is a prospective therapeutic strategy for ischemic heart disease. However, its efficacy in nonischemic cardiomyopathy, such as DCM, remains to be elucidated, and the feasible modality for iNKT cell activation in humans is yet to be validated. METHODS Dendritic cells isolated from human volunteers were pulsed with α-galactosylceramide ex vivo, which were used as α-galactosylceramide-pulsed dendritic cells (αGCDCs). We treated DCM mice harboring mutated troponin TΔK210/ΔK210 with αGCDCs and evaluated the efficacy of iNKT cell activation on heart failure in DCM mice. Furthermore, we investigated the molecular basis underlying its therapeutic effects in these mice and analyzed primary cardiac cells under iNKT cell-secreted cytokines. RESULTS The number of iNKT cells in the spleens of DCM mice was reduced compared with that in wild-type mice, whereas αGCDC treatment activated iNKT cells, prolonged survival of DCM mice, and prevented decline in the left ventricular ejection fraction for 4 weeks, accompanied by suppressed interstitial fibrosis. Mechanistically, αGCDC treatment suppressed TGF (transforming growth factor)-β signaling and expression of fibrotic genes and restored vasculature that was impaired in DCM hearts by upregulating angiopoietin 1 (Angpt1) expression. Consistently, IFNγ (interferon gamma) suppressed TGF-β-induced Smad2/3 signaling and the expression of fibrotic genes in cardiac fibroblasts and upregulated Angpt1 expression in cardiomyocytes via Stat1. CONCLUSIONS Immunomodulatory cell therapy with αGCDCs is a novel therapeutic strategy for heart failure in DCM.
Collapse
Affiliation(s)
- Masataka Ikeda
- Department of Cardiovascular Medicine (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Immunoregulatory Cardiovascular Medicine (M.I., T.I.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomomi Ide
- Department of Cardiovascular Medicine (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Immunoregulatory Cardiovascular Medicine (M.I., T.I.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shouji Matsushima
- Department of Cardiovascular Medicine (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Soichiro Ikeda
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kosuke Okabe
- Department of Cardiovascular Medicine (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akihito Ishikita
- Department of Cardiovascular Medicine (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomonori Tadokoro
- Department of Cardiovascular Medicine (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masashi Sada
- Department of Cardiovascular Medicine (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ko Abe
- Department of Cardiovascular Medicine (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Midori Sato
- Department of Cardiovascular Medicine (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akiko Hanada
- Department of Cardiovascular Medicine (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shinobu Arai
- Department of Early Childhood and Elementary Education, Faculty of Education, Nakamura Gakuen University, Fukuoka, Japan (S.A.)
| | - Kisho Ohtani
- Department of Cardiovascular Medicine (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Atsushi Nonami
- Center for Advanced Medical Innovation, Kyushu University Hospital, Fukuoka, Japan (A.N.)
| | - Shinichi Mizuno
- Department of Health Sciences (S. Mizuno), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Sachio Morimoto
- Department of Health Sciences at Fukuoka, International University of Health and Welfare, Japan (S. Morimoto)
| | - Shinichiro Motohashi
- Department of Cardiovascular Medicine (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Medical Immunology, Graduate School of Medicine, Chiba University, Japan (S. Motohashi)
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science (K. Akashi), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaru Taniguchi
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan (M.T.)
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
23
|
Elevated Serum Levels of IgG4 in Patients with Heart Failure with Reduced Ejection Fraction: A Prospective Controlled Study. BIOLOGY 2022; 11:biology11081168. [PMID: 36009795 PMCID: PMC9404706 DOI: 10.3390/biology11081168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 11/29/2022]
Abstract
(1) Background: Immunoglobulin gamma subclass 4 (IgG4) is a serum protein belonging to the immunoglobulin superfamily. It has a central role in certain immune-mediated conditions defined as IgG4-related disease. There is a paucity of data regarding the potential association of IgG4 and cardiovascular diseases. Our aim is to study the serum levels of IgG4 in patients with ischemic and non-ischemic dilated cardiomyopathy (DCM). (2) Methods: patients with ischemic and non-ischemic DCM were included in this study. Non-ischemic DCM was defined as a left ventricular ejection fraction (LVEF) < 40% without coronary artery disease (CAD). Ischemic DCM was defined as a LVEF < 40% and proven CAD. The serum concentrations of IgG4 were measured by turbidimetry. (3) Results: Overall 98 patients with cardiomyopathy had significantly higher levels of IgG4 compared with the control group (77.4 ± 64.0 vs. 50.3 ± 28.8 mg/dL, p < 0.01). Although there was no difference in the total IgG levels in patients with ischemic DCM, the serum concentrations of IgG4 were significantly higher than the corresponding values in the control group (89.8 ± 67.3 vs. 50.3 ± 28.8 mg/dL; interquartile ranges: 40.4−126.5 vs. 31.8−66.8 mg/dL, p < 0.01). This was altered by gender and smoking. (4) Conclusions: The patients with ischemic DCM had increased serum concentrations of IgG4. Future studies are warranted to explore the potential role of an IgG4-mediated process in patients with heart failure with reduced LVEF.
Collapse
|
24
|
Montera MW, Marcondes-Braga FG, Simões MV, Moura LAZ, Fernandes F, Mangine S, Oliveira Júnior ACD, Souza ALADAGD, Ianni BM, Rochitte CE, Mesquita CT, de Azevedo Filho CF, Freitas DCDA, Melo DTPD, Bocchi EA, Horowitz ESK, Mesquita ET, Oliveira GH, Villacorta H, Rossi Neto JM, Barbosa JMB, Figueiredo Neto JAD, Luiz LF, Hajjar LA, Beck-da-Silva L, Campos LADA, Danzmann LC, Bittencourt MI, Garcia MI, Avila MS, Clausell NO, Oliveira NAD, Silvestre OM, Souza OFD, Mourilhe-Rocha R, Kalil Filho R, Al-Kindi SG, Rassi S, Alves SMM, Ferreira SMA, Rizk SI, Mattos TAC, Barzilai V, Martins WDA, Schultheiss HP. Brazilian Society of Cardiology Guideline on Myocarditis - 2022. Arq Bras Cardiol 2022; 119:143-211. [PMID: 35830116 PMCID: PMC9352123 DOI: 10.36660/abc.20220412] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
| | - Fabiana G Marcondes-Braga
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | - Marcus Vinícius Simões
- Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo, São Paulo, SP - Brasil
| | | | - Fabio Fernandes
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | - Sandrigo Mangine
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | | | | | - Bárbara Maria Ianni
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | - Carlos Eduardo Rochitte
- Instituto do Coração (InCor) - Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil
- Hospital do Coração (HCOR), São Paulo, SP - Brasil
| | - Claudio Tinoco Mesquita
- Hospital Pró-Cardíaco, Rio de Janeiro, RJ - Brasil
- Universidade Federal Fluminense,Rio de Janeiro, RJ - Brasil
- Hospital Vitória, Rio de Janeiro, RJ - Brasil
| | | | | | | | - Edimar Alcides Bocchi
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | | | - Evandro Tinoco Mesquita
- Universidade Federal Fluminense,Rio de Janeiro, RJ - Brasil
- Centro de Ensino e Treinamento Edson de Godoy Bueno / UHG, Rio de Janeiro, RJ - Brasil
| | | | | | | | | | | | | | - Ludhmila Abrahão Hajjar
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
- Instituto do Câncer do Estado de São Paulo da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil
| | - Luis Beck-da-Silva
- Hospital de Clínicas de Porto Alegre, Porto Alegre, RS - Brasil
- Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS - Brasil
| | | | | | - Marcelo Imbroise Bittencourt
- Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ - Brasil
- Hospital Universitário Pedro Ernesto, Rio de Janeiro, RJ - Brasil
| | - Marcelo Iorio Garcia
- Hospital Universitário Clementino Fraga Filho (HUCFF) da Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ - Brasil
| | - Monica Samuel Avila
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | | | | | | | | | | | | | - Sadeer G Al-Kindi
- Harrington Heart and Vascular Institute, University Hospitals and Case Western Reserve University,Cleveland, Ohio - EUA
| | | | - Silvia Marinho Martins Alves
- Pronto Socorro Cardiológico de Pernambuco (PROCAPE), Recife, PE - Brasil
- Universidade de Pernambuco (UPE), Recife, PE - Brasil
| | - Silvia Moreira Ayub Ferreira
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | - Stéphanie Itala Rizk
- Instituto do Câncer do Estado de São Paulo da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil
- Hospital Sírio Libanês, São Paulo, SP - Brasil
| | | | - Vitor Barzilai
- Instituto de Cardiologia do Distrito Federal, Brasília, DF - Brasil
| | - Wolney de Andrade Martins
- Universidade Federal Fluminense,Rio de Janeiro, RJ - Brasil
- DASA Complexo Hospitalar de Niterói, Niterói, RJ - Brasil
| | | |
Collapse
|
25
|
Sozzi FB, Gherbesi E, Faggiano A, Gnan E, Maruccio A, Schiavone M, Iacuzio L, Carugo S. Viral Myocarditis: Classification, Diagnosis, and Clinical Implications. Front Cardiovasc Med 2022; 9:908663. [PMID: 35795363 PMCID: PMC9250986 DOI: 10.3389/fcvm.2022.908663] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Myocarditis is an inflammatory disease of the myocardium with focal or diffuse involvement. Viral infections are the most common cause of myocarditis, especially in Western countries. A recent viral illness with gastroenteric or upper respiratory symptoms often precedes myocarditis. The absence of specific pathognomonic features in conjunction with the wide spectrum of clinical manifestations that range from subclinical cases to sudden cardiac death (SCD) makes myocarditis diagnosis particularly challenging. Moreover, myocarditis might represent a cause of initially unexplained dilated cardiomyopathy (DCM) and heart failure (HF), especially among children and young adults. Cardiac magnetic resonance imaging (CMR) is crucial for myocarditis diagnosis, because of its ability to detect interstitial edema during acute inflammation. Assessment of subepicardial or mid-myocardial fibrosis by late gadolinium enhancement (LGE) is typical for myocarditis. Cardiac arrhythmias are frequent events that may arise especially in more severe myocarditis cases. The most common form of arrhythmia is atrial fibrillation, followed by ventricular tachycardia. Documented arrhythmias have been reported more commonly with HIV myocarditis than other more common infections such as Adenovirus, Parvovirus B19, human Herpes virus 6, and Enterovirus. The mechanisms of arrhythmogenesis in myocardial inflammation are not fully understood; in the acute phase, the spectrum of arrhythmogenesis ranges from a direct effect on cardiomyocytes that leads to electrical instability and ion channel impairment to ischemia from coronary macro- or microvascular disease. In chronic myocarditis, instead, myocardial replacement with fibrosis promotes scar-mediated re-entrant ventricular arrhythmias. Observational data suggested the important role of CMR, with LGE being the strongest independent predictor of SCD, cardiac, and all-cause mortality. In acute myocarditis, the most common localization of subepicardial LGE dwells in the lateral wall. Patients with myocarditis that develop HF and arrhythmias usually show a larger LGE distribution involving several myocardial segments. Moreover, a mid-layer LGE in the interventricular septum is more frequent in acute myocarditis than in acute coronary syndromes cases. The risk of SCD in patients with wide areas of LGE is significant, and a shared decision-making approach is warranted. Nevertheless, there is no formal consensus about the extension of LGE to justify implantable cardioverter defibrillator (ICD) implantation in primary prevention.
Collapse
Affiliation(s)
- Fabiola B. Sozzi
- Cardiology Unit, Internal Medicine Department, Fondazione Ospedale Maggiore Policlinico IRCCS Cà Granda, University of Milan, Milan, Italy
| | - Elisa Gherbesi
- Cardiology Unit, Internal Medicine Department, Fondazione Ospedale Maggiore Policlinico IRCCS Cà Granda, University of Milan, Milan, Italy
| | - Andrea Faggiano
- Cardiology Unit, Internal Medicine Department, Fondazione Ospedale Maggiore Policlinico IRCCS Cà Granda, University of Milan, Milan, Italy
| | - Eleonora Gnan
- Cardiology Unit, Internal Medicine Department, Fondazione Ospedale Maggiore Policlinico IRCCS Cà Granda, University of Milan, Milan, Italy
| | - Alessio Maruccio
- Cardiology Unit, Internal Medicine Department, Fondazione Ospedale Maggiore Policlinico IRCCS Cà Granda, University of Milan, Milan, Italy
| | - Marco Schiavone
- Cardiology Unit, Luigi Sacco University Hospital, Milan, Italy
| | | | - Stefano Carugo
- Cardiology Unit, Internal Medicine Department, Fondazione Ospedale Maggiore Policlinico IRCCS Cà Granda, University of Milan, Milan, Italy
| |
Collapse
|
26
|
Ho CL, Ju TR, Lee CC, Lin HT, Wang AL, Chen RJ, Lin YC. The Early Dynamic Change in Cardiac Enzymes and Renal Function Is Associated with Mortality in Patients with Fulminant Myocarditis on Extracorporeal Membrane Oxygenation: Analysis of a Single Center's Experience. Healthcare (Basel) 2022; 10:1063. [PMID: 35742115 PMCID: PMC9222735 DOI: 10.3390/healthcare10061063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/04/2022] [Accepted: 06/06/2022] [Indexed: 11/23/2022] Open
Abstract
(1) Background: Fulminant myocarditis (FM) could result in hemodynamic derangement and fatal arrhythmia. Veno-arterial extracorporeal membrane oxygenation (V-A ECMO) is used to maintain organ perfusion in FM patients complicating cardiogenic shock. The present study aims to assess the static and dynamic factors in association with mortality in FM patients on V-A ECMO (2) Methods: Twenty-eight patients were enrolled between 2013 to 2019 for analysis (3) Results: In-hospital survival rate was 78.5%. There was no statistical difference in demographics and baseline laboratory data between survivors and non-survivors. However, within 24 h after ECMO support, CK-MB increased by 96.8% among non-survivors, but decreased by 23.7% among survivors (p = 0.022). Troponin I increased by 378% among non-survivors and 1.7% among survivors (p = 0.032). Serum creatinine increased by 108% among non-survivors, but decreased by 8.5% among survivors (p = 0.005). The receiver operating characteristic curve suggested an increase in serum creatinine by 68% within 24 h after ECMO support was associated with increased mortality with an area under the curve of 0.91. (4) Conclusions: V-A ECMO is an excellent tool to support FM patients with cardiogenic shock. The early dynamic change of renal function and cardiac enzymes may be useful for outcome assessment.
Collapse
Affiliation(s)
- Ching-Lin Ho
- Department of Surgery, Division of Cardiovascular Surgery, China Medical University Hospital, Taichung City 406040, Taiwan;
| | - Teressa Reanne Ju
- Department of Internal Medicine, New York Presbyterian Queens, New York, NY 11355, USA;
| | - Chi Chan Lee
- Department of Critical Care, Guam Regional Medical City, Tamuning, GU 96929, USA;
| | - Hsin-Ti Lin
- College of Medicine, China Medical University, Taichung City 406040, Taiwan; (H.-T.L.); (A.-L.W.)
| | - Alexander-Lee Wang
- College of Medicine, China Medical University, Taichung City 406040, Taiwan; (H.-T.L.); (A.-L.W.)
| | - Robert Jeenchen Chen
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA;
| | - You-Cian Lin
- Department of Surgery, Division of Cardiovascular Surgery, China Medical University Hospital, Taichung City 406040, Taiwan;
| |
Collapse
|
27
|
Okor I, Bob-Manuel T, Price J, Sleem A, Amoran O, Kelly J, Ekerete MF, Bamgbose MO, Bolaji OA, Krim SR. COVID-19 Myocarditis: An Emerging Clinical Conundrum. Curr Probl Cardiol 2022; 47:101268. [PMID: 35644500 PMCID: PMC9135640 DOI: 10.1016/j.cpcardiol.2022.101268] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 05/23/2022] [Indexed: 01/08/2023]
Abstract
Coronavirus Disease 2019 (COVID-19) has been a significant cause of global mortality and morbidity since it was first reported in December 2019 in Wuhan, China. COVID19 like previous coronaviruses primarily affects the lungs causing pneumonia, interstitial pneumonitis, and severe acute respiratory distress syndrome (ARDS). However, there is increasing evidence linking COVID-19 to cardiovascular complications such as arrhythmias, heart failure, cardiogenic shock, fulminant myocarditis, and cardiac death. Given the novelty of this virus, there is paucity of data on some cardiovascular complications of COVID-19, specifically myocarditis. Myocarditis is an inflammatory disease of the heart muscle with a heterogenous clinical presentation and progression. It is mostly caused by viral infections and is the result of interaction of the virus and the host's immune system. There have been several case reports linking COVID-19 with myocarditis, however the true mechanism of cardiac injury remains under investigation. In this paper we review the clinical presentation, proposed pathophysiology, differential diagnoses and management of myocarditis in COVID-19 patients.
Collapse
Affiliation(s)
- Ivana Okor
- John Ochsner Heart and Vascular Center, New Orleans, LA, USA
| | | | - Justin Price
- John Ochsner Heart and Vascular Center, New Orleans, LA, USA
| | - Amber Sleem
- John Ochsner Heart and Vascular Center, New Orleans, LA, USA
| | - Olayiwola Amoran
- Pennsylvania Hospital of the University of Pennsylvania Health System, Philadelphia, PA, USA
| | - Jayna Kelly
- John Ochsner Heart and Vascular Center, New Orleans, LA, USA
| | | | | | - Olayiwola A Bolaji
- Department of Internal Medicine, University of Maryland Capital Regional Medical Centre, Lake Arbor, MD, USA
| | - Selim R Krim
- John Ochsner Heart and Vascular Center, New Orleans, LA, USA; The University of Queensland Ochsner Clinical School, Faculty of Medicine, The University of Queensland, New Orleans, LA, USA.
| |
Collapse
|
28
|
Oh JH, Kim GB, Seok H. Implication of microRNA as a potential biomarker of myocarditis. Clin Exp Pediatr 2022; 65:230-238. [PMID: 35240034 PMCID: PMC9082251 DOI: 10.3345/cep.2021.01802] [Citation(s) in RCA: 8] [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: 12/05/2021] [Accepted: 01/29/2022] [Indexed: 12/15/2022] Open
Abstract
Myocarditis was previously attributed to an epidemic viral infection. Additional harmful reagents, in addition to viruses, play a role in its etiology. Coronavirus disease 2019 (COVID-19) vaccine-induced myocarditis has recently been described, drawing attention to vaccine-induced myocarditis in children and adolescents. Its pathology is based on a series of complex immune responses, including initial innate immune responses in response to viral entry, adaptive immune responses leading to the development of antigen-specific antibodies, and autoimmune responses to cellular injury caused by cardiomyocyte rupture that releases antigens. Chronic inflammation and fibrosis in the myocardium eventually result in cardiac failure. Recent advancements in molecular biology have remarkably increased our understanding of myocarditis. In particular, microRNAs (miRNAs) are a hot topic in terms of the role of new biomarkers and the pathophysiology of myocarditis. Myocarditis has been linked with microRNA-221/222 (miR-221/222), miR-155, miR-10a*, and miR-590. Despite the lack of clinical trials of miRNA intervention in myocarditis yet, multiple clinical trials of miRNAs in other cardiac diseases have been aggressively conducted to help pave the way for future research, which is bolstered by the success of recently U.S. Food and Drug Administration-approved small-RNA medications. This review presents basic information and recent research that focuses on myocarditis and related miRNAs as a potential novel biomarker and the therapeutics.
Collapse
Affiliation(s)
- Jin-Hee Oh
- Department of Pediatrics, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Gi Beom Kim
- Department of Pediatrics, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Heeyoung Seok
- Department of Transdisciplinary Research and Collaboration, Genomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
29
|
Zhang X, Liu W. Engineering Injectable Anti‐Inflammatory Hydrogels to Treat Acute Myocardial Infarction. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Xiaoping Zhang
- Tianjin Key Laboratory of Composite and Functional Materials School of Material Science and Engineering Tianjin University Tianjin 300350 China
| | - Wenguang Liu
- Tianjin Key Laboratory of Composite and Functional Materials School of Material Science and Engineering Tianjin University Tianjin 300350 China
| |
Collapse
|
30
|
Kuć A, Kubik D, Kościelecka K, Szymanek W, Męcik-Kronenberg T. The Relationship Between Peripartum Cardiomyopathy and Preeclampsia – Pathogenesis, Diagnosis and Management. J Multidiscip Healthc 2022; 15:857-867. [PMID: 35496718 PMCID: PMC9045831 DOI: 10.2147/jmdh.s357872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/14/2022] [Indexed: 12/03/2022] Open
Abstract
Peripartum cardiomyopathy (PPCM) is a condition with an incompletely understood etiology, although many risk factors for this disorder have been mentioned. Preeclampsia (PE) is a rare but undoubtedly very important cause of PPCM. Early recognition and prompt treatment of preeclampsia and peripartum cardiomyopathy are essential to optimize pregnancy outcomes. An extensive manual search of major electronic databases was conducted in November 2021. The following literature review provides a comprehensive discussion of peripartum cardiomyopathy and preeclampsia and quantifies the prevalence of PE in women with PPCM. The authors highlighted aspects such as epidemiology, risk factors, cardiovascular changes, diagnosis and clinical presentation, and management and complications. Accumulating data indicate that both conditions have a similar pathogenesis characterized by vascular abnormalities. In both conditions we can observe an increase in interleukin-6 and gamma interferon, CCL2/MCP1, and decreased SOD activity. sFLT1 (a soluble form of fms-like tyrosine kinase 1), a substance with antiangiogenic and probably cardiotoxic effects, may be important. Preeclampsia and peripartum cardiomyopathy are characterized by recurrence rates that follow a similar pattern in subsequent pregnancies, and mortality remains a concern. Our analysis highlights the need to better understand the co-morbidity of PE and PPCM, and the need to qualify patients for the same clinical trials because of the common origin of these conditions.
Collapse
Affiliation(s)
- Aleksandra Kuć
- Student Research Group at the Chair and Department of Pathomorphology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Zabrze, Poland
- Correspondence: Aleksandra Kuć, 1E Street, Siedlce, 08-110, Poland, Tel +48 504 188 178, Email
| | - Daria Kubik
- Student Research Group at the Chair and Department of Pathomorphology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Zabrze, Poland
| | - Klaudia Kościelecka
- Student Research Group at the Chair and Department of Pathomorphology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Zabrze, Poland
| | - Wojciech Szymanek
- Hospital Emergency Department, St. John Paul II Mazovia Regional Hospital in Siedlce, Siedlce, Poland
- Nursing at Collegium Mazovia Innovative University in Siedlce, Siedlce, Poland
| | - Tomasz Męcik-Kronenberg
- Chair and Department of Pathomorphology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Zabrze, Poland
| |
Collapse
|
31
|
Seidman MA, McManus B. Myocarditis. Cardiovasc Pathol 2022. [DOI: 10.1016/b978-0-12-822224-9.00005-0] [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: 10/17/2022] Open
|
32
|
Wei X, Zhang A, Yang W, Fang Y. Depletion of β3-adrenergic receptor relieves pressure overload-induced cardiac hypertrophy and heart failure via enhancing innate immune response. Biomed Pharmacother 2021; 143:112194. [PMID: 34563949 DOI: 10.1016/j.biopha.2021.112194] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 11/16/2022] Open
Abstract
Cardiac pressure overload is a crucial risk factor for cardiac hypertrophy and heart failure. Our previous study showed that depletion of the β3-adrenergic receptor (ADRB3) induced left ventricular diastolic dysfunction via potential regulation of energy metabolism and cardiac contraction. However, the effects of ADRB3 on pressure overload-induced heart failure remain unclear. In the present study, systemic ADRB3-knockout mice suffering from transverse aortic constriction (TAC) surgery were used to identify the effects of ADRB3 on pressure overload-induced heart failure. Compared to wild-type mice, ADRB3 depletion significantly improved the left ventricular ejection fraction, reduced left ventricular posterior wall thickness and interventricular septum thickness, and decreased the area of cardiomyocytes after TAC. RNA sequencing and bioinformatics analysis showed that ADRB3 depletion up-regulated 275 mRNAs and down-regulated 105 mRNAs in mice suffering TAC surgery. GO analysis, GO-tree analysis, and GSEA showed that ADRB3 depletion mainly enhanced the innate immune response of hearts in cardiac pressure overload mice. In addition, pathway analysis and Pathway-Act analysis presented that innate immune response-related pathways, including RIG-I-like receptor signaling pathway, antigen processing and presentation, Toll-like receptor signaling pathway, and cell adhesion molecules, were significantly enriched in ADRB3-KO-TAC mice. Ten hub genes were identified using protein-protein interaction network, MCODE, and cytoHubba analysis. Furthermore, the depletion and activation of ADRB3 validated the effects of ADRB3 on the innate immune response of hearts after TAC. In conclusion, ADRB3 depletion relieves pressure overload-induced cardiac hypertrophy and heart failure, and these effects could be explained by the enhancement of innate immune response.
Collapse
Affiliation(s)
- Xuemei Wei
- Department of Cardiovascular medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Andi Zhang
- Department of Cardiovascular medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenbo Yang
- Department of Cardiovascular medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yuehua Fang
- Department of Cardiovascular medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
33
|
Skrzypiec-Spring M, Sapa-Wojciechowska A, Haczkiewicz-Leśniak K, Piasecki T, Kwiatkowska J, Podhorska-Okołów M, Szeląg A. HMG-CoA Reductase Inhibitor, Simvastatin Is Effective in Decreasing Degree of Myocarditis by Inhibiting Metalloproteinases Activation. Biomolecules 2021; 11:biom11101415. [PMID: 34680049 PMCID: PMC8533153 DOI: 10.3390/biom11101415] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/24/2021] [Accepted: 09/24/2021] [Indexed: 12/17/2022] Open
Abstract
Background: Acute myocarditis often progresses to heart failure because there is no effective, etiology-targeted therapy of this disease. Simvastatin has been shown to be cardioprotective by decreasing matrix metalloproteinases’ (MMPs) activity. The study was designed to determine whether simvastatin inhibits MMPs activity, decreases the severity of inflammation and contractile dysfunction of the heart in experimental autoimmune myocarditis (EAM). Methods: Simvastatin (3 or 30 mg/kg/day) was given to experimental rats with EAM by gastric gavage for 21 days. Then transthoracic echocardiography was performed, MMPs activity and troponin I level were determined and tissue samples were assessed under a light and transmission electron microscope. Results: Hearts treated with simvastatin did not show left ventricular enlargement. As a result of EAM, there was an enhanced activation of MMP-9, which was significantly reduced in the high-dose simvastatin group compared to the low-dose group. It was accompanied by prevention of myofilaments degradation and reduction of severity of inflammation. Conclusions: The cardioprotective effects of simvastatin in the acute phase of EAM are, at least in part, due to its ability to decrease MMP-9 activity and subsequent decline in myofilaments degradation and suppression of inflammation. These effects were achieved in doses equivalent to therapeutic doses in humans.
Collapse
Affiliation(s)
- Monika Skrzypiec-Spring
- Department of Pharmacology, Wroclaw Medical University, 50-345 Wrocław, Poland; (J.K.); (A.S.)
- Correspondence: ; Tel.: +48-71-7841438
| | | | | | - Tomasz Piasecki
- Department of Epizootiology and Clinic of Bird and Exotic Animals, Wroclaw University of Environmental and Life Sciences, 50-013 Wrocław, Poland;
| | - Joanna Kwiatkowska
- Department of Pharmacology, Wroclaw Medical University, 50-345 Wrocław, Poland; (J.K.); (A.S.)
| | - Marzenna Podhorska-Okołów
- Department of Ultrastructural Research, Wroclaw Medical University, 50-013 Wrocław, Poland; (K.H.-L.); (M.P.-O.)
| | - Adam Szeląg
- Department of Pharmacology, Wroclaw Medical University, 50-345 Wrocław, Poland; (J.K.); (A.S.)
| |
Collapse
|
34
|
Zhong Z, Yang Z, Peng Y, Wang L, Yuan X. Diagnosis and treatment of eosinophilic myocarditis. J Transl Autoimmun 2021; 4:100118. [PMID: 35005589 PMCID: PMC8716607 DOI: 10.1016/j.jtauto.2021.100118] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 08/25/2021] [Accepted: 08/30/2021] [Indexed: 12/05/2022] Open
Abstract
Eosinophilic myocarditis is a type of inflammatory cardiomyopathy characterized by eosinophilic infiltration into myocardial tissue. The accurate myocarditis incidence rate is difficult to determine because of the clinical limitations of an endomyocardial biopsy. The primary pathogenesis of eosinophilic myocarditis is the release of related substances by eosinophils, leading to cell membrane damage and cell destruction. However, evidence suggests that specific genes play a role in myocarditis development.As CMR imaging availability increases, the diagnosis rate of eosinophilic myocarditis will increase. The diagnosis of myocarditis mainly depends on an endocardial biopsy. Glucocorticoids can relieve patients' symptoms, but the early use of steroids may prevent intermediate disease stage development (i.e., thrombonecrosis and fibrosis with wall thrombosis). Anticoagulant therapy may also affect disease development. In addition to routine follow-up, a regular myocardial biopsy should be considered for discharged patients, if possible.
Collapse
Key Words
- ANCA, anti-neutrophil cytoplasmic antibody
- CEL, chronic eosinophilic leukemia.
- CMR, cardiac magnetic resonance
- Cardiac magnetic resonance
- EAM, experimental autoimmune myocarditis
- ECG, electrocardiogram
- ECP, eosinophilic cationic protein
- EGE, early gadolinium enhancement, LGE, late gadolinium enhancement
- EGPA, eosinophilic granulomatosis with polyangiitis
- EMB, endomyocardial biopsy
- Endomyocardial biopsy
- Eosinophilic myocarditis
- FIP1L1-PDGFRA, FIP1-like1-platelet-derived growth factor receptor α
- Glucocorticoids
- HES, hypereosinophilic syndrome
- IFNγ, interferon gamma
Collapse
Affiliation(s)
- Zezhong Zhong
- Department of Cardiology, Liuyang People's Hospital, Nanhua University, Hunan Province, 410300, China
| | - Zicong Yang
- People's Hospital of Guangxi Zhuang Autonomous Region, 530021, China
| | - Yiming Peng
- Department of Cardiology, Liuyang People's Hospital, Nanhua University, Hunan Province, 410300, China
| | - Lei Wang
- Department of Cardiology, Liuyang People's Hospital, Nanhua University, Hunan Province, 410300, China
| | - Xuming Yuan
- Department of Cardiology, Liuyang People's Hospital, Nanhua University, Hunan Province, 410300, China
| |
Collapse
|
35
|
Li B, Lento PA, Pan S. Inflammatory Cardiomyopathy: Case-based Review on Clinical Presentation, Diagnosis, and Management. Cardiol Rev 2021; 29:230-237. [PMID: 33165090 DOI: 10.1097/crd.0000000000000369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Inflammatory cardiomyopathy is a broad term encompassing any disease leading to myocardial inflammation with associated cardiac dysfunction. While endomyocardial biopsy remains the gold standard for diagnosis, noninvasive imaging techniques, such as cardiac magnetic resonance imaging and positron emission tomography, have become powerful tools to facilitate the identification of underlying myocardial inflammation. This review presents a series of clinical cases with some common etiologies of inflammatory cardiomyopathy, including diagnosis and management.
Collapse
Affiliation(s)
- Boyangzi Li
- From the Department of Medicine, New York University Grossman School of Medicine, New York, NY
| | - Patrick A Lento
- Department of Pathology, New York Medical College, Valhalla, NY
| | - Stephen Pan
- Department of Medicine, Division of Cardiology, Westchester Medical Center/New York Medical College, Valhalla, NY
| |
Collapse
|
36
|
Soma G, Simmons MA, Canarie MF, Karnik R, Steele J, Tiyyagura G, Asnes J. A 13-Year-Old With Fever and Chest Pain. Pediatrics 2021; 148:peds.2021-050960. [PMID: 34344802 DOI: 10.1542/peds.2021-050960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/15/2021] [Indexed: 11/24/2022] Open
Affiliation(s)
| | | | - Michael F Canarie
- Critical Care, Department of Pediatrics, School of Medicine, Yale University, New Haven, Connecticut
| | | | | | | | | |
Collapse
|
37
|
Abstract
Importance Peripartum cardiomyopathy is a rare form of heart failure due to left ventricular systolic dysfunction that affects women late in pregnancy and the postpartum period. A diagnosis of exclusion, peripartum cardiomyopathy can be difficult to diagnose in the context of the normal physiologic changes of pregnancy and requires a high index of suspicion. Evidence Acquisition Original research articles, review articles, and guidelines on peripartum cardiomyopathy were reviewed. Results The etiology of peripartum cardiomyopathy remains poorly defined, but theories include genetic predisposition, as well as myocardial inflammation and angiogenic dysregulation. Risk factors for this condition include hypertensive disorders of pregnancy, Black race, and maternal age older than 30 years. Patients with peripartum cardiomyopathy are at increased risk of acute clinical decompensation, cardiac arrhythmias, thromboembolic complications, and death. Primary treatment modalities include initiation of a medication regimen aimed at the optimization of preload and reduction of afterload. Maternal clinical status is the primary determinant for timing of delivery. Conclusions Prompt diagnosis and medical management by an interdisciplinary care team are vital for improving outcomes in patients with peripartum cardiomyopathy.
Collapse
|
38
|
Rikhi R, Karnuta J, Hussain M, Collier P, Funchain P, Tang WHW, Chan TA, Moudgil R. Immune Checkpoint Inhibitors Mediated Lymphocytic and Giant Cell Myocarditis: Uncovering Etiological Mechanisms. Front Cardiovasc Med 2021; 8:721333. [PMID: 34434981 PMCID: PMC8381278 DOI: 10.3389/fcvm.2021.721333] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 07/14/2021] [Indexed: 12/26/2022] Open
Abstract
The advent of immune checkpoint inhibitors (ICIs) has revolutionized the field of oncology, but these are associated with immune related adverse events. One such adverse event, is myocarditis, which has limited the continued immunosuppressive treatment options in patients afflicted by the disease. Pre-clinical and clinical data have found that specific ICI targets and precipitate distinct myocardial infiltrates, consistent with lymphocytic or giant cell myocarditis. Specifically, it has been reported that CTLA-4 inhibition preferentially results in giant cell myocarditis with a predominately CD4+ T cell infiltrate and PD-1 inhibition leads to lymphocytic myocarditis, with a predominately CD8+ T cell infiltrate. Our manuscript discusses the latest literature surrounding ICI pathways and targets, while detailing proposed mechanisms behind ICI mediated myocarditis.
Collapse
Affiliation(s)
- Rishi Rikhi
- Department of Medicine, Cleveland Clinic Foundation, Cleveland, OH, United States.,Department of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Jaret Karnuta
- Department of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Muzna Hussain
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Patrick Collier
- Department of Medicine, Case Western Reserve University, Cleveland, OH, United States.,Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Pauline Funchain
- Department of Medicine, Case Western Reserve University, Cleveland, OH, United States.,Department of Hematology and Medical Oncology, Taussig Cancer Center Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Wai Hong Wilson Tang
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Timothy A Chan
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH, United States
| | - Rohit Moudgil
- Department of Medicine, Case Western Reserve University, Cleveland, OH, United States.,Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| |
Collapse
|
39
|
Haley KE, Almas T, Shoar S, Shaikh S, Azhar M, Cheema FH, Hameed A. The role of anti-inflammatory drugs and nanoparticle-based drug delivery models in the management of ischemia-induced heart failure. Biomed Pharmacother 2021; 142:112014. [PMID: 34391184 DOI: 10.1016/j.biopha.2021.112014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/27/2021] [Accepted: 08/03/2021] [Indexed: 12/20/2022] Open
Abstract
Ongoing advancements in the treatment of acute myocardial infarction (MI) have significantly decreased MI related mortality. Consequently, the number of patients experiencing post-MI heart failure (HF) has continued to rise. Infarction size and the extent of left ventricular (LV) remodeling are largely determined by the extent of ischemia at the time of myocardial injury. In the setting of MI or acute phase of post-MI LV remodeling, anti-inflammatory drugs including intravenous immunoglobulin (IVIG) and Pentoxifylline have shown potential efficacy in preventing post-MI remodeling in-vitro and in some clinical trials. However, systemic administration of anti-inflammatory drugs are not without their off-target side effects. Herein, we explore the clinical feasibility of targeted myocardial delivery of anti-inflammatory drugs via biodegradable polymers, liposomes, hydrogels, and nano-particle based drug delivery models (NDDM) based on existing pre-clinical and clinical models. We summarize the barriers to clinical application of targeted anti-inflammatory delivery post-MI, including challenges in achieving sufficient retention and distribution, as well as the potential need for multiple dosing. Collectively, we suggest that localized delivery of anti-inflammatory agents to the myocardium using NDDM is a promising approach for successful treatment of ischemic HF. Future studies will be instrumental in determining the most effective target and delivery modalities for orchestrating NDDM-mediated treatment of HF.
Collapse
Affiliation(s)
- Kathryn E Haley
- Graduate Entry Medicine, RCSI University of Medicine and Health Sciences, Dublin 2 Dublin, Ireland; Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin 2 Dublin, Ireland
| | - Talal Almas
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin 2 Dublin, Ireland; School of Medicine, RCSI University of Medicine and Health Sciences, Dublin 2 Dublin, Ireland
| | - Saeed Shoar
- HCA Healthcare Gulf Coast Division, Houston, TX, USA
| | - Shan Shaikh
- HCA Healthcare Gulf Coast Division, Houston, TX, USA
| | - Maimoona Azhar
- Graduate Entry Medicine, RCSI University of Medicine and Health Sciences, Dublin 2 Dublin, Ireland; Department of Surgery, St. Vincent's University Hospital, Dublin 4 Dublin, Ireland
| | - Faisal Habib Cheema
- HCA Healthcare Gulf Coast Division, Houston, TX, USA; University of Houston, College of Medicine, Houston, TX, USA
| | - Aamir Hameed
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin 2 Dublin, Ireland; Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin (TCD), Dublin, Ireland.
| |
Collapse
|
40
|
Bian RT, Wang ZT, Li WY. Immunoadsorption treatment for dilated cardiomyopathy: A PRISMA-compliant systematic review and meta-analysis. Medicine (Baltimore) 2021; 100:e26475. [PMID: 34190171 PMCID: PMC8257917 DOI: 10.1097/md.0000000000026475] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 06/07/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND As one of the leading causes of heart failure, dilated cardiomyopathy (DCM) is characterized by dysfunctional muscle contraction and enlarged ventricular chamber. Patients with DCM have been shown to respond well to immunoadsorption (IA) therapies. However, the efficacy and safety of IA treatment for DCM patients remained to be evaluated. METHODS This study was designed in accordance with the Preferred Reporting Items for Systematic Review and Meta-analysis. We searched the databases such as Cochrane library, Cochrane Central Register of Controlled Trials, Embase, OVID, and Web of Science from January 1990 to March 20, 2020, and performed meta-analysis using Stata MP Version 13.0. RESULTS We performed meta-analysis on 12 studies that included a total of 395 patients with DCM. Overall, IA treatment significantly improved the left ventricular ejection fraction (6.01, 95% confidence interval [CI] [4.84-7.19]), reduced the left ventricular end diastolic diameter (-3.62, 95% CI [-4.06 to -3.19]), reduced severity of symptoms according to the New York Heart Association (NYHA) functional classification (-1.37, 95% CI [-1.73 to -1.02]) as compared with the controls, but had no effect on values for safety parameters (1.13, 95% CI [0.58-2.19]). CONCLUSIONS Results of this meta-analysis indicated that the IA treatment can improve the left ventricular ejection fraction, reduce left ventricular end diastolic diameter, and thus improve clinical outcome in DCM patients. However, further evidence are required to validate the relative safety of IA treatment. Multi-center, double blind studies should be conducted to elucidate the precise effect of IA treatment in DCM patients.
Collapse
Affiliation(s)
- Ru-tao Bian
- Department of Cardiology, Henan University of Chinese Medicine
| | - Zhen-tao Wang
- Department of Cardiology, The Second Affiliated Hospital of Henan University of Chinese Medicine
| | - Wei-yu Li
- Department of Nephropathy, Zhengzhou Traditional Chinese Medicine Hospital, Zhengzhou, China
| |
Collapse
|
41
|
Diagnosis and Management of Myocarditis: An Evidence-Based Review for the Emergency Medicine Clinician. J Emerg Med 2021; 61:222-233. [PMID: 34108120 DOI: 10.1016/j.jemermed.2021.03.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/17/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Myocarditis is a potentially fatal condition that can be misdiagnosed in the emergency department (ED) setting. OBJECTIVE The purpose of this narrative review article is to provide a summary of the background, pathophysiology, diagnosis, and management of myocarditis, with a focus on emergency clinicians. DISCUSSION Myocarditis occurs when inflammation of the heart musculature causes cardiac dysfunction. Symptoms may range from mild to severe and are often preceded by a viral prodrome. Laboratory assessment and an electrocardiogram can be helpful for the diagnosis, but echocardiography is the ideal test in the ED setting. Some patients may also require advanced imaging, though this will often occur during hospitalization or follow-up. Treatment is primarily focused on respiratory and hemodynamic support. Initial hemodynamic management includes vasopressors and inotropes, whereas more severe cases may require an intra-aortic balloon pump, extracorporeal membrane oxygenation, or a ventricular assist device. Nonsteroidal anti-inflammatory drugs should be avoided while intravenous immunoglobulin is controversial. CONCLUSION Myocarditis is a serious condition with the potential for significant morbidity and mortality. It is important for clinicians to be aware of the current evidence regarding the diagnosis, management, and disposition of these patients.
Collapse
|
42
|
Palaskas NL, Segura A, Lelenwa L, Siddiqui BA, Subudhi SK, Lopez-Mattei J, Durand JB, Deswal A, Zhao B, Maximilian Buja L, Iliescu C. Immune checkpoint inhibitor myocarditis: elucidating the spectrum of disease through endomyocardial biopsy. Eur J Heart Fail 2021; 23:1725-1735. [PMID: 34114291 DOI: 10.1002/ejhf.2265] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 05/17/2021] [Accepted: 06/04/2021] [Indexed: 12/11/2022] Open
Abstract
AIMS Although immune checkpoint inhibitor (ICI) myocarditis carries a high reported mortality, increasing reports of smoldering myocarditis suggest a clinical spectrum of disease. Endomyocardial biopsy (EMB) remains the gold standard for diagnosis of ICI myocarditis, but different pathologic diagnostic criteria exist. The objective of this study was to classify the spectrum of ICI myocarditis and myocardial inflammation by pathology findings on EMB and correlate this with clinical outcomes. METHODS AND RESULTS All patients who had EMB at MD Anderson Cancer Center between January 2018 and August 2019 for suspected ICI myocarditis were retrospectively reviewed. A grading system (Grades 0-2) based on the degree of inflammatory infiltrate was developed by pathologists. Cardiovascular outcomes and treatment were compared between grades of pathology. We identified 28 patients who had EMB for suspected ICI myocarditis, of which 18 were positive for myocarditis/inflammation. There were four deaths (two in Grade 2 and two in Grade 1), but only one was attributable to myocarditis. Grade 2 patients had no myocarditis-associated deaths despite having the highest troponin T values (median 2063 pg/mL). Four patients with Grade 1 myocardial inflammation continued ICI without any immunomodulation, and all were alive without adverse cardiovascular events at follow-up. CONCLUSION We defined an EMB grading system for ICI myocarditis encompassing a spectrum of histologic findings of inflammatory infiltrates. A subset of low-grade myocardial inflammation patients were able to continue ICI without immunosuppressive therapy. Further studies are needed to identify low-risk patients who can be safely treated with ICI.
Collapse
Affiliation(s)
- Nicolas L Palaskas
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Laura Lelenwa
- University of Texas Health Science Center, Houston, TX, USA
| | - Bilal A Siddiqui
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sumit K Subudhi
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Juan Lopez-Mattei
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Thoracic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jean B Durand
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anita Deswal
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bihong Zhao
- University of Texas Health Science Center, Houston, TX, USA
| | | | - Cezar Iliescu
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
43
|
Rroku A, Kottwitz J, Heidecker B. Update on myocarditis - what we know so far and where we may be heading. EUROPEAN HEART JOURNAL. ACUTE CARDIOVASCULAR CARE 2021; 10:455–467. [PMID: 32319308 DOI: 10.1177/2048872620910109] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Affiliation(s)
- Andi Rroku
- Charite Universitätsmedizin Berlin, Campus Benjamin Franklin, Germany
| | | | - Bettina Heidecker
- Charite Universitätsmedizin Berlin, Campus Benjamin Franklin, Germany
| |
Collapse
|
44
|
Meridor K, Shoenfeld Y, Tayer-Shifman O, Levy Y. Lupus acute cardiomyopathy is highly responsive to intravenous immunoglobulin treatment: Case series and literature review. Medicine (Baltimore) 2021; 100:e25591. [PMID: 33950936 PMCID: PMC8104142 DOI: 10.1097/md.0000000000025591] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/31/2021] [Indexed: 01/04/2023] Open
Abstract
INTRODUCTION Intravenous immunoglobulin (IVIg) is currently used with considerable success for the treatment of many autoimmune diseases, including systemic lupus erythematosus (SLE). Among its various indications, IVIg has also been found to be beneficial in myocarditis, whether or not it is associated with an autoimmune disease. Nevertheless, data regarding IVIg treatment for myocarditis/cardiomyopathy in patients with SLE are sparse. The objective of this case series was to describe our experience with IVIg as a treatment for lupus myocarditis and to review the literature for IVIg for this indication. PATIENT CONCERNS We report 5 female patients with SLE, who presented with signs of acute heart failure including pulmonary congestion and arrhythmias. DIAGNOSIS Echocardiography demonstrated new reduced left ventricular ejection fraction of 20% to 30%. Two patients underwent coronary artery angiography, which demonstrated normal coronary arteries, supporting the diagnosis of myocarditis or nonischemic cardiomyopathy. INTERVENTIONS High-dose IVIg treatment was initiated in all 5 patients. OUTCOMES Following the treatment, clinical and echocardiographic improvement in cardiac function occurred within a few days to 1 month. This dramatic improvement persisted for several years. CONCLUSION Based on our case series, we believe that IVIg has an important role in the management of lupus acute cardiomyopathy. This safe, well-tolerated optional treatment should be considered, especially in severe cases.
Collapse
Affiliation(s)
- Katya Meridor
- Department of Internal Medicine E, Meir Medical Center, Kfar Saba
- Affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel HaShomer
- Affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Oshrat Tayer-Shifman
- Rheumatology Unit, Meir Medical Center, Kfar Saba
- Affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yair Levy
- Department of Internal Medicine E, Meir Medical Center, Kfar Saba
- Affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
45
|
Kadhi A, Mohammed F, Nemer G. The Genetic Pathways Underlying Immunotherapy in Dilated Cardiomyopathy. Front Cardiovasc Med 2021; 8:613295. [PMID: 33937353 PMCID: PMC8079649 DOI: 10.3389/fcvm.2021.613295] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 03/22/2021] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) is a global public health threat affecting 26 million individuals worldwide with an estimated prevalence increase of 46% by 2030. One of the main causes of HF and sudden death in children and adult is Dilated Cardiomyopathy (DCM). DCM is characterized by dilation and systolic dysfunction of one or both ventricles. It has an underlying genetic basis or can develop subsequent to various etiologies that cause myocardium inflammation (secondary causes). The morbidity and mortality rates of DCM remains high despite recent advancement to manage the disease. New insights have been dedicated to better understand the pathogenesis of DCM in respect to genetic and inflammatory basis by linking the two entities together. This cognizance in the field of cardiology might have an innovative approach to manage DCM through targeted treatment directed to the causative etiology. The following review summarizes the genetical and inflammatory causes underlying DCM and the pathways of the novel precision-medicine-based immunomodulatory strategies to salvage and prevent the associated heart failure linked to the disease.
Collapse
Affiliation(s)
- Ayat Kadhi
- Division of Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Fathima Mohammed
- Division of Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Georges Nemer
- Division of Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.,Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
46
|
Khawaja A, Bromage DI. The innate immune response in myocarditis. Int J Biochem Cell Biol 2021; 134:105973. [PMID: 33831592 DOI: 10.1016/j.biocel.2021.105973] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 03/12/2021] [Accepted: 03/31/2021] [Indexed: 12/14/2022]
Abstract
Acute myocarditis is an inflammatory condition of the heart characterised by cellular injury and the influx of leucocytes, including neutrophils, monocytes, macrophages and lymphocytes. While this response is vital for tissue repair, excessive scar deposition and maladaptive ventricular remodelling can result in a legacy of heart failure. It is increasingly recognised as a clinical phenomenon due, in part, to increased availability of cardiac magnetic resonance imaging in patients presenting with chest pain in the absence of significant coronary artery disease. Emerging epidemiological evidence has associated myocarditis with poor outcomes in the context of left ventricular impairment, and even when the left ventricle is preserved outcomes are less benign than once thought. Despite this, our understanding of the contribution of the inflammatory response to the pathophysiology of acute myocarditis lags behind that of acute myocardial infarction, which is the vanguard cardiovascular condition for inflammation research. We recently reviewed monocyte and macrophage phenotype and function in acute myocardial infarction, concluding that their plasticity and heterogeneity might account for conflicting evidence from attempts to target specific leucocyte subpopulations. Here, we revise our understanding of myocardial inflammation, which is predominantly derived from myocardial infarction research, review experimental evidence for the immune response in acute myocarditis, focusing on innate immunity, and discuss potential future directions for immunotherapy research in acute myocarditis.
Collapse
Affiliation(s)
- Abdullah Khawaja
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Daniel I Bromage
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK.
| |
Collapse
|
47
|
Ota S, Orii M, Nishiguchi T, Yokoyama M, Matsushita R, Takemoto K, Tanimoto T, Hirata K, Hozumi T, Akasaka T. Implications of multiple late gadolinium enhancement lesions on the frequency of left ventricular reverse remodeling and prognosis in patients with non-ischemic cardiomyopathy. J Cardiovasc Magn Reson 2021; 23:32. [PMID: 33761955 PMCID: PMC7992777 DOI: 10.1186/s12968-021-00734-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 02/09/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Non-ischemic cardiomyopathy (NICM) is a heterogeneous disease, and its prognosis varies. Although late gadolinium enhancement (LGE)-cardiovascular magnetic resonance (CMR) demonstrates a linear pattern in the mid-wall of the septum or multiple LGE lesions in patients with NICM, the therapeutic response and prognosis of multiple LGE lesions have not been elucidated. This study aimed to investigate the frequency of left ventricular (LV) reverse remodeling (LVRR) and prognosis in patients with NICM who have multiple LGE lesions. METHODS This single-center retrospective study included 101 consecutive patients with NICM who were divided into 3 groups according to LGE-CMR results: patients without LGE (no LGE group = 48 patients), patients with a typical mid-wall LGE pattern (n = 29 patients), and patients with multiple LGE lesions (n = 24 patients). LVRR was defined as an increase in LV ejection fraction (LVEF) ≥ 10 % and a final value of LVEF > 35 %, which was accompanied by a decrease in LV end-systolic volume ≥ 15 % at 12-month follow-up using echocardiography. The frequency of composite cardiac events, defined as sudden cardiac death (SCD), aborted SCD (non-fatal ventricular fibrillation, sustained ventricular tachycardia, or adequate implantable cardioverter-defibrillator therapies), and heart failure death or hospitalization for worsening heart failure, were summarized and compared between the groups. RESULTS Among the 3 groups, the frequency of LVRR was significantly lower in the multiple lesions group than in the no LGE and mid-wall groups (no LGE vs. mid-wall vs. multiple lesions: 49 % vs. 52 % vs. 19 %, p = 0.03). There were 24 composite cardiac events among the patients: 2 in patients without LGE (hospitalization for worsening heart failure; 2), 7 in patients of the mid-wall group (SCD; 1, aborted SCD; 1 and hospitalization for worsening heart failure; 5), and 15 in patients of the multiple lesions group (SCD; 1, aborted SCD; 8 and hospitalization for worsening heart failure; 6). The multiple LGE lesions was an independent predictor of composite cardiac events (hazard ratio: 11.40 [95 % confidence intervals: 1.49-92.01], p = 0.020). CONCLUSIONS Patients with multiple LGE lesions have a higher risk of cardiac events and poorer LVRR. The LGE pattern may be useful for an improved risk stratification in patients with NICM.
Collapse
Affiliation(s)
- Shingo Ota
- Department of Cardiovascular Medicine, Wakayama Medical University, Wakayama, Japan
| | - Makoto Orii
- Department of Radiology, Iwate Medical University, 1-1, Idaidori, Yahaba, 028-3695, Iwate, Japan.
| | - Tsuyoshi Nishiguchi
- Department of Cardiovascular Medicine, Wakayama Medical University, Wakayama, Japan
| | - Mao Yokoyama
- Department of Cardiovascular Medicine, Wakayama Medical University, Wakayama, Japan
| | - Ryoko Matsushita
- Clinical Laboratory, Wakayama Medical University, Wakayama, Japan
| | - Kazushi Takemoto
- Department of Cardiovascular Medicine, Wakayama Medical University, Wakayama, Japan
| | - Takashi Tanimoto
- Department of Cardiovascular Medicine, Wakayama Medical University, Wakayama, Japan
| | - Kumiko Hirata
- Department of Education, Division of medical science, Osaka Educational University, Osaka, Japan
| | - Takeshi Hozumi
- Department of Cardiovascular Medicine, Wakayama Medical University, Wakayama, Japan
| | - Takashi Akasaka
- Department of Cardiovascular Medicine, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
48
|
Lyle MA, Cooper LT. Failure of intravenous immunoglobulin to improve cardiac function in parvovirus B19-associated chronic dilated cardiomyopathy. Eur J Heart Fail 2021; 23:310-311. [PMID: 33496358 DOI: 10.1002/ejhf.2110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 02/06/2023] Open
Affiliation(s)
- Melissa A Lyle
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Leslie T Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
49
|
Kessler EL, Oerlemans MIFJ, van den Hoogen P, Yap C, Sluijter JPG, de Jager SCA. Immunomodulation in Heart Failure with Preserved Ejection Fraction: Current State and Future Perspectives. J Cardiovasc Transl Res 2021; 14:63-74. [PMID: 32444946 PMCID: PMC7892675 DOI: 10.1007/s12265-020-10026-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 05/07/2020] [Indexed: 12/14/2022]
Abstract
The heart failure (HF) epidemic is growing and approximately half of the HF patients have heart failure with preserved ejection fraction (HFpEF). HFpEF is a heterogeneous syndrome, characterized by a preserved left ventricular ejection fraction (LVEF ≥ 50%) with diastolic dysfunction, and is associated with high morbidity and mortality. Underlying comorbidities of HFpEF, i.e., hypertension, type 2 diabetes mellitus, obesity, and renal failure, lead to a systemic pro-inflammatory state, thereby affecting normal cardiac function. Increased inflammatory biomarkers predict incident HFpEF and are higher in patients with HFpEF as compared with heart failure with reduced ejection fraction (HFrEF). Randomized trials in HFpEF patients using traditional HF medication failed to demonstrate a clear benefit on hard endpoints (mortality and/or HF hospitalization). Therefore, therapies targeting underlying comorbidities and systemic inflammation in early HFpEF may provide better opportunities. Here, we provide an overview of the current state and future perspectives of immunomodulatory therapies for HFpEF.
Collapse
Affiliation(s)
- Elise L Kessler
- Laboratory of Experimental Cardiology, Cardiology, UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, Netherlands
- Netherlands Heart Institute, 3511 EP, Utrecht, Netherlands
- Circulatory Health Laboratory, Utrecht University, University Medical Center Utrecht, Utrecht, Netherlands
| | - Martinus I F J Oerlemans
- Circulatory Health Laboratory, Utrecht University, University Medical Center Utrecht, Utrecht, Netherlands
- Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Patricia van den Hoogen
- Laboratory of Experimental Cardiology, Cardiology, UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, Netherlands
- Circulatory Health Laboratory, Utrecht University, University Medical Center Utrecht, Utrecht, Netherlands
| | - Carmen Yap
- Laboratory of Experimental Cardiology, Cardiology, UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, Netherlands
- Circulatory Health Laboratory, Utrecht University, University Medical Center Utrecht, Utrecht, Netherlands
| | - Joost P G Sluijter
- Laboratory of Experimental Cardiology, Cardiology, UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, Netherlands
- Circulatory Health Laboratory, Utrecht University, University Medical Center Utrecht, Utrecht, Netherlands
| | - Saskia C A de Jager
- Laboratory of Experimental Cardiology, Cardiology, UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, Netherlands.
- Circulatory Health Laboratory, Utrecht University, University Medical Center Utrecht, Utrecht, Netherlands.
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands.
| |
Collapse
|
50
|
Hazebroek MR, Henkens MTHM, Raafs AG, Verdonschot JAJ, Merken JJ, Dennert RM, Eurlings C, Abdul Hamid MA, Wolffs PFG, Winkens B, Brunner-La Rocca HP, Knackstedt C, van Paassen P, van Empel VPM, Heymans SRB. Intravenous immunoglobulin therapy in adult patients with idiopathic chronic cardiomyopathy and cardiac parvovirus B19 persistence: a prospective, double-blind, randomized, placebo-controlled clinical trial. Eur J Heart Fail 2021; 23:302-309. [PMID: 33347677 PMCID: PMC8048650 DOI: 10.1002/ejhf.2082] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/16/2020] [Accepted: 12/17/2020] [Indexed: 12/19/2022] Open
Abstract
AIMS Previous uncontrolled studies suggested a possible benefit of intravenous immunoglobulin (IVIg) in parvovirus B19 (B19V)-related dilated cardiomyopathy (DCM). This randomized, double-blind, placebo-controlled, single-centre trial investigated the benefits of IVIg beyond conventional therapy in idiopathic chronic DCM patients with B19V persistence. METHODS AND RESULTS Fifty patients (39 men; mean age 54 ± 11 years) with idiopathic chronic (>6 months) DCM on optimal medical therapy, left ventricular ejection fraction (LVEF) <45%, and endomyocardial biopsy (EMB) B19V load of >200 copies/µg DNA were blindly randomized to either IVIg (n = 26, 2 g/kg over 4 days) or placebo (n = 24). The primary outcome was change in LVEF at 6 months after randomization. Secondary outcomes were change in functional capacity assessed by 6-min walk test (6MWT), quality of life [Minnesota Living with Heart Failure Questionnaire (MLHFQ)], left ventricular end-diastolic volume (LVEDV), and EMB B19V load at 6 months after randomization. LVEF significantly improved in both IVIg and placebo groups (absolute mean increase 5 ± 9%, P = 0.011 and 6 ± 10%, P = 0.008, respectively), without a significant difference between groups (P = 0.609). Additionally, change in 6MWT [median (interquartile range) IVIg 36 (13;82) vs. placebo 32 (5;80) m; P = 0.573], MLHFQ [IVIg 0 (-7;5) vs. placebo -2 (-6;6), P = 0.904] and LVEDV (IVIg -16 ± 49 mL/m2 vs. placebo -29 ± 40 mL/m2 ; P = 0.334) did not significantly differ between groups. Moreover, despite increased circulating B19V antibodies upon IVIg administration, reduction in cardiac B19V did not significantly differ between groups. CONCLUSION Intravenous immunoglobulin therapy does not significantly improve cardiac systolic function or functional capacity beyond standard medical therapy in patients with idiopathic chronic DCM and cardiac B19V persistence. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov ID NCT00892112.
Collapse
Affiliation(s)
- Mark R Hazebroek
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - Michiel T H M Henkens
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - Anne G Raafs
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - Job A J Verdonschot
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands.,Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Jort J Merken
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - Robert M Dennert
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - Casper Eurlings
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - Myrurgia A Abdul Hamid
- Department of Pathology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Petra F G Wolffs
- Department of Medical Microbiology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Bjorn Winkens
- Department of Methodology and Statistics, Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, The Netherlands
| | - Hans-Peter Brunner-La Rocca
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - Christian Knackstedt
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - Pieter van Paassen
- Department of Nephrology and Clinical Immunology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Vanessa P M van Empel
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - Stephane R B Heymans
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands.,Department of Cardiovascular Research, University of Leuven, Leuven, Belgium.,Netherlands Heart Institute (Nl-HI), Utrecht, The Netherlands
| |
Collapse
|