1
|
Alberti M, Marcucci A, Biondi F, Chiusolo S, Masini G, Faggioni L, Cioni D, Morrone D, De Caterina R, Neri E, Aquaro GD. Pancreatitis-associated Myocarditis: Systematic Review and Meta-analysis of a Deadly Duo. J Cardiovasc Echogr 2024; 34:160-169. [PMID: 39895893 PMCID: PMC11784726 DOI: 10.4103/jcecho.jcecho_59_24] [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: 09/14/2024] [Revised: 10/30/2024] [Accepted: 11/03/2024] [Indexed: 01/03/2025] Open
Abstract
Myocardial injury is a recognized complication of acute pancreatitis, whereas myocarditis has only been occasionally reported and has not been systematically evaluated. We systematically reviewed PubMed literature published up to January 2024 for studies including both "myocarditis" and "pancreatitis" as keywords. Relevant data regarding patient characteristics and outcomes were collected and analyzed. A total of 31 patients from 31 independent studies were included. The etiology of pancreatitis was viral in 52%, bacterial in 20%, toxic in 16%, autoimmune in 9%, and idiopathic in 3%. 23% of patients were immunocompromised. Median high sensitivity-cardiac troponin T was 342 (IQR 73-890) ng/L and N-terminus-pro-brain natriuretic peptide was 11053 (IQR 1397-26150) pg/mL. The average left ventricular ejection fraction was 33±13%. Fulminant myocarditis, presenting with cardiogenic shock and/or malignant ventricular arrhythmias occurred in 48% of patients, more frequently in men than in women (P=0.026). Severe myocarditis occurred in 42% of edematous and 60% of necrotizing pancreatitis (P=0.56). No association was found between the severity of myocarditis and plasma levels of amylase (P=0.98) and lipase (P=0.83). The relative frequency of severe myocarditis was 80% in pancreatitis due to Leptospirosis, and 40% in pancreatitis due to viral infections. The mortality rate was 22%: 13% died during hospitalization and 9% after. Myocarditis is a potentially lethal complication of pancreatitis and is more frequently associated with viral etiology in immunocompromised individuals. Based on such findings, cardiac troponin measurements and an electrocardiogram are advisable to exclude myocardial involvement in selected patients. Confirmatory diagnosis and prognostic assessments should be based on cardiac magnetic resonance imaging.
Collapse
Affiliation(s)
- Mattia Alberti
- Department of Surgical, Medical and Molecular Pathology and Critical Area, Cardiology Division, University of Pisa, Pisa, Italy
| | - Alessandro Marcucci
- Department of Surgical, Medical and Molecular Pathology and Critical Area, Radiology Division, University of Pisa, Pisa, Italy
| | - Filippo Biondi
- Department of Surgical, Medical and Molecular Pathology and Critical Area, Cardiology Division, University of Pisa, Pisa, Italy
| | - Simona Chiusolo
- Department of Surgical, Medical and Molecular Pathology and Critical Area, Cardiology Division, University of Pisa, Pisa, Italy
| | - Gabriele Masini
- Department of Surgical, Medical and Molecular Pathology and Critical Area, Cardiology Division, University of Pisa, Pisa, Italy
| | - Lorenzo Faggioni
- Department of Surgical, Medical and Molecular Pathology and Critical Area, Radiology Division, University of Pisa, Pisa, Italy
| | - Dania Cioni
- Department of Surgical, Medical and Molecular Pathology and Critical Area, Radiology Division, University of Pisa, Pisa, Italy
| | - Doralisa Morrone
- Department of Surgical, Medical and Molecular Pathology and Critical Area, Cardiology Division, University of Pisa, Pisa, Italy
| | - Raffaele De Caterina
- Department of Surgical, Medical and Molecular Pathology and Critical Area, Cardiology Division, University of Pisa, Pisa, Italy
| | - Emanuele Neri
- Department of Surgical, Medical and Molecular Pathology and Critical Area, Radiology Division, University of Pisa, Pisa, Italy
| | - Giovanni Donato Aquaro
- Department of Surgical, Medical and Molecular Pathology and Critical Area, Radiology Division, University of Pisa, Pisa, Italy
| |
Collapse
|
2
|
Li Y, Zhai B, Yang B, Wang B, Wang Y, Qu M, Tang Y. Immune myocarditis induced by sintilimab therapy: A case report. Exp Ther Med 2024; 28:333. [PMID: 39006500 PMCID: PMC11240266 DOI: 10.3892/etm.2024.12622] [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: 11/06/2023] [Accepted: 05/09/2024] [Indexed: 07/16/2024] Open
Abstract
Immunotherapy is a potent tool used in cancer treatment, but the occurrence of immune-related adverse events induced by immune checkpoint inhibitors (ICIs) cannot be overlooked. This is particularly true for rare but potentially fatal cardiovascular complications, such as myocarditis; heart muscle inflammation may lead to heart dysfunction and arrhythmia. The present case is a 68-year-old female breast cancer patient who developed palpitations and elevated cardiac enzyme levels after 1 day of ICI therapy, and the patient was eventually diagnosed with immune myocarditis. After receiving hormonal shock therapy, Ctn I, CK, CK-MB and other cardiac enzyme-related markers improved significantly, and electrocardiogram test returned to normal, and the patient recovered during hospitalization without any major adverse cardiac events. Furthermore, the present study reviewed the mechanism of immune myocarditis induced by ICI therapy, with the aim of providing a clinical foundation for the prevention and diagnosis of cardiovascular adverse events in ICI therapy.
Collapse
Affiliation(s)
- Yu Li
- Translational Medical Center, Weifang Second People's Hospital, Weifang Respiratory Disease Hospital, Weifang, Shandong 261041, P.R. China
- Department of Cardiology, Weifang Second People's Hospital, Weifang Respiratory Disease Hospital, Weifang, Shandong 261041, P.R. China
| | - Baowei Zhai
- Department of Cardiology, Weifang Second People's Hospital, Weifang Respiratory Disease Hospital, Weifang, Shandong 261041, P.R. China
| | - Ben Yang
- Translational Medical Center, Weifang Second People's Hospital, Weifang Respiratory Disease Hospital, Weifang, Shandong 261041, P.R. China
| | - Bin Wang
- Department of Cardiology, Weifang Second People's Hospital, Weifang Respiratory Disease Hospital, Weifang, Shandong 261041, P.R. China
| | - Yubing Wang
- Translational Medical Center, Weifang Second People's Hospital, Weifang Respiratory Disease Hospital, Weifang, Shandong 261041, P.R. China
| | - Meihua Qu
- Translational Medical Center, Weifang Second People's Hospital, Weifang Respiratory Disease Hospital, Weifang, Shandong 261041, P.R. China
| | - Yuanyuan Tang
- Translational Medical Center, Weifang Second People's Hospital, Weifang Respiratory Disease Hospital, Weifang, Shandong 261041, P.R. China
| |
Collapse
|
3
|
Zhou J, Wang C, Hua X, Liu Y, Song J, Hou R. Exploring the Feasibility of Investigating Myocarditis Progression Using Histopathological Features from Whole-Slide Images of H&E Tissue. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2024; 2024:1-4. [PMID: 40039394 DOI: 10.1109/embc53108.2024.10781865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Myocarditis is an inflammatory condition affecting the heart muscle, if left unaddressed, may lead to severe complications such as heart failure, arrhythmias, and sudden cardiac death. Current subtyping methods lack sufficient pathological analysis to guide healthcare. Here, we present a clustering analysis using pathological features of H&E stained whole-slide images of from 39 patients with myocarditis who have gone through heart transplantation. Totally, 3,791 pathological features were extracted using each nucleus segmented from StarDist segmentation network. After feature dimensionality reduction, 181 features were selected for clustering with K-Modes method. Two groups of patients with significant differences in time from heart failure to transplantation and time from onset to transplantation were identified. This study demonstrates the feasibility of using pathohistological images to enhance the progression assessment of patients with myocarditis, which may help improve diagnostic accuracy and facilitating targeted therapeutic interventions for cardiac-related diseases.
Collapse
|
4
|
Wang T, Wang S, Jia X, Li C, Ma X, Tong H, Liu M, Li L. Baicalein alleviates cardiomyocyte death in EAM mice by inhibiting the JAK-STAT1/4 signalling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155558. [PMID: 38547614 DOI: 10.1016/j.phymed.2024.155558] [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: 11/24/2023] [Revised: 03/12/2024] [Accepted: 03/19/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND The experimental autoimmune myocarditis (EAM) model is valuable for investigating myocarditis pathogenesis. M1-type macrophages and CD4+T cells exert key pathogenic effects on EAM initiation and progression. Baicalein (5,6,7-trihydroxyflavone, C15H10O5, BAI), which is derived from the Scutellaria baicalensis root, is a primary bioactive compound with potent anti-inflammatory and antioxidant properties. BAI exerts good therapeutic effects against various autoimmune diseases; however, its effect in EAM has not been thoroughly researched. PURPOSE This study aimed to explore the possible inhibitory effect of BAI on M1 macrophage polarisation and CD4+T cell differentiation into Th1 cells via modulation of the JAK-STAT1/4 signalling pathway, which reduces the secretion of pro-inflammatory factors, namely, TNF-α and IFN-γ, and consequently inhibits TNF-α- and IFN-γ-triggered apoptosis in cardiomyocytes of the EAM model mice. STUDY DESIGN AND METHODS Flow cytometry, immunofluorescence, real-time quantitative polymerase chain reaction (q-PCR), and western blotting were performed to determine whether BAI alleviated M1/Th1-secreted TNF-α- and IFN-γ-induced myocyte death in the EAM model mice through the inhibition of the JAK-STAT1/4 signalling pathway. RESULTS These results indicate that BAI intervention in mice resulted in mild inflammatory infiltrates. BAI inhibited JAK-STAT1 signalling in macrophages both in vivo and in vitro, which attenuated macrophage polarisation to the M1 type and reduced TNF-α secretion. Additionally, BAI significantly inhibited the differentiation of CD4+T cells to Th1 cells and IFN-γ secretion both in vivo and in vitro by modulating the JAK-STAT1/4 signalling pathway. This ultimately led to decreased TNF-α and IFN-γ levels in cardiac tissues and reduced myocardial cell apoptosis. CONCLUSION This study demonstrates that BAI alleviates M1/Th1-secreted TNF-α- and IFN-γ-induced cardiomyocyte death in EAM mice by inhibiting the JAK-STAT1/4 signalling pathway.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical, Qingdao University, 308 Ningxia Road, Qingdao, Shandong 266071, China
| | - Shuang Wang
- Department of Biochemistry, School of Basic Medical, Qingdao University, Qingdao, China
| | - Xihui Jia
- Department of Biochemistry, School of Basic Medical, Qingdao University, Qingdao, China
| | - Chenglin Li
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical, Qingdao University, 308 Ningxia Road, Qingdao, Shandong 266071, China
| | - Xiaoran Ma
- School of Medicine, Qing dao Binhai University, Qingdao, China
| | - Huimin Tong
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical, Qingdao University, 308 Ningxia Road, Qingdao, Shandong 266071, China
| | - Meng Liu
- Department of Biochemistry, School of Basic Medical, Qingdao University, Qingdao, China
| | - Ling Li
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical, Qingdao University, 308 Ningxia Road, Qingdao, Shandong 266071, China.
| |
Collapse
|
5
|
Azzam M, Awad A, Abugharbyeh A, Kahaleh B. Myocarditis in connective tissue diseases: an often-overlooked clinical manifestation. Rheumatol Int 2023; 43:1983-1992. [PMID: 37587233 DOI: 10.1007/s00296-023-05428-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/08/2023] [Indexed: 08/18/2023]
Abstract
To discuss what is currently known about myocarditis in the context of major connective tissue diseases, including Systemic lupus erythematosus, Rheumatoid Arthritis, Sjogren, Dermato-myositis and Polymyositis, Systemic Sclerosis, and Mixed connective tissue disease. Variability exists between studies regarding the incidence of myocarditis in connective tissue diseases, which is hypothesized to be the result of its subclinical course in most cases. Extensive gaps of knowledge exist in the field of pathophysiology. Although endomyocardial biopsy remains to be the gold standard for diagnosis, the advancement in non-invasive modalities such as cardiac MRI, echocardiography, and nuclear medicine has allowed for earlier and more frequent detection of myocarditis. A lack of treatment guidelines was found across the different connective tissue diseases. Most of the literature available revolved around myocarditis in the context of Systemic lupus erythematosus. Numerous recent studies were published that contributed to advancements in diagnosis and treatment however, there remains a lack of diagnostic and treatment guidelines.
Collapse
Affiliation(s)
- Muayad Azzam
- Faculty of Medicine, The University of Jordan, Amman, 11942, Jordan.
| | - Amro Awad
- Faculty of Medicine, The University of Jordan, Amman, 11942, Jordan
| | - Aya Abugharbyeh
- Division of Rheumatology and Immunology, University of Toledo Medical Center, Toledo, USA
| | - Bashar Kahaleh
- Division of Rheumatology and Immunology, University of Toledo Medical Center, Toledo, USA
| |
Collapse
|
6
|
Fu L, Zou Y, Yu B, Hong D, Guan T, Hu J, Xu Y, Wu Y, Kou J, Lv Y. Background and roles: myosin in autoimmune diseases. Front Cell Dev Biol 2023; 11:1220672. [PMID: 37691828 PMCID: PMC10484797 DOI: 10.3389/fcell.2023.1220672] [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: 05/13/2023] [Accepted: 08/09/2023] [Indexed: 09/12/2023] Open
Abstract
The myosin superfamily is a group of molecular motors. Autoimmune diseases are characterized by dysregulation or deficiency of the immune tolerance mechanism, resulting in an immune response to the human body itself. The link between myosin and autoimmune diseases is much more complex than scientists had hoped. Myosin itself immunization can induce experimental autoimmune diseases of animals, and myosins were abnormally expressed in a number of autoimmune diseases. Additionally, myosin takes part in the pathological process of multiple sclerosis, Alzheimer's disease, Parkinson's disease, autoimmune myocarditis, myositis, hemopathy, inclusion body diseases, etc. However, research on myosin and its involvement in the occurrence and development of diseases is still in its infancy, and the underlying pathological mechanisms are not well understood. We can reasonably predict that myosin might play a role in new treatments of autoimmune diseases.
Collapse
Affiliation(s)
- Longsheng Fu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yonghui Zou
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Boyang Yu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangxi, China
| | - Daojun Hong
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Teng Guan
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada
| | - Jinfang Hu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yi Xu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yaoqi Wu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Junping Kou
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangxi, China
| | - Yanni Lv
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
7
|
Interdonato L, Impellizzeri D, D’Amico R, Cordaro M, Siracusa R, D’Agostino M, Genovese T, Gugliandolo E, Crupi R, Fusco R, Cuzzocrea S, Di Paola R. Modulation of TLR4/NFκB Pathways in Autoimmune Myocarditis. Antioxidants (Basel) 2023; 12:1507. [PMID: 37627502 PMCID: PMC10451772 DOI: 10.3390/antiox12081507] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/14/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
Myocarditis is an inflammatory and oxidative disorder characterized by immune cell recruitment in the damaged tissue and organ dysfunction. In this paper, we evaluated the molecular pathways involved in myocarditis using a natural compound, Coriolus versicolor, in an experimental model of autoimmune myocarditis (EAM). Animals were immunized with an emulsion of pig cardiac myosin and complete Freund's adjuvant supplemented with mycobacterium tuberculosis; thereafter, Coriolus versicolor (200 mg/Kg) was orally administered for 21 days. At the end of the experiment, blood pressure and heart rate measurements were recorded and the body and heart weights as well. From the molecular point of view, the Coriolus versicolor administration reduced the activation of the TLR4/NF-κB pathway and the levels of pro-inflammatory cytokines (INF-γ, TNF-α, IL-6, IL-17, and IL-2) and restored the levels of anti-inflammatory cytokines (IL-10). These anti-inflammatory effects were accompanied with a reduced lipid peroxidation and nitrite levels and restored the antioxidant enzyme activities (SOD and CAT) and GSH levels. Additionally, it reduced the histological injury and the immune cell recruitment (CD4+ and CD68+ cells). Moreover, we observed an antiapoptotic activity in both intrinsic (Fas/FasL/caspase-3) and extrinsic (Bax/Bcl-2) pathways. Overall, our data showed that Coriolus versicolor administration modulates the TLR4/NF-κB signaling in EAM.
Collapse
Affiliation(s)
- Livia Interdonato
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Ramona D’Amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Marika Cordaro
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, Consolare Valeria, 98100 Messina, Italy
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Melissa D’Agostino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Tiziana Genovese
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Enrico Gugliandolo
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Rosalia Crupi
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Rosanna Di Paola
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| |
Collapse
|
8
|
Almalki ME, Alshumrani FA, Almalki HA, Saati AA, Alzahrani SE, Khouj SM. Regional Myopericarditis Mimicking Inferior Myocardial Infarction Following COVID-19 Vaccination: A Rare Adverse Event. Cureus 2023; 15:e41168. [PMID: 37529509 PMCID: PMC10389750 DOI: 10.7759/cureus.41168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2023] [Indexed: 08/03/2023] Open
Abstract
We present the case of a 41-year-old man who developed myopericarditis after receiving the Pfizer COVID-19 vaccine. The patient experienced a sudden onset of chest and abdominal pain 16 days after vaccination. Electrocardiogram findings revealed deep T-wave inversion and minimal ST-segment elevation. Further investigation through coronary artery angiography and computed tomography identified an anomalous left main coronary artery. Magnetic resonance imaging confirmed the diagnosis of myopericarditis. This case highlights the importance of considering myopericarditis as a potential cause of chest pain and elevated cardiac biomarkers following COVID-19 vaccination, particularly in young individuals. Clinicians should be aware of this adverse event and include it in the differential diagnosis for patients presenting with similar symptoms after vaccination.
Collapse
Affiliation(s)
- Mohammed E Almalki
- Department of Medicine, College of Medicine, Umm Al-Qura University, Makkah, SAU
| | - Fahad A Alshumrani
- Department of Medicine, College of Medicine, Umm Al-Qura University, Makkah, SAU
| | - Hussam A Almalki
- Department of Medicine, College of Medicine, Umm Al-Qura University, Makkah, SAU
| | - Asim A Saati
- Department of Medicine, College of Medicine, Umm Al-Qura University, Makkah, SAU
| | - Saeed E Alzahrani
- Department of Medicine, College of Medicine, Umm Al-Qura University, Makkah, SAU
| | - Saleh M Khouj
- Department of Interventional Cardiology and Structural Heart Disease, King Abdullah Medical City, Makkah, SAU
| |
Collapse
|
9
|
Xu J, Zhou Z, Zheng Y, Yang S, Huang K, Li H. Roles of inflammasomes in viral myocarditis. Front Cell Infect Microbiol 2023; 13:1149911. [PMID: 37256114 PMCID: PMC10225676 DOI: 10.3389/fcimb.2023.1149911] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/28/2023] [Indexed: 06/01/2023] Open
Abstract
Viral myocarditis (VMC), characterized by viral infection-induced inflammation, is a life-threatening disease associated with dilated cardiomyopathy or heart failure. Innate immunity plays a crucial role in the progression of inflammation, in which inflammasomes provide a platform for the secretion of cytokines and mediate pyroptosis. Inflammasomes are rising stars gaining increasing attention. The nucleotide oligomerization domain-, leucine-rich repeat-, and pyrin domain-containing protein 3 (NLRP3) inflammasome, the caspase recruitment domain-containing protein 8 (CARD8) inflammasome, and the caspase-11 inflammasome are three inflammasomes that were reported to affect the process and prognosis of VMC. These inflammasomes can be activated by a wide range of cellular events. Accumulating evidence has suggested that inflammasomes are involved in different stages of VMC, including the trigger and progression of myocardial injury and remodeling after infection. In this review, we summarized the pathways involving inflammasomes in VMC and discussed the potential therapies targeting inflammasomes and related pathways.
Collapse
Affiliation(s)
- Jingyu Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zihao Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yidan Zheng
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sai Yang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Huang
- Institution of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huili Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
10
|
Kyaw T, Drummond G, Bobik A, Peter K. Myocarditis: causes, mechanisms, and evolving therapies. Expert Opin Ther Targets 2023; 27:225-238. [PMID: 36946552 DOI: 10.1080/14728222.2023.2193330] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
INTRODUCTION Myocarditis is a severe lymphocyte-mediated inflammatory disorder of the heart, mostly caused by viruses and immune checkpoint inhibitors (ICIs). Recently, myocarditis as a rare adverse event of mRNA vaccines for SARS-CoV-2 has caused global attention. The clinical consequences of myocarditis can be very severe, but specific treatment options are lacking or not yet clinically proven. AREAS COVERED This paper offers a brief overview of the biology of viruses that frequently cause myocarditis, focusing on mechanisms important for viral entry and replication following host infection. Current and new potential therapeutic targets/strategies especially for viral myocarditis are reviewed systematically. In particular, the immune system in myocarditis is dissected with respect to infective viral and non-infective, ICI-induced myocarditis. EXPERT OPINION Vaccination is an excellent emerging preventative strategy for viral myocarditis, but most vaccines still require further development. Anti-viral treatments that inhibit viral replication need to be considered following viral infection in host myocardium, as lower viral load reduces inflammation severity. Understanding how the immune system continues to damage the heart even after viral clearance will define novel therapeutic targets/strategies. We propose that viral myocarditis can be best treated using a combination of antiviral agents and immunotherapies that control cytotoxic T cell activity.
Collapse
Affiliation(s)
- Tin Kyaw
- Inflammation and Cardiovascular Disease Laboratory, Baker Heart and Diabetes Institute
- Centre for Inflammatory Diseases, Monash Medical Centre, Monash University, Melbourne, Australia
- Department of Cardiometabolic Health, University of Melbourne Melbourne Australia
| | - Grant Drummond
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University Melbourne Australia
- Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, Australia
| | - Alex Bobik
- Inflammation and Cardiovascular Disease Laboratory, Baker Heart and Diabetes Institute
- Centre for Inflammatory Diseases, Monash Medical Centre, Monash University, Melbourne, Australia
- Department of Cardiometabolic Health, University of Melbourne Melbourne Australia
- Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, Australia
- Heart Centre, Alfred Hospital, Melbourne, Australia
| | - Karlheinz Peter
- Inflammation and Cardiovascular Disease Laboratory, Baker Heart and Diabetes Institute
- Department of Cardiometabolic Health, University of Melbourne Melbourne Australia
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University Melbourne Australia
- Heart Centre, Alfred Hospital, Melbourne, Australia
- Department of Immunology, Monash University Melbourne Australia
| |
Collapse
|
11
|
Feng G, Zhu C, Lin CY, Bredemeyer A, Förster I, Kreisel D, Lavine KJ. CCL17 Protects Against Viral Myocarditis by Suppressing the Recruitment of Regulatory T Cells. J Am Heart Assoc 2023; 12:e028442. [PMID: 36752267 PMCID: PMC10111487 DOI: 10.1161/jaha.122.028442] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Background Viral myocarditis is characterized by leukocyte infiltration of the heart and cardiomyocyte death. We recently identified C-C chemokine ligand (CCL) 17 as a proinflammatory effector of C-C chemokine receptor 2-positive macrophages and dendritic cells that are recruited to the heart and contribute to adverse left ventricular remodeling following myocardial infarction and pressure overload. Methods and Results Mouse encephalomyocarditis virus was used to investigate the function of CCL17 in a viral myocarditis model. Ccl17Gfp reporter and knockout mice were used to identify the cell types that express CCL17 and delineate the functional importance of CCL17 in encephalomyocarditis virus clearance and myocardial inflammation. Cardiac CCL17 was expressed in C-C chemokine receptor 2-positive macrophages and dendritic cells following encephalomyocarditis virus infection. Colony-stimulating factor 2 (granulocyte-macrophage colony-stimulating factor) signaling was identified as a key regulator of CCL17 expression. Ccl17 deletion resulted in impaired encephalomyocarditis virus clearance, increased cardiomyocyte death, and higher mortality during infection early stage, and aggravated hypertrophy and fibrotic responses in infection long-term stage. An increased abundance of regulatory T cells was detected in the myocardium of injured Ccl17-deficient mice. Depletion of regulatory T cells in Ccl17-deficient mice abrogated the detrimental role of CCL17 deletion by restoring interferon signaling. Conclusions Collectively, these findings identify CCL17 as an important mediator of the host immune response during cardiac viral infection early stage and suggest that CCL17 targeted therapies should be avoided in acute viral myocarditis.
Collapse
Affiliation(s)
- Guoshuai Feng
- Cardiovascular Division, Department of Medicine Washington University School of Medicine St. Louis MO
| | - Cuige Zhu
- Division of Oncology Washington University School of Medicine St. Louis MO
| | - Chieh-Yu Lin
- Department of Pathology and Immunology Washington University St. Louis MO
| | - Andrea Bredemeyer
- Cardiovascular Division, Department of Medicine Washington University School of Medicine St. Louis MO
| | - Irmgard Förster
- Immunology and Environment, LIMES Institute University of Bonn Germany
| | - Daniel Kreisel
- Department of Surgery Washington University St. Louis MO
| | - Kory J Lavine
- Cardiovascular Division, Department of Medicine Washington University School of Medicine St. Louis MO.,Department of Pathology and Immunology Washington University St. Louis MO.,Department of Developmental Biology Washington University St. Louis MO
| |
Collapse
|
12
|
Makrutzki-Zlotek K, Escher F, Karadeniz Z, Aleshcheva G, Pietsch H, Küchler K, Schultheiss HP, Heidecker B, Poller W, Landmesser U, Scheibenbogen C, Thevathasan T, Skurk C. FOXO3A acts as immune response modulator in human virus-negative inflammatory cardiomyopathy. HEART (BRITISH CARDIAC SOCIETY) 2023; 109:846-856. [PMID: 36702542 DOI: 10.1136/heartjnl-2022-321732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 01/02/2023] [Indexed: 01/27/2023]
Abstract
OBJECTIVE Inflammatory cardiomyopathy is characterised by inflammatory infiltrates leading to cardiac injury, left ventricular (LV) dilatation and reduced LV ejection fraction (LVEF). Several viral pathogens and autoimmune phenomena may cause cardiac inflammation.The effects of the gain of function FOXO3A single-nucleotide polymorphism (SNP) rs12212067 on inflammation and outcome were studied in a cohort of patients with inflammatory dilated cardiomyopathy (DCMi) in relation to cardiac viral presence. METHODS Distribution of the SNP was determined in virus-positive and virus-negative DCMi patients and in control subjects without myocardial pathology. Baseline and outcome data were compared in 221 virus-negative patients with detection of cardiac inflammation and reduced LVEF according to their carrier status of the SNP. RESULTS Distribution of SNP rs12212067 did not differ between virus-positive (n=22, 19.3%), virus-negative (n=45, 20.4 %) and control patients (n=18, 23.4 %), indicating the absence of susceptibility for viral infection or inflammation per se (p=0.199). Patients in the virus-negative DCMi group were characterised by reduced LVEF 35.5% (95% CI) 33.5 to 37.4) and increased LVEDD (LV end-diastolic diameter) 59.8 mm (95% CI 58.5 to 61.2). Within the group, SNP and non-SNP carriers had similarly impaired LVEF 39.2% (95% CI 34.3% to 44.0%) vs 34.5% (95% CI 32.4 to 36.5), p=0.083, and increased LVEDD 58.9 mm (95% CI 56.3 to 61.5) vs 60.1 mm (95% CI 58.6 to 61.6), p=0.702, respectively. The number of inflammatory infiltrates was not different in both SNP groups at baseline. Outcome after 6 months showed a significant improvement in LVEF and clinical symptoms in SNP rs12212067 carriers 50.9% (95% CI 45.4 to 56.3) versus non-SNP carriers 41.7% (95% CI 39.2 to 44.2), p≤0.01. The improvement in clinical symptoms and LVEF was associated with a significant reduction in cardiac inflammation (ΔCD45RO+ p≤0.05; ΔMac-1+ p≤0.05; ΔLFA-1+ p≤0.01; ΔCD54+ p≤0.01) in the SNP cohort versus non-SNP cohort, respectively. Subgroup analyses identified ΔMac-1+, ΔLFA-1+, ΔCD3+ and Δperforin+ as predictors for improvement in cardiac function in SNP-positive patients. CONCLUSION FOXO3A might act as modulator of the cardiac immune response, diminishing cardiac inflammation and injury in pathogen-negative DCMi.
Collapse
Affiliation(s)
- Kamila Makrutzki-Zlotek
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany
| | - Felicitas Escher
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Virchow-Klinikum, Berlin, Germany.,DZHK, German Center for Cardiovascular Research, Berlin, Germany.,IKDT, Institute for Cardiac Diagnostics and Therapy, Berlin, Germany
| | - Zehra Karadeniz
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany
| | - Ganna Aleshcheva
- IKDT, Institute for Cardiac Diagnostics and Therapy, Berlin, Germany
| | - Heiko Pietsch
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Virchow-Klinikum, Berlin, Germany.,IKDT, Institute for Cardiac Diagnostics and Therapy, Berlin, Germany
| | - Konstanze Küchler
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany
| | | | - Bettina Heidecker
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany.,Institute of Medical Informatics, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Wolfgang Poller
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany.,Institute of Medical Informatics, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Ulf Landmesser
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany.,Institute of Medical Informatics, Charité Universitätsmedizin Berlin, Berlin, Germany.,BIH, Berlin Institute of Health at Charité, Berlin, Germany
| | - Carmen Scheibenbogen
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin - Campus Virchow-Klinikum, Berlin, Germany
| | - Tharusan Thevathasan
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany.,DZHK, German Center for Cardiovascular Research, Berlin, Germany.,Institute of Medical Informatics, Charité Universitätsmedizin Berlin, Berlin, Germany.,BIH, Berlin Institute of Health at Charité, Berlin, Germany
| | - Carsten Skurk
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany .,DZHK, German Center for Cardiovascular Research, Berlin, Germany
| |
Collapse
|
13
|
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: 1.0] [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
|
14
|
Pediatric Myocarditis: What Have We Learnt So Far? J Cardiovasc Dev Dis 2022; 9:jcdd9050143. [PMID: 35621854 PMCID: PMC9144089 DOI: 10.3390/jcdd9050143] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/23/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023] Open
Abstract
Myocarditis is an inflammatory disease of the myocardium that is troublesome to diagnose and manage, especially in children. Since the introduction of endomyocardial biopsy (EMB), new diagnostic tools have provided useful data. Especially when enhanced with immunohistochemistry and polymerase chain reaction (PCR) studies, EMB remains the gold standard for the diagnosis. Notably, cardiac magnetic resonance (MRI) is a non-invasive tool that can confirm the diagnosis and has a particular usefulness during the follow-up. The causes of myocarditis are heterogeneous (mostly viral in children). The course and outcome of the illness in the pediatric population represent a complex interaction between etiologic agents and the immune system, which is still not fully understood. The clinical presentation and course of myocarditis vary widely from paucisymptomatic illness to acute heart failure refractory to therapy, arrhythmias, angina-like presentation and sudden cardiac death. In this setting, cardiac biomarkers (i.e., troponins and BNP), although unspecific, can be used to support the diagnosis. Finally, the efficacy of therapeutic strategies is controversial and not confirmed by clinical trials. In this review, we summarized the milestones in diagnosis and provided an overview of the therapeutic options for myocarditis in children.
Collapse
|
15
|
Yu K, Zhou L, Wang Y, Yu C, Wang Z, Liu H, Wei H, Han L, Cheng J, Wang F, Wang DW, Zhao C. Mechanisms and Therapeutic Strategies of Viral Myocarditis Targeting Autophagy. Front Pharmacol 2022; 13:843103. [PMID: 35479306 PMCID: PMC9035591 DOI: 10.3389/fphar.2022.843103] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
Viral myocarditis is caused by infection with viruses or bacteria, including coxsackievirus B3 (CVB3), and is characterized by acute or chronic inflammatory responses in the heart. The mortality associated with severe viral myocarditis is considerable. In some patients, viral myocarditis may develop into dilated cardiomyopathy or heart failure. Autophagy is involved in a wide range of physiological processes, including viral infection and replication. In the present review, we focus on the responses of cardiac tissues, cardiomyocytes, and cardiac fibroblasts to CVB3 infection. Subsequently, the effects of altered autophagy on the development of viral myocarditis are discussed. Finally, this review also examined and assessed the use of several popular autophagy modulating drugs, such as metformin, resveratrol, rapamycin, wortmannin, and 3-methyladenine, as alternative treatment strategies for viral myocarditis.
Collapse
Affiliation(s)
- Kun Yu
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Zhou
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yinhui Wang
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengxin Yu
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ziyi Wang
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Liu
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haoran Wei
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liang Han
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Jia Cheng
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Wang
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dao Wen Wang
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunxia Zhao
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Chunxia Zhao,
| |
Collapse
|
16
|
Cardiovascular Magnetic Resonance Imaging Pattern in Campylobacter jejuni-related Myocarditis. Microorganisms 2022; 10:microorganisms10020208. [PMID: 35208663 PMCID: PMC8878248 DOI: 10.3390/microorganisms10020208] [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: 12/10/2021] [Revised: 12/29/2021] [Accepted: 01/17/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Campylobacter jejuni (C. jejuni) is a common cause of mostly self-limiting enterocolitis. Although rare, myocarditis has been increasingly documented as a complication following campylobacteriosis. Such cases have occurred predominantly in younger males and involved a single causative species, namely C. jejuni. Case report: We report herein a case of myocarditis complicating gastroenteritis in a 23-year-old immunocompetent patient, caused by this bacterium with a favorable outcome. Cardiac magnetic resonance imagining was useful in establishing an early diagnosis. Conclusions: Myocarditis should be considered in younger patients presenting with chest pain and plasmatic troponin elevations. The occurrence of myocarditis complicating C. jejuni is reviewed.
Collapse
|
17
|
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
|
18
|
Zhang MW, Wang XH, Shi J, Yu JG. Sinomenine in Cardio-Cerebrovascular Diseases: Potential Therapeutic Effects and Pharmacological Evidences. Front Cardiovasc Med 2021; 8:749113. [PMID: 34660748 PMCID: PMC8517137 DOI: 10.3389/fcvm.2021.749113] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/08/2021] [Indexed: 12/12/2022] Open
Abstract
Cardio-cerebrovascular diseases, as a major cause of health loss all over the world, contribute to an important part of the global burden of disease. A large number of traditional Chinese medicines have been proved effective both clinically and in pharmacological investigations, with the acceleration of the modernization of Chinese medicine. Sinomenine is the main active constituent of sinomenium acutum and has been generally used in therapies of rheumatoid arthritis and neuralgia. Varieties of pharmacological effects of sinomenine in cardio-cerebrovascular system have been discovered recently, suggesting an inspiring application prospect of sinomenine in cardio-cerebrovascular diseases. Sinomenine may retard the progression of atherosclerosis by attenuating endothelial inflammation, regulating immune cells function, and inhibiting the proliferation of vascular smooth muscle cells. Sinomenine also alleviates chronic cardiac allograft rejection relying on its anti-inflammatory and anti-hyperplastic activities and suppresses autoimmune myocarditis by immunosuppression. Prevention of myocardial or cerebral ischemia-reperfusion injury by sinomenine is associated with its modulation of cardiomyocyte death, inflammation, calcium overload, and oxidative stress. The regulatory effects on vasodilation and electrophysiology make sinomenine a promising drug to treat hypertension and arrhythmia. Here, in this review, we will illustrate the pharmacological activities of sinomenine in cardio-cerebrovascular system and elaborate the underlying mechanisms, as well as give an overview of the potential therapeutic roles of sinomenine in cardio-cerebrovascular diseases, trying to provide clues and bases for its clinical usage.
Collapse
Affiliation(s)
- Meng-Wan Zhang
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Hui Wang
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Shi
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jian-Guang Yu
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
19
|
Bockstahler M, Salbach C, Müller AM, Kübler A, Müller OJ, Katus HA, Frey N, Kaya Z. LNA oligonucleotide mediates an anti-inflammatory effect in autoimmune myocarditis via targeting lactate dehydrogenase B. Immunology 2021; 165:158-170. [PMID: 34606637 PMCID: PMC9426621 DOI: 10.1111/imm.13421] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 08/18/2021] [Accepted: 09/14/2021] [Indexed: 11/29/2022] Open
Abstract
Treatment of myocarditis is often limited to symptomatic treatment due to unknown pathomechanisms. In order to identify new therapeutic approaches, the contribution of locked nucleic acid antisense oligonucleotides (LNA ASOs) in autoimmune myocarditis was investigated. Hence, A/J mice were immunized with cardiac troponin I (TnI) to induce experimental autoimmune myocarditis (EAM) and treated with LNA ASOs. The results showed an unexpected anti‐inflammatory effect for one administered LNA ASO MB_1114 by reducing cardiac inflammation and fibrosis. The target sequence of MB_1114 was identified as lactate dehydrogenase B (mLDHB). For further analysis, mice received mLdhb‐specific GapmeR during induction of EAM. Here, mice receiving the mLdhb‐specific GapmeR showed increased protein levels of cardiac mLDHB and a reduced cardiac inflammation and fibrosis. The effect of increased cardiac mLDHB protein level was associated with a downregulation of genes of reactive oxygen species (ROS)‐associated proteins, indicating a reduction in ROS. Here, the suppression of murine pro‐apoptotic Bcl‐2‐associated X protein (mBax) was also observed. In our study, an unexpected anti‐inflammatory effect of LNA ASO MB_1114 and mLdhb‐specific GapmeR during induction of EAM could be demonstrated in vivo. This effect was associated with increased protein levels of cardiac mLDHB, mBax suppression and reduced ROS activation. Thus, LDHB and LNA ASOs may be considered as a promising target for directed therapy of myocarditis. Nevertheless, further investigations are necessary to clarify the mechanism of action of anti‐inflammatory LDHB‐triggered effects.
Collapse
Affiliation(s)
- Mariella Bockstahler
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Christian Salbach
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Anna-Maria Müller
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Andrea Kübler
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Oliver J Müller
- Department of Internal Medicine III, University of Kiel, Kiel, Germany
| | - Hugo A Katus
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Norbert Frey
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Ziya Kaya
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
20
|
Suh HN, Kim YK, Lee JY, Kang GH, Hwang JH. Dissect the immunity using cytokine profiling and NF-kB target gene analysis in systemic inflammatory minipig model. PLoS One 2021; 16:e0252947. [PMID: 34086835 PMCID: PMC8177627 DOI: 10.1371/journal.pone.0252947] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/25/2021] [Indexed: 11/18/2022] Open
Abstract
Minipigs have remarkably similar physiology to humans, therefore, they it can be a good animal model for inflammation study. Thus, the conventional (serum chemistry, histopathology) and novel analytic tools [immune cell identification in tissue, cytokine level in peripheral blood mononuclear cells (PBMC) and serum, NF-kB target gene analysis in tissue] were applied to determine inflammation in Chicago Miniature Swine (CMS) minipig. Lipopolysaccharide (LPS)-induced acute systemic inflammation caused liver and kidney damage in serum chemistry and histopathology. Immunohistochemistry (IHC) also showed an increase of immune cell distribution in spleen and lung during inflammation. Moreover, NF-kB-target gene expression was upregulated in lung and kidney in acute inflammation and in heart, liver, and intestine in chronic inflammation. Cytokine mRNA was elevated in PBMC under acute inflammation along with elevated absolute cytokine levels in serum. Overall, LPS-mediated systemic inflammation affects the various organs, and can be detected by IHC of immune cells, gene analysis in PBMC, and measuring the absolute cytokine in serum along with conventional inflammation analytic tools.
Collapse
Affiliation(s)
- Han Na Suh
- Animal Model Research Group, Korea Institute of Toxicology, Jeongeup, Jeollabuk-do, Republic of Korea
- * E-mail: (HNS); (JHH)
| | - Young Kyu Kim
- Animal Model Research Group, Korea Institute of Toxicology, Jeongeup, Jeollabuk-do, Republic of Korea
| | - Ju Young Lee
- Animal Model Research Group, Korea Institute of Toxicology, Jeongeup, Jeollabuk-do, Republic of Korea
| | - Goo-Hwa Kang
- Animal Model Research Group, Korea Institute of Toxicology, Jeongeup, Jeollabuk-do, Republic of Korea
| | - Jeong Ho Hwang
- Animal Model Research Group, Korea Institute of Toxicology, Jeongeup, Jeollabuk-do, Republic of Korea
- * E-mail: (HNS); (JHH)
| |
Collapse
|
21
|
Siegel PM, Bojti I, Bassler N, Holien J, Flierl U, Wang X, Waggershauser P, Tonnar X, Vedecnik C, Lamprecht C, Stankova I, Li T, Helbing T, Wolf D, Anto-Michel N, Mitre LS, Ehrlich J, Orlean L, Bender I, Przewosnik A, Mauler M, Hollederer L, Moser M, Bode C, Parker MW, Peter K, Diehl P. A DARPin targeting activated Mac-1 is a novel diagnostic tool and potential anti-inflammatory agent in myocarditis, sepsis and myocardial infarction. Basic Res Cardiol 2021; 116:17. [PMID: 33721106 PMCID: PMC7960600 DOI: 10.1007/s00395-021-00849-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 01/18/2021] [Indexed: 12/15/2022]
Abstract
The monocyte β2-integrin Mac-1 is crucial for leukocyte–endothelium interaction, rendering it an attractive therapeutic target for acute and chronic inflammation. Using phage display, a Designed-Ankyrin-Repeat-Protein (DARPin) was selected as a novel binding protein targeting and blocking the αM I-domain, an activation-specific epitope of Mac-1. This DARPin, named F7, specifically binds to activated Mac-1 on mouse and human monocytes as determined by flow cytometry. Homology modelling and docking studies defined distinct interaction sites which were verified by mutagenesis. Intravital microscopy showed reduced leukocyte–endothelium adhesion in mice treated with this DARPin. Using mouse models of sepsis, myocarditis and ischaemia/reperfusion injury, we demonstrate therapeutic anti-inflammatory effects. Finally, the activated Mac-1-specific DARPin is established as a tool to detect monocyte activation in patients receiving extra-corporeal membrane oxygenation, as well as suffering from sepsis and ST-elevation myocardial infarction. The activated Mac-1-specific DARPin F7 binds preferentially to activated monocytes, detects inflammation in critically ill patients, and inhibits monocyte and neutrophil function as an efficient new anti-inflammatory agent.
Collapse
Affiliation(s)
- Patrick M Siegel
- Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - István Bojti
- Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nicole Bassler
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Jessica Holien
- ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Melbourne, Australia
| | - Ulrike Flierl
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Xiaowei Wang
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia.,Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia.,Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Philipp Waggershauser
- Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Xavier Tonnar
- Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christopher Vedecnik
- Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Constanze Lamprecht
- BIOSS Centre for Biological Signalling Studies/Synthetic Biology of Signalling Processes, University of Freiburg, Freiburg, Germany
| | - Ivana Stankova
- Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tian Li
- Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Thomas Helbing
- Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dennis Wolf
- Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nathaly Anto-Michel
- Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lucia Sol Mitre
- Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julia Ehrlich
- Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lukas Orlean
- Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ileana Bender
- Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anne Przewosnik
- Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maximilian Mauler
- Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Laura Hollederer
- Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Martin Moser
- Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christoph Bode
- Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Michael W Parker
- ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Melbourne, Australia.,Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia.,Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia. .,Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia. .,Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia. .,Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia.
| | - Philipp Diehl
- Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia.,Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| |
Collapse
|
22
|
Zhang MW, Shen YJ, Shi J, Yu JG. MiR-223-3p in Cardiovascular Diseases: A Biomarker and Potential Therapeutic Target. Front Cardiovasc Med 2021; 7:610561. [PMID: 33553260 PMCID: PMC7854547 DOI: 10.3389/fcvm.2020.610561] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 12/23/2020] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular diseases, involving vasculopathy, cardiac dysfunction, or circulatory disturbance, have become the major cause of death globally and brought heavy social burdens. The complexity and diversity of the pathogenic factors add difficulties to diagnosis and treatment, as well as lead to poor prognosis of these diseases. MicroRNAs are short non-coding RNAs to modulate gene expression through directly binding to the 3′-untranslated regions of mRNAs of target genes and thereby to downregulate the protein levels post-transcriptionally. The multiple regulatory effects of microRNAs have been investigated extensively in cardiovascular diseases. MiR-223-3p, expressed in multiple cells such as macrophages, platelets, hepatocytes, and cardiomyocytes to modulate their cellular activities through targeting a variety of genes, is involved in the pathological progression of many cardiovascular diseases. It participates in regulation of several crucial signaling pathways such as phosphatidylinositol 3-kinase/protein kinase B, insulin-like growth factor 1, nuclear factor kappa B, mitogen-activated protein kinase, NOD-like receptor family pyrin domain containing 3 inflammasome, and ribosomal protein S6 kinase B1/hypoxia inducible factor 1 α pathways to affect cell proliferation, migration, apoptosis, hypertrophy, and polarization, as well as electrophysiology, resulting in dysfunction of cardiovascular system. Here, in this review, we will discuss the role of miR-223-3p in cardiovascular diseases, involving its verified targets, influenced signaling pathways, and regulation of cell function. In addition, the potential of miR-223-3p as therapeutic target and biomarker for diagnosis and prediction of cardiovascular diseases will be further discussed, providing clues for clinicians.
Collapse
Affiliation(s)
- Meng-Wan Zhang
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yun-Jie Shen
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Shi
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jian-Guang Yu
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
23
|
De Luca G, Campochiaro C, Sartorelli S, Peretto G, Dagna L. Therapeutic strategies for virus-negative myocarditis: a comprehensive review. Eur J Intern Med 2020; 77:9-17. [PMID: 32402564 DOI: 10.1016/j.ejim.2020.04.050] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 12/19/2022]
Abstract
Virus-negative or autoimmune myocarditis(VNM) is an inflammatory disease affecting the myocardium that may occur as a distinct disease with exclusive cardiac involvement, or in the context of systemic autoimmune or inflammatory disorders. The pathogenesis of VNM involves both innate and acquired immunity and is not completely elucidated: an early immune-mediated pathogenic process lead to subacute and chronic stages and eventually results in tissue remodeling, fibrosis, contractile dysfunction, dilated cardiomyopathy and arrhythmic burden, accounting for a dismal prognosis. Treatment interventions effectively curbing the acute inflammatory process at an early stage can prevent late cardiac remodeling and improve patient's outcome. The mainstay of treatment of VNM remains symptomatic therapy of heart failure and arrhythmia, while the use of immunosuppressive treatments has long been considered controversial until recently, and strategies effectively targeting the inflammatory and immune-mediated substrate of the disease remain elusive. Only steroids and azathioprine have been tested in clinical trials, and nowadays represent the therapy of choice. A substantial proportion of patients are resistant to first line strategies, suggesting that some critical inflammatory mechanisms are not responsive to conventional immunosuppression with steroids and azathioprine, or experience drug-related adverse events. Thus, second-line targeted therapeutic strategies to treat VNM are eagerly awaited. Recent data on the pathogenic mechanisms underlying myocardial inflammation are paving the way to novel, promising treatment strategies for myocarditis, which could reformulate future treatment strategies for VNM. In this review, we summarize the current therapeutic opportunities, beyond corticosteroids, to treat VNM, including conventional and biologic immunosuppressive drugs and cytokine blocking agents.
Collapse
Affiliation(s)
- Giacomo De Luca
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Via Olgettina, 60-20132, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy.
| | - Corrado Campochiaro
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Via Olgettina, 60-20132, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Silvia Sartorelli
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Via Olgettina, 60-20132, Milan, Italy
| | - Giovanni Peretto
- Department of Cardiac Electrophysiology and Arrhythmology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Via Olgettina, 60-20132, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
24
|
Wagner MJ, Khan M, Mohsin S. Healing the Broken Heart; The Immunomodulatory Effects of Stem Cell Therapy. Front Immunol 2020; 11:639. [PMID: 32328072 PMCID: PMC7160320 DOI: 10.3389/fimmu.2020.00639] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/20/2020] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular Disease (CVD) is a leading cause of mortality within the United States. Current treatments being administered to patients who suffered a myocardial infarction (MI) have increased patient survival, but do not facilitate the replacement of damaged myocardium. Recent studies demonstrate that stem cell-based therapies promote myocardial repair; however, the poor engraftment of the transferred stem cell populations within the infarcted myocardium is a major limitation, regardless of the cell type. One explanation for poor cell retention is attributed to the harsh inflammatory response mounted following MI. The inflammatory response coupled to cardiac repair processes is divided into two distinct phases. The first phase is initiated during ischemic injury when necrosed myocardium releases Danger Associated Molecular Patterns (DAMPs) and chemokines/cytokines to induce the activation and recruitment of neutrophils and pro-inflammatory M1 macrophages (MΦs); in turn, facilitating necrotic tissue clearance. During the second phase, a shift from the M1 inflammatory functional phenotype to the M2 anti-inflammatory and pro-reparative functional phenotype, permits the resolution of inflammation and the establishment of tissue repair. T-regulatory cells (Tregs) are also influential in mediating the establishment of the pro-reparative phase by directly regulating M1 to M2 MΦ differentiation. Current studies suggest CD4+ T-lymphocyte populations become activated when presented with autoantigens released from the injured myocardium. The identity of the cardiac autoantigens or paracrine signaling molecules released from the ischemic tissue that directly mediate the phenotypic plasticity of T-lymphocyte populations in the post-MI heart are just beginning to be elucidated. Stem cells are enriched centers that contain a diverse paracrine secretome that can directly regulate responses within neighboring cell populations. Previous studies identify that stem cell mediated paracrine signaling can influence the phenotype and function of immune cell populations in vitro, but how stem cells directly mediate the inflammatory microenvironment of the ischemic heart is poorly characterized and is a topic of extensive investigation. In this review, we summarize the complex literature that details the inflammatory microenvironment of the ischemic heart and provide novel insights regarding how paracrine mediated signaling produced by stem cell-based therapies can regulate immune cell subsets to facilitate pro-reparative myocardial wound healing.
Collapse
Affiliation(s)
- Marcus J Wagner
- Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Mohsin Khan
- Center for Metabolic Disease, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Sadia Mohsin
- Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| |
Collapse
|
25
|
Tong R, Jia T, Shi R, Yan F. Inhibition of microRNA-15 protects H9c2 cells against CVB3-induced myocardial injury by targeting NLRX1 to regulate the NLRP3 inflammasome. Cell Mol Biol Lett 2020; 25:6. [PMID: 32099552 PMCID: PMC7031959 DOI: 10.1186/s11658-020-00203-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 02/06/2020] [Indexed: 02/07/2023] Open
Abstract
Background Viral myocarditis (VMC) is a type of cardiac inflammation that is generally caused by coxsackievirus B3 (CVB3) infection. Several MicroRNAs (miRNAs) are known to play crucial roles in VMC pathogenesis. MiR-15 is reportedly associated with myocardial injury, inflammatory responses and viral infection. Whether miR-15 affects the occurrence and development of VMC remains largely unknown. The roles of miR-15 and their underlying mechanisms in CVB3-stimulated H9c2 cells were assessed in this study. Methods We infected H9c2 cells with CVB3 to establish a VMC cellular model. We then determined the effects of miR-15 inhibition on three cardiomyocyte injury markers: lactate dehydrogenase (LDH), creatine kinase-MB (CK-MB) and cardiac troponin-I (cTn-I). The impact on CVB3-induced cell apoptosis and pro-inflammatory cytokines was also investigated. The effects of miR-15 inhibition on NLRP3 inflammasome activation were also assessed. The target relationship between miR-15 and NOD-like receptor X1 (NLRX1) was determined using a luciferase reporter assay. Results MiR-15 expression was significantly upregulated in H9c2 cells after CVB3 infection. Inhibition of miR-15 significantly decreased the CVB3-induced levels of LDH, CK-MB and cTn-I. It also elevated cell viability, reduced CVB3-induced cell apoptosis and decreased the generation of the interleukins IL-1β, IL-6 and IL-18. Furthermore, we determined that miR-15 inhibition suppressed NLRP3 inflammasome activation by downregulating NLRP3 and caspase-1 p20 expression. We found a direct target relationship between miR-15 and NLRX1. Additionally, inhibition of NLRX1 reversed the protective effects of miR-15 inhibition against CVB3-induced myocardial cell injury by regulating the NLRP3 inflammasome. Conclusion Our results indicate that miR-15 inhibition alleviates CVB3-induced myocardial inflammation and cell injury. This may be partially due to NLRX1-mediated NLRP3 inflammasome inactivation.
Collapse
Affiliation(s)
- Ru Tong
- 1Laboratory Dept., Second Hospital of Shanxi Medical University, Taiyuan, 030001 Shanxi China
| | - Tiewen Jia
- 1Laboratory Dept., Second Hospital of Shanxi Medical University, Taiyuan, 030001 Shanxi China
| | - Ruijie Shi
- 2Laboratory Dept., Shaanxi Provincial People's Hospital, No. 256, West Youyi Road, Xi'an, 710068 Shaanxi province China
| | - Futang Yan
- 2Laboratory Dept., Shaanxi Provincial People's Hospital, No. 256, West Youyi Road, Xi'an, 710068 Shaanxi province China
| |
Collapse
|
26
|
Shao M, Wang D, Zhou Y, Du K, Liu W. Interleukin-10 delivered by mesenchymal stem cells attenuates experimental autoimmune myocarditis. Int Immunopharmacol 2020; 81:106212. [PMID: 32062070 DOI: 10.1016/j.intimp.2020.106212] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/08/2020] [Accepted: 01/08/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUNDS Autoimmune myocarditis is characterized by over-activated immune system attacking the cardiomyocytes, resulting in heart function decline. In the current study, we investigated the therapeutic advantages of delivering Interleukin-10 (IL-10) by mesenchymal stem cells (MSCs), both of which had immune suppression functions, in treating experimental autoimmune myocarditis. METHODS The mouse model of autoimmune myocarditis was established by subcutaneous injection of troponin I in A/J mice. Mouse bone marrow derived mesenchymal stem cells (BM-MSCs) with or without IL-10 overexpression, or the recombinant IL-10 protein were delivered into the mice via tail-vein injection. The inflammation and fibrosis levels of the heart were evaluated with qPCR, ELISA and histological staining. Serum level of anti-troponin-I was assessed by ELISA. Heart function analysis was conducted with echocardiography. RESULTS BM-MSCs overexpressing IL-10 had enhanced immune suppression functions. They also showed improved therapeutic effects from the perspective of heart function and cardiac fibrosis. The anti-troponin-I level was significantly reduced by MSCs overexpressing IL-10 when comparing with the MSCs or IL-10 protein injection. CONCLUSION IL-10 delivered by MSCs showed therapeutic advantages in treating experimental autoimmune myocarditis.
Collapse
Affiliation(s)
- Minkun Shao
- Department of Newborn, Shangqiu First People's Hospital, Shangqiu 476100, Henan, China
| | - Dong Wang
- Department of Newborn, Shangqiu First People's Hospital, Shangqiu 476100, Henan, China
| | - Yan Zhou
- Department of Newborn, Shangqiu First People's Hospital, Shangqiu 476100, Henan, China
| | - Kun Du
- Department of Newborn, Shangqiu First People's Hospital, Shangqiu 476100, Henan, China
| | - Wei Liu
- Department of Newborn, Shangqiu First People's Hospital, Shangqiu 476100, Henan, China.
| |
Collapse
|
27
|
Kociol RD, Cooper LT, Fang JC, Moslehi JJ, Pang PS, Sabe MA, Shah RV, Sims DB, Thiene G, Vardeny O. Recognition and Initial Management of Fulminant Myocarditis: A Scientific Statement From the American Heart Association. Circulation 2020; 141:e69-e92. [PMID: 31902242 DOI: 10.1161/cir.0000000000000745] [Citation(s) in RCA: 375] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Fulminant myocarditis (FM) is an uncommon syndrome characterized by sudden and severe diffuse cardiac inflammation often leading to death resulting from cardiogenic shock, ventricular arrhythmias, or multiorgan system failure. Historically, FM was almost exclusively diagnosed at autopsy. By definition, all patients with FM will need some form of inotropic or mechanical circulatory support to maintain end-organ perfusion until transplantation or recovery. Specific subtypes of FM may respond to immunomodulatory therapy in addition to guideline-directed medical care. Despite the increasing availability of circulatory support, orthotopic heart transplantation, and disease-specific treatments, patients with FM experience significant morbidity and mortality as a result of a delay in diagnosis and initiation of circulatory support and lack of appropriately trained specialists to manage the condition. This scientific statement outlines the resources necessary to manage the spectrum of FM, including extracorporeal life support, percutaneous and durable ventricular assist devices, transplantation capabilities, and specialists in advanced heart failure, cardiothoracic surgery, cardiac pathology, immunology, and infectious disease. Education of frontline providers who are most likely to encounter FM first is essential to increase timely access to appropriately resourced facilities, to prevent multiorgan system failure, and to tailor disease-specific therapy as early as possible in the disease process.
Collapse
|
28
|
Zarak-Crnkovic M, Kania G, Jaźwa-Kusior A, Czepiel M, Wijnen WJ, Czyż J, Müller-Edenborn B, Vdovenko D, Lindner D, Gil-Cruz C, Bachmann M, Westermann D, Ludewig B, Distler O, Lüscher TF, Klingel K, Eriksson U, Błyszczuk P. Heart non-specific effector CD4 + T cells protect from postinflammatory fibrosis and cardiac dysfunction in experimental autoimmune myocarditis. Basic Res Cardiol 2019; 115:6. [PMID: 31863205 PMCID: PMC6925074 DOI: 10.1007/s00395-019-0766-6] [Citation(s) in RCA: 18] [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: 04/01/2019] [Accepted: 12/04/2019] [Indexed: 12/14/2022]
Abstract
Heart-specific CD4+ T cells have been implicated in development and progression of myocarditis in mice and in humans. Here, using mouse models of experimental autoimmune myocarditis (EAM) we investigated the role of heart non-specific CD4+ T cells in the progression of the disease. Heart non-specific CD4+ T cells were obtained from DO11.10 mice expressing transgenic T cell receptor recognizing chicken ovalbumin. We found that heart infiltrating CD4+ T cells expressed exclusively effector (Teff) phenotype in the EAM model and in hearts of patients with lymphocytic myocarditis. Adoptive transfer experiments showed that while heart-specific Teff infiltrated the heart shortly after injection, heart non-specific Teff effectively accumulated during myocarditis and became the major heart-infiltrating CD4+ T cell subset at later stage. Restimulation of co-cultured heart-specific and heart non-specific CD4+ T cells with alpha-myosin heavy chain antigen showed mainly Th1/Th17 response for heart-specific Teff and up-regulation of a distinct set of extracellular signalling molecules in heart non-specific Teff. Adoptive transfer of heart non-specific Teff in mice with myocarditis did not affect inflammation severity at the peak of disease, but protected the heart from adverse post-inflammatory fibrotic remodelling and cardiac dysfunction at later stages of disease. Furthermore, mouse and human Teff stimulated in vitro with common gamma cytokines suppressed expression of profibrotic genes, reduced amount of α-smooth muscle actin filaments and decreased contraction of cardiac fibroblasts. In this study, we provided a proof-of-concept that heart non-specific Teff cells could effectively contribute to myocarditis and protect the heart from the dilated cardiomyopathy outcome.
Collapse
Affiliation(s)
- Martina Zarak-Crnkovic
- Cardioimmunology, Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Gabriela Kania
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | | | - Marcin Czepiel
- Department of Clinical Immunology, Jagiellonian University Medical College, University Children's Hospital, Wielicka 265, 30-663, Cracow, Poland
| | - Winandus J Wijnen
- Cardioimmunology, Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Jarosław Czyż
- Department of Cell Biology, Jagiellonian University, Cracow, Poland
| | - Björn Müller-Edenborn
- Cardioimmunology, Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
- Department of Medicine, GZO-Zurich Regional Health Center, Wetzikon, Switzerland
| | - Daria Vdovenko
- Cardioimmunology, Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Diana Lindner
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, Hamburg, Germany
| | - Cristina Gil-Cruz
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Marta Bachmann
- Cardioimmunology, Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Dirk Westermann
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, Hamburg, Germany
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Oliver Distler
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Thomas F Lüscher
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | - Karin Klingel
- Cardiopathology, Institute for Pathology and Neuropathology, University of Tubingen, Tubingen, Germany
| | - Urs Eriksson
- Cardioimmunology, Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
- Department of Medicine, GZO-Zurich Regional Health Center, Wetzikon, Switzerland
| | - Przemysław Błyszczuk
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, Zurich, Switzerland.
- Department of Clinical Immunology, Jagiellonian University Medical College, University Children's Hospital, Wielicka 265, 30-663, Cracow, Poland.
| |
Collapse
|
29
|
MicroRNA-223-3p modulates dendritic cell function and ameliorates experimental autoimmune myocarditis by targeting the NLRP3 inflammasome. Mol Immunol 2019; 117:73-83. [PMID: 31743855 DOI: 10.1016/j.molimm.2019.10.027] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 10/15/2019] [Accepted: 10/31/2019] [Indexed: 12/17/2022]
Abstract
Autoimmune myocarditis is a cause of dilated cardiomyopathy and heart failure. MicroRNAs regulate many immune processes, but their role in aberrant inflammation during autoimmune myocarditis remains unclear. In this study, we investigated the role of miR-223-3p in experimental autoimmune myocarditis (EAM). We found that miR-223-3p expression was significantly lower in EAM mice than that in normal mice. miR-223-3p inhibited NLRP3 inflammasome expression, promoting the polarization of dendritic cells (DCs) towards a tolerogenic DC phenotype. miR-223-3p effectively induced regulatory T cell (Treg) generation by inhibiting the function of antigen-presenting DCs. Transfer of miR-223-3p-overexpressing DCs protected mice against the development of EAM. Our findings suggest that miR-223-3p is involved in the induction of the tolerogenic DC phenotype and regulates tolerance in autoimmune myocarditis.
Collapse
|
30
|
The spectrum of myocarditis: from pathology to the clinics. Virchows Arch 2019; 475:279-301. [DOI: 10.1007/s00428-019-02615-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 06/15/2019] [Accepted: 06/20/2019] [Indexed: 12/14/2022]
|
31
|
Błyszczuk P. Myocarditis in Humans and in Experimental Animal Models. Front Cardiovasc Med 2019; 6:64. [PMID: 31157241 PMCID: PMC6532015 DOI: 10.3389/fcvm.2019.00064] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 04/30/2019] [Indexed: 12/21/2022] Open
Abstract
Myocarditis is defined as an inflammation of the cardiac muscle. In humans, various infectious and non-infectious triggers induce myocarditis with a broad spectrum of histological presentations and clinical symptoms of the disease. Myocarditis often resolves spontaneously, but some patients develop heart failure and require organ transplantation. The need to understand cellular and molecular mechanisms of inflammatory heart diseases led to the development of mouse models for experimental myocarditis. It has been shown that pathogenic agents inducing myocarditis in humans can often trigger the disease in mice. Due to multiple etiologies of inflammatory heart diseases in humans, a number of different experimental approaches have been developed to induce myocarditis in mice. Accordingly, experimental myocarditis in mice can be induced by infection with cardiotropic agents, such as coxsackievirus B3 and protozoan parasite Trypanosoma cruzi or by activating autoimmune responses against heart-specific antigens. In certain models, myocarditis is followed by the phenotype of dilated cardiomyopathy and the end stage of heart failure. This review describes the most commonly used mouse models of experimental myocarditis with a focus on the role of the innate and adaptive immune systems in induction and progression of the disease. The review discusses also advantages and limitations of individual mouse models in the context of the clinical manifestation and the course of the disease in humans. Finally, animal-free alternatives in myocarditis research are outlined.
Collapse
Affiliation(s)
- Przemysław Błyszczuk
- Department of Clinical Immunology, Jagiellonian University Medical College, Cracow, Poland.,Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
32
|
Lijuan S, Shu L, Yonghua Z, Qingmin X, Chungang Z, Lan L. Effect of Shenqi Yangxin decoction on high mobility group box 1 and inflammatory signal pathway in a rat model of dilated cardiomyopathy. J TRADIT CHIN MED 2018. [DOI: 10.1016/s0254-6272(18)30985-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
33
|
Beling A, Kespohl M. Proteasomal Protein Degradation: Adaptation of Cellular Proteolysis With Impact on Virus-and Cytokine-Mediated Damage of Heart Tissue During Myocarditis. Front Immunol 2018; 9:2620. [PMID: 30546359 PMCID: PMC6279938 DOI: 10.3389/fimmu.2018.02620] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 10/24/2018] [Indexed: 12/26/2022] Open
Abstract
Viral myocarditis is an inflammation of the heart muscle triggered by direct virus-induced cytolysis and immune response mechanisms with most severe consequences during early childhood. Acute and long-term manifestation of damaged heart tissue and disturbances of cardiac performance involve virus-triggered adverse activation of the immune response and both immunopathology, as well as, autoimmunity account for such immune-destructive processes. It is a matter of ongoing debate to what extent subclinical virus infection contributes to the debilitating sequela of the acute disease. In this review, we conceptualize the many functions of the proteasome in viral myocarditis and discuss the adaptation of this multi-catalytic protease complex together with its implications on the course of disease. Inhibition of proteasome function is already highly relevant as a strategy in treating various malignancies. However, cardiotoxicity and immune-related adverse effects have proven significant hurdles, representative of the target's wide-ranging functions. Thus, we further discuss the molecular details of proteasome-mediated activity of the immune response for virus-mediated inflammatory heart disease. We summarize how the spatiotemporal flexibility of the proteasome might be tackled for therapeutic purposes aiming to mitigate virus-mediated adverse activation of the immune response in the heart.
Collapse
Affiliation(s)
- Antje Beling
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Biochemistry, Berlin, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Partner Site Berlin, Berlin, Germany
| | - Meike Kespohl
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Biochemistry, Berlin, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Partner Site Berlin, Berlin, Germany
| |
Collapse
|
34
|
Meyer IS, Goetzke CC, Kespohl M, Sauter M, Heuser A, Eckstein V, Vornlocher HP, Anderson DG, Haas J, Meder B, Katus HA, Klingel K, Beling A, Leuschner F. Silencing the CSF-1 Axis Using Nanoparticle Encapsulated siRNA Mitigates Viral and Autoimmune Myocarditis. Front Immunol 2018; 9:2303. [PMID: 30349538 PMCID: PMC6186826 DOI: 10.3389/fimmu.2018.02303] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 09/17/2018] [Indexed: 12/11/2022] Open
Abstract
Myocarditis is an inflammatory disease of the heart muscle most commonly caused by viral infection and often maintained by autoimmunity. Virus-induced tissue damage triggers chemokine production and, subsequently, immune cell infiltration with pro-inflammatory and pro-fibrotic cytokine production follows. In patients, the overall inflammatory burden determines the disease outcome. Following the aim to define specific molecules that drive both immunopathology and/or autoimmunity in inflammatory heart disease, here we report on increased expression of colony stimulating factor 1 (CSF-1) in patients with myocarditis. CSF-1 controls monocytes originating from hematopoietic stem cells and subsequent progenitor stages. Both, monocytes and macrophages are centrally involved in mediating tissue damage and fibrotic scarring in the heart. CSF-1 influences monocytes via engagement of CSF-1 receptor, and it is also produced by cells of the mononuclear phagocyte system themselves. Based on this, we sought to modulate the virus-triggered inflammatory response in an experimental model of Coxsackievirus B3-induced myocarditis by silencing the CSF-1 axis in myeloid cells using nanoparticle-encapsulated siRNA. siCSF-1 inverted virus-mediated immunopathology as reflected by lower troponin T levels, a reduction of accumulating myeloid cells in heart tissue and improved cardiac function. Importantly, pathogen control was maintained and the virus was efficiently cleared from heart tissue. Since viral heart disease triggers heart-directed autoimmunity, in a second approach we investigated the influence of CSF-1 upon manifestation of heart tissue inflammation during experimental autoimmune myocarditis (EAM). EAM was induced in Balb/c mice by immunization with a myocarditogenic myosin-heavy chain-derived peptide dissolved in complete Freund's adjuvant. siCSF-1 treatment initiated upon established disease inhibited monocyte infiltration into heart tissue and this suppressed cardiac injury as reflected by diminished cardiac fibrosis and improved cardiac function at later states. Mechanistically, we found that suppression of CSF-1 production arrested both differentiation and maturation of monocytes and their precursors in the bone marrow. In conclusion, during viral and autoimmune myocarditis silencing of the myeloid CSF-1 axis by nanoparticle-encapsulated siRNA is beneficial for preventing inflammatory tissue damage in the heart and preserving cardiac function without compromising innate immunity's critical defense mechanisms.
Collapse
Affiliation(s)
- Ingmar Sören Meyer
- Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Heidelberg, Germany
| | - Carl Christoph Goetzke
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Meike Kespohl
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Martina Sauter
- Cardiopathology, Institute for Pathology and Neuropathology, University Hospital Tuebingen, Tuebingen, Germany
| | - Arnd Heuser
- Max-Delbrueck-Center for Molecular Medicine Berlin, Berlin, Germany
| | - Volker Eckstein
- Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States.,Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States.,Department of Chemical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, United States
| | - Jan Haas
- Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Heidelberg, Germany
| | - Benjamin Meder
- Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Heidelberg, Germany
| | - Hugo Albert Katus
- Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Heidelberg, Germany
| | - Karin Klingel
- Cardiopathology, Institute for Pathology and Neuropathology, University Hospital Tuebingen, Tuebingen, Germany
| | - Antje Beling
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Florian Leuschner
- Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Heidelberg, Germany
| |
Collapse
|
35
|
De Luca G, Cavalli G, Campochiaro C, Tresoldi M, Dagna L. Myocarditis: An Interleukin-1-Mediated Disease? Front Immunol 2018; 9:1335. [PMID: 29951067 PMCID: PMC6008311 DOI: 10.3389/fimmu.2018.01335] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 05/29/2018] [Indexed: 12/20/2022] Open
Affiliation(s)
- Giacomo De Luca
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Giulio Cavalli
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy.,Department of Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Corrado Campochiaro
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Moreno Tresoldi
- Unit of General Medicine and Advanced Care, IRCCS San Raffaele Hospital and Scientific Institute, Milan, Italy
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
36
|
Approximation of the Incidence of Myocarditis by Systematic Screening With Cardiac Magnetic Resonance Imaging. JACC-HEART FAILURE 2018; 6:573-579. [PMID: 29885953 DOI: 10.1016/j.jchf.2018.03.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 02/27/2018] [Accepted: 03/07/2018] [Indexed: 01/02/2023]
Abstract
OBJECTIVES This study sought to obtain an approximation of the true incidence of myocarditis by systematic screening of patients at risk using cardiac magnetic resonance imaging (CMR) in a tertiary care center. BACKGROUND Underdiagnosis of myocarditis and resulting uncertainty about its incidence remain a clinical dilemma. The authors hypothesized that systematic screening of patients presenting with angina-like symptoms, elevated troponin T, and no significant coronary artery disease using CMR will provide an approximation of the true incidence of myocarditis. METHODS The authors performed a retrospective chart review of patients presenting with angina-like symptoms and elevated high-sensitivity troponin T (TnT-hs ≥14 ng/l) in 2015 and 2016. During the year 2015, only patients with elevated TnT-hs, no significant coronary artery disease, and moderate-to-high clinical likelihood of myocarditis underwent CMR. Starting in 2016, CMR was obtained in patients with similar presentation, but independent of clinical likelihood of myocarditis. RESULTS A total of 1,788 patients (74% male, age 69 ± 14 years) qualified for our analysis. In 2015, 521 patients presented with angina-like symptoms and TnT-hs elevation. In 2016, the number increased to 1,267 patients. Although in the year 2015, a total of 4 of 88 (5%) CMRs were positive for myocarditis, the percentage of positive CMRs doubled (26 of 199; 13%; p = 0.03) in 2016. CONCLUSIONS A novel diagnostic screening algorithm led to a 6.3-fold increase of the incidence of myocarditis in our hospital. Furthermore, the percentage of CMRs positive for myocarditis doubled, supporting the diagnostic value of this method. Considering the potentially lethal adverse events of myocarditis if left untreated, we recommend a low threshold for the use of CMR in patients with angina-like symptoms and elevated TnT-hs after exclusion of coronary artery disease.
Collapse
|
37
|
Roles of Host Immunity in Viral Myocarditis and Dilated Cardiomyopathy. J Immunol Res 2018; 2018:5301548. [PMID: 29854842 PMCID: PMC5964556 DOI: 10.1155/2018/5301548] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 04/09/2018] [Accepted: 04/19/2018] [Indexed: 02/07/2023] Open
Abstract
The pathogenesis of viral myocarditis includes both the direct damage mediated by viral infection and the indirect lesion resulted from host immune responses. Myocarditis can progress into dilated cardiomyopathy that is also associated with immunopathogenesis. T cell-mediated autoimmunity, antibody-mediated autoimmunity (autoantibodies), and innate immunity, working together, contribute to the development of myocarditis and dilated cardiomyopathy.
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW In this paper we will review the modern diagnostic approach to patients with clinically suspected myocarditis as well as the treatment modalities and strategy in light of up-to-date clinical experience and scientific evidence. RECENT FINDINGS Rapidly expanding evidence suggests that myocardial inflammation is frequently underdiagnosed or overlooked in clinical practice, although new therapeutic options have been validated. Moreover, the available evidence suggests that subclinical cardiac involvement has negative prognostic impact on morbidity and mortality and should be actively investigated and adequately treated. Myocarditis represents a growing challenge for physicians, due to increased referral of patients for endomyocardial biopsy (EMB) or cardiac magnetic resonance (CMR), and requires a highly integrated management by a team of caring physicians.
Collapse
|
39
|
Lu H, Zong G, Zhou S, Jiang Y, Chen R, Su Z, Wu Y. Angiotensin II-C-C chemokine receptor2/5 axis-dependent monocyte/macrophage recruitment contributes to progression of experimental autoimmune myocarditis. Microbiol Immunol 2017; 61:539-546. [PMID: 29052263 DOI: 10.1111/1348-0421.12548] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 09/12/2017] [Accepted: 10/11/2017] [Indexed: 01/20/2023]
Abstract
Angiotensin II (ANG II) plays critical roles in modulation of circulatory homeostasis and activation of innate and adaptive immunity and has also been implicated in several mouse models of autoimmune disease. However, how ANG II regulates macrophages and is involved in development of experimental autoimmune myocarditis (EAM) remains unclear. Therefore, the present study aimed to address the above question and explore possible mechanisms. EAM was induced in BALB/c mice. ANG II was quantitated by ELISA and hematoxylin and eosin staining was employed to analyze pathological changes and macrophage infiltration. The chemotactic ability of ANG II was assessed by using a Transwell system. It was found that ANG II is up-regulated in serum and heart tissues of mice with EAM and that ANG II significantly drives monocyte/macrophage infiltration through the C-C chemokine receptor 2/5 (CCR2/5) axis. CCR2/5 antagonists and ANG II receptor inhibitor could all abrogate monocyte/macrophage infiltration and ameliorate development of EAM. Our results have firstly identified a novel function of ANG II: that it is a critical chemokine for monocyte/macrophage recruitment. Furthermore, our results indicate that ANG II is a potential candidate for treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Hongxiang Lu
- Central Laboratory, Fourth Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.,Department of Immunology, Jiangsu University, Zhenjiang 212013, China
| | - Gangjun Zong
- Department of Cardiology, Wuxi Clinical Hospital, Anhui Medical University, Wuxi 214000, China.,Department of Cardiology, 101 Hospital of PLA, Wuxi 214000, China
| | - Shanshan Zhou
- Department of Immunology, Jiangsu University, Zhenjiang 212013, China
| | - Yuanyuan Jiang
- Department of Immunology, Jiangsu University, Zhenjiang 212013, China
| | - Rong Chen
- Department of Immunology, Jiangsu University, Zhenjiang 212013, China
| | - Zhaoliang Su
- Central Laboratory, Fourth Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.,Department of Immunology, Jiangsu University, Zhenjiang 212013, China
| | - Yan Wu
- Central Laboratory, Fourth Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.,Department of Physiology, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, China
| |
Collapse
|
40
|
Shintaku M, Uchiyama K, Kobayashi Y. Chronic myocarditis with a long clinical course: Report of an autopsy case of probable autoimmune myocarditis. Pathol Int 2017; 67:521-525. [DOI: 10.1111/pin.12568] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 07/18/2017] [Indexed: 11/29/2022]
Affiliation(s)
- Masayuki Shintaku
- Department of Pathology; Shiga Medical Center for Adults; Moriyama Shiga Japan
| | | | - Yohei Kobayashi
- Department of Cardiology; Osaka Red Cross Hospital; Osaka Japan
| |
Collapse
|
41
|
Ludwig RJ, Vanhoorelbeke K, Leypoldt F, Kaya Z, Bieber K, McLachlan SM, Komorowski L, Luo J, Cabral-Marques O, Hammers CM, Lindstrom JM, Lamprecht P, Fischer A, Riemekasten G, Tersteeg C, Sondermann P, Rapoport B, Wandinger KP, Probst C, El Beidaq A, Schmidt E, Verkman A, Manz RA, Nimmerjahn F. Mechanisms of Autoantibody-Induced Pathology. Front Immunol 2017; 8:603. [PMID: 28620373 PMCID: PMC5449453 DOI: 10.3389/fimmu.2017.00603] [Citation(s) in RCA: 336] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/08/2017] [Indexed: 12/22/2022] Open
Abstract
Autoantibodies are frequently observed in healthy individuals. In a minority of these individuals, they lead to manifestation of autoimmune diseases, such as rheumatoid arthritis or Graves' disease. Overall, more than 2.5% of the population is affected by autoantibody-driven autoimmune disease. Pathways leading to autoantibody-induced pathology greatly differ among different diseases, and autoantibodies directed against the same antigen, depending on the targeted epitope, can have diverse effects. To foster knowledge in autoantibody-induced pathology and to encourage development of urgently needed novel therapeutic strategies, we here categorized autoantibodies according to their effects. According to our algorithm, autoantibodies can be classified into the following categories: (1) mimic receptor stimulation, (2) blocking of neural transmission, (3) induction of altered signaling, triggering uncontrolled (4) microthrombosis, (5) cell lysis, (6) neutrophil activation, and (7) induction of inflammation. These mechanisms in relation to disease, as well as principles of autoantibody generation and detection, are reviewed herein.
Collapse
Affiliation(s)
- Ralf J. Ludwig
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Karen Vanhoorelbeke
- Laboratory for Thrombosis Research, IRF Life Sciences, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Frank Leypoldt
- Neuroimmunology, Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Kiel, Germany
- Neuroimmunology, Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Lübeck, Germany
- Department of Neurology, University of Kiel, Kiel, Germany
| | - Ziya Kaya
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Katja Bieber
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Sandra M. McLachlan
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Medical Center, UCLA School of Medicine, Los Angeles, CA, United States
| | - Lars Komorowski
- Institute for Experimental Immunology, Affiliated to Euroimmun AG, Lübeck, Germany
| | - Jie Luo
- Department of Neuroscience, University of Pennsylvania Medical School, Philadelphia, PA, United States
| | | | | | - Jon M. Lindstrom
- Department of Neuroscience, University of Pennsylvania Medical School, Philadelphia, PA, United States
| | - Peter Lamprecht
- Department of Rheumatology, University of Lübeck, Lübeck, Germany
| | - Andrea Fischer
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany
| | | | - Claudia Tersteeg
- Laboratory for Thrombosis Research, IRF Life Sciences, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | | | - Basil Rapoport
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Medical Center, UCLA School of Medicine, Los Angeles, CA, United States
| | - Klaus-Peter Wandinger
- Department of Neurology, Institute of Clinical Chemistry, University Medical-Centre Schleswig-Holstein, Lübeck, Germany
| | - Christian Probst
- Institute for Experimental Immunology, Affiliated to Euroimmun AG, Lübeck, Germany
| | - Asmaa El Beidaq
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Enno Schmidt
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Alan Verkman
- Department of Medicine, University of California, San Francisco, CA, United States
- Department of Physiology, University of California, San Francisco, CA, United States
| | - Rudolf A. Manz
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Falk Nimmerjahn
- Department of Biology, Institute of Genetics, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
42
|
Cardiac Function Remains Impaired Despite Reversible Cardiac Remodeling after Acute Experimental Viral Myocarditis. J Immunol Res 2017; 2017:6590609. [PMID: 28352641 PMCID: PMC5352897 DOI: 10.1155/2017/6590609] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 12/01/2016] [Accepted: 12/15/2016] [Indexed: 12/15/2022] Open
Abstract
Background. Infection with Coxsackievirus B3 induces myocarditis. We aimed to compare the acute and chronic phases of viral myocarditis to identify the immediate effects of cardiac inflammation as well as the long-term effects after resolved inflammation on cardiac fibrosis and consequently on cardiac function. Material and Methods. We infected C57BL/6J mice with Coxsackievirus B3 and determined the hemodynamic function 7 as well as 28 days after infection. Subsequently, we analyzed viral burden and viral replication in the cardiac tissue as well as the expression of cytokines and matrix proteins. Furthermore, cardiac fibroblasts were infected with virus to investigate if viral infection alone induces profibrotic signaling. Results. Severe cardiac inflammation was determined and cardiac fibrosis was consistently colocalized with inflammation during the acute phase of myocarditis. Declined cardiac inflammation but no significantly improved hemodynamic function was observed 28 days after infection. Interestingly, cardiac fibrosis declined to basal levels as well. Both cardiac inflammation and fibrosis were reversible, whereas the hemodynamic function remains impaired after healed viral myocarditis in C57BL/6J mice.
Collapse
|
43
|
Lymphocytes at the Heart of Wound Healing. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1003:225-250. [DOI: 10.1007/978-3-319-57613-8_11] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
44
|
Inflammatory Cardiomyopathy: A Current View on the Pathophysiology, Diagnosis, and Treatment. BIOMED RESEARCH INTERNATIONAL 2016; 2016:4087632. [PMID: 27382566 PMCID: PMC4921131 DOI: 10.1155/2016/4087632] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 03/20/2016] [Indexed: 12/31/2022]
Abstract
Inflammatory cardiomyopathy is defined as inflammation of the heart muscle associated with impaired function of the myocardium. In our region, its etiology is most often viral. Viral infection is a possible trigger of immune and autoimmune mechanisms which contributed to the damage of myocardial function. Myocarditis is considered the most common cause of dilated cardiomyopathy. Typical manifestation of this disease is heart failure, chest pain, or arrhythmias. The most important noninvasive diagnostic method is magnetic resonance imaging, but the gold standard of diagnostics is invasive examination, endomyocardial biopsy. In a significant proportion of cases with impaired left ventricular systolic function, recovery occurs spontaneously in several weeks and therefore it is appropriate to postpone critical therapeutic decisions about 3–6 months after start of the treatment. Therapy is based on standard heart failure treatment; immunosuppressive or antimicrobial treatment may be considered in some cases depending on the results of endomyocardial biopsy. If severe dysfunction of the left ventricle persists, device therapy may be needed.
Collapse
|
45
|
Liu ZL, Liu ZJ, Liu JP, Kwong JSW, Cochrane Heart Group. WITHDRAWN: Herbal medicines for viral myocarditis. Cochrane Database Syst Rev 2016; 2016:CD003711. [PMID: 27142939 PMCID: PMC10680413 DOI: 10.1002/14651858.cd003711.pub6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The editors of Cochrane Heart consider this title as low priority for the current portfolio of the Heart Group and therefore this title is not open to a new author team. The editorial group responsible for this previously published document have withdrawn it from publication.
Collapse
Affiliation(s)
- Zhao Lan Liu
- Beijing University of Chinese MedicineCentre for Evidence‐Based Chinese Medicine11 Bei San Huan Dong Lu, Chaoyang DistrictBeijingChina100029
| | - Zhi Jun Liu
- Beijing University of Chinese MedicineCentre for Evidence‐Based Chinese Medicine11 Bei San Huan Dong Lu, Chaoyang DistrictBeijingChina100029
| | - Jian Ping Liu
- Beijing University of Chinese MedicineCentre for Evidence‐Based Chinese Medicine11 Bei San Huan Dong Lu, Chaoyang DistrictBeijingChina100029
| | - Joey SW Kwong
- West China Hospital, Sichuan UniversityChinese Evidence‐Based Medicine CenterNo. 37, Guo Xue XiangChengduSichuanChina610041
| | | |
Collapse
|
46
|
Gagliardi MG, Fierabracci A, Pilati M, Chinali M, Bassano C, Saura F, Giovannoni I, Francalanci P. The Impact of Specific Viruses on Clinical Outcome in Children Presenting with Acute Heart Failure. Int J Mol Sci 2016; 17:486. [PMID: 27043551 PMCID: PMC4848942 DOI: 10.3390/ijms17040486] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 03/14/2016] [Accepted: 03/28/2016] [Indexed: 11/17/2022] Open
Abstract
The presence and type of viral genomes have been suggested as the main etiology for inflammatory dilated cardiomyopathy. Information on the clinical implication of this finding in a large population of children is lacking. We evaluated the prevalence, type, and clinical impact of specific viral genomes in endomyocardial biopsies (EMB) collected between 2001 and 2013 among 63 children admitted to our hospital for acute heart failure (median age 2.8 years). Viral genome was searched by polymerase chain reaction (PCR). Patients underwent a complete two-dimensional echocardiographic examination at hospital admission and at discharge and were followed-up for 10 years. Twenty-seven adverse events (7 deaths and 20 cardiac transplantations) occurred during the follow-up. Viral genome was amplified in 19/63 biopsies (35%); PVB19 was the most commonly isolated virus. Presence of specific viral genome was associated with a significant recovery in ejection fraction, compared to patients without viral evidence (p < 0.05). In Cox-regression analysis, higher survival rate was related to virus-positive biopsies (p < 0.05). When comparing long-term prognosis among different viral groups, a trend towards better prognosis was observed in the presence of isolated Parvovirus B19 (PVB19) (p = 0.07). In our series, presence of a virus-positive EMB (mainly PVB19) was associated with improvement over time in cardiac function and better long-term prognosis.
Collapse
Affiliation(s)
- Maria Giulia Gagliardi
- Medical and Surgical Department of Cardiology, Children's Hospital Bambino Gesù, Piazza S. Onofrio 4, 00165 Rome, Italy.
| | - Alessandra Fierabracci
- Immuno-Infectivology and Clinical Trials Area, Children's Hospital Bambino Gesù, Viale S. Paolo 15, 00146 Rome, Italy.
| | - Mara Pilati
- Medical and Surgical Department of Cardiology, Children's Hospital Bambino Gesù, Piazza S. Onofrio 4, 00165 Rome, Italy.
| | - Marcello Chinali
- Medical and Surgical Department of Cardiology, Children's Hospital Bambino Gesù, Piazza S. Onofrio 4, 00165 Rome, Italy.
| | - Carlo Bassano
- Operative Unit and Chair of Cardiac Surgery, Tor Vergata University, Viale Oxford 81, 00133 Rome, Italy.
| | - Francesca Saura
- Department of Laboratory Medicine, Children's Hospital Bambino Gesù, Piazza S. Onofrio 4, 00165 Rome, Italy.
| | - Isabella Giovannoni
- Department of Pathology, Children's Hospital Bambino Gesù, Piazza S. Onofrio 4, 00165 Rome, Italy.
| | - Paola Francalanci
- Department of Pathology, Children's Hospital Bambino Gesù, Piazza S. Onofrio 4, 00165 Rome, Italy.
| |
Collapse
|
47
|
|
48
|
Affiliation(s)
- Alida L P Caforio
- Cardiology, Department of Cardiological Thoracic and Vascular Sciences, University of Padua, Padua, Italy
| | - Renzo Marcolongo
- Clinical Immunology, Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Cristina Basso
- Cardiovascular Pathology, Department of Cardiological, Thoracic and Vascular Sciences, University of Padua, Padua, Italy
| | - Sabino Iliceto
- Cardiology, Department of Cardiological Thoracic and Vascular Sciences, University of Padua, Padua, Italy
| |
Collapse
|
49
|
Amoah BP, Yang H, Zhang P, Su Z, Xu H. Immunopathogenesis of Myocarditis: The Interplay Between Cardiac Fibroblast Cells, Dendritic Cells, Macrophages and CD4+T Cells. Scand J Immunol 2015; 82:1-9. [DOI: 10.1111/sji.12298] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Revised: 02/21/2015] [Accepted: 03/14/2015] [Indexed: 02/06/2023]
Affiliation(s)
- B. Prince Amoah
- Department of Immunology; School of Medical Science and Laboratory Medicine; Jiangsu University; Zhenjiang China
- Department of Biomedical and Forensic Sciences; School of Biological Sciences; University of Cape Coast; Cape Coast Ghana
| | - H. Yang
- Department of Immunology; School of Medical Science and Laboratory Medicine; Jiangsu University; Zhenjiang China
| | - P. Zhang
- Department of Immunology; School of Medical Science and Laboratory Medicine; Jiangsu University; Zhenjiang China
| | - Z. Su
- Department of Immunology; School of Medical Science and Laboratory Medicine; Jiangsu University; Zhenjiang China
- The Central Laboratory; The Fourth Affiliated Hospital of Jiangsu University; Zhenjiang China
| | - H. Xu
- Department of Immunology; School of Medical Science and Laboratory Medicine; Jiangsu University; Zhenjiang China
| |
Collapse
|
50
|
Aoyama Y, Kobayashi K, Morishita Y, Maeda K, Murohara T. Wnt11 gene therapy with adeno-associated virus 9 improves the survival of mice with myocarditis induced by coxsackievirus B3 through the suppression of the inflammatory reaction. J Mol Cell Cardiol 2015; 84:45-51. [PMID: 25886696 DOI: 10.1016/j.yjmcc.2015.04.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 04/03/2015] [Accepted: 04/09/2015] [Indexed: 01/09/2023]
Abstract
The wnt signaling pathway plays important roles in development and in many diseases. Recently several reports suggest that non-canonical Wnt proteins contribute to the inflammatory response in adult animals. However, the effects of Wnt proteins on virus-induced myocarditis have not been explored. Here, we investigated the effect of Wnt11 protein in a model of myocarditis induced by coxsackievirus B3 (CVB3) using recombinant adeno-associated virus 9 (rAAV9). The effect of Wnt11 gene therapy on a CVB3-induced myocarditis model was examined using male BALB/c mice. Mice received a single intravenous injection of either rAAV9-Wnt11 or rAAV9-LacZ 2 weeks before intraperitoneal administration of CVB3. Intravenous injection of the rAAV9 vector resulted in efficient, durable, and relatively cardiac-specific transgene expression. Survival was significantly greater among rAAV9-Wnt11 treated mice than among mice treated with rAAV9-LacZ (87.5% vs. 54.1%, P < 0.05). Wnt11 expression also reduced the infiltration of inflammatory cells, necrosis of the myocardium, and suppressed the mRNA expression of inflammatory cytokines. This is the first report to show that Wnt11 expression improves the survival of mice with CVB3-induced myocarditis. AAV9-mediated Wnt11 gene therapy produces beneficial effects on cardiac function and increases the survival of mice with CVB3-induced myocarditis through the suppression of both infiltration of inflammatory cells and gene expression of inflammatory cytokines.
Collapse
Affiliation(s)
- Yutaka Aoyama
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya 466, Japan
| | - Koichi Kobayashi
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya 466, Japan.
| | - Yoshihiro Morishita
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya 466, Japan
| | - Kengo Maeda
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya 466, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya 466, Japan
| |
Collapse
|