1
|
Tian F, Xiao Y, Peng Z, Zhang L, Ni F, Gui S, Fan Y, Xi Z, Zhang Z. Fulminant myocarditis caused by influenza B virus in a male child: a case report and literature review. J Cardiothorac Surg 2024; 19:492. [PMID: 39182151 PMCID: PMC11344422 DOI: 10.1186/s13019-024-02997-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 08/13/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Influenza B virus induced myocarditis is a rare complication with potentially wide variations in severity and clinical presentation, and the pathogenesis is unclear. CASE PRESENTATION We describe a rare case of a 7-year-old boy who developed fulminant myocarditis (FM) due to influenza B virus infection. Treatment measures included mechanical ventilation, vasoactive agents, Extracorporeal membrane oxygenation (ECMO), Continuous Renal Replacement Therapy (CRRT), anti-inflammatory, antiviral, anti-infection, and enteral nutrition support. After 10 days of treatment, the patient succumbed to multiorgan failure. CONCLUSIONS After a systematic review of the literature, we found that this disease predominantly affects females, with pediatric cases exceedingly rare. Fulminant myocarditis (FM) progresses rapidly, poses significant treatment challenges sporadic, and carries a poor prognosis. Interestingly, literature reports suggest that anti-thymocyte globulin therapy may have a positive impact in treating FM, potentially offering new insights into its pathogenesis and clinical management.
Collapse
Affiliation(s)
- Fei Tian
- Department of Critical Care Medicine, Yichang Central People's Hospital, The First College of Clinical Medical Science of China Three Gorges University, Yichang, China
- Center of Clinical Nursing Research, China Three Gorges University, Yichang, 443002, China
| | - Yi Xiao
- Department of Endocrinology, Yichang Central People's Hospital, The First College of Clinical Medical Science of China Three Gorges University, Yichang, China
| | - Zhekang Peng
- Department of Critical Care Medicine, Yichang Central People's Hospital, The First College of Clinical Medical Science of China Three Gorges University, Yichang, China
| | - Lingyun Zhang
- Department of Respiratory and Critical Care Medicine, Yichang Central People's Hospital, The First College of Clinical Medical Science of China Three Gorges University, Yichang, China
| | - Fu Ni
- Department of Nursing, Yichang Central People's Hospital, The First College of Clinical Medical Science of China Three Gorges University, Yichang, China
| | - Shengmin Gui
- Department of Nursing, Yichang Central People's Hospital, The First College of Clinical Medical Science of China Three Gorges University, Yichang, China
- Center of Clinical Nursing Research, China Three Gorges University, Yichang, 443002, China
| | - Yuqing Fan
- Department of Nursing, Yichang Central People's Hospital, The First College of Clinical Medical Science of China Three Gorges University, Yichang, China
| | - Zuyang Xi
- Department of Nursing, Yichang Central People's Hospital, The First College of Clinical Medical Science of China Three Gorges University, Yichang, China.
- Center of Clinical Nursing Research, China Three Gorges University, Yichang, 443002, China.
| | - Zhaohui Zhang
- Department of Critical Care Medicine, Yichang Central People's Hospital, The First College of Clinical Medical Science of China Three Gorges University, Yichang, China.
| |
Collapse
|
2
|
Won T, Song EJ, Kalinoski HM, Moslehi JJ, Čiháková D. Autoimmune Myocarditis, Old Dogs and New Tricks. Circ Res 2024; 134:1767-1790. [PMID: 38843292 DOI: 10.1161/circresaha.124.323816] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 05/08/2024] [Indexed: 06/12/2024]
Abstract
Autoimmunity significantly contributes to the pathogenesis of myocarditis, underscored by its increased frequency in autoimmune diseases such as systemic lupus erythematosus and polymyositis. Even in cases of myocarditis caused by viral infections, dysregulated immune responses contribute to pathogenesis. However, whether triggered by existing autoimmune conditions or viral infections, the precise antigens and immunologic pathways driving myocarditis remain incompletely understood. The emergence of myocarditis associated with immune checkpoint inhibitor therapy, commonly used for treating cancer, has afforded an opportunity to understand autoimmune mechanisms in myocarditis, with autoreactive T cells specific for cardiac myosin playing a pivotal role. Despite their self-antigen recognition, cardiac myosin-specific T cells can be present in healthy individuals due to bypassing the thymic selection stage. In recent studies, novel modalities in suppressing the activity of pathogenic T cells including cardiac myosin-specific T cells have proven effective in treating autoimmune myocarditis. This review offers an overview of the current understanding of heart antigens, autoantibodies, and immune cells as the autoimmune mechanisms underlying various forms of myocarditis, along with the latest updates on clinical management and prospects for future research.
Collapse
Affiliation(s)
- Taejoon Won
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois Urbana-Champaign (T.W.)
| | - Evelyn J Song
- Section of Cardio-Oncology and Immunology, Division of Cardiology and the Cardiovascular Research Institute, University of California San Francisco (E.J.S., J.J.M.)
| | - Hannah M Kalinoski
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD (H.M.K., D.Č)
| | - Javid J Moslehi
- Section of Cardio-Oncology and Immunology, Division of Cardiology and the Cardiovascular Research Institute, University of California San Francisco (E.J.S., J.J.M.)
| | - Daniela Čiháková
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD (H.M.K., D.Č)
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD (D.Č)
| |
Collapse
|
3
|
Xiong C, Li B, Song R, Ma Z, Huber SA, Liu W. IFITM3 mediates inflammation induced myocardial injury through JAK2/STAT3 signaling pathway. Mol Immunol 2024; 167:1-15. [PMID: 38306778 DOI: 10.1016/j.molimm.2024.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/11/2023] [Accepted: 01/23/2024] [Indexed: 02/04/2024]
Abstract
Myocarditis is an inflammation of the heart muscle often associated with viral infections and can lead to dilated cardiomyopathy. Interferon-induced transmembrane protein 3 (IFITM3) is a small endosomal membrane protein with anti-viral activity against multiple viruses and is also implicated in non-infectious diseases such as cancer and Alzheimer's Disease. Since the IFITM3 proteins are expressed both in T cells and in cardiomyocytes, it is reasonable to hypothesize that these molecules could affect myocarditis either through their effect on the autoimmune response or through direct modulation of cardiomyocyte damage. The aim of this study was to investigate the role of IFITM3 in experimental autoimmune myocarditis (EAM)-mediated myocardial injury. Immunization of rats with cardiac myosin results in substantial cardiac inflammation and is associated with increased expression of IFITM3 after 21 days. In vivo IFITM3 shRNA knockdown using the lentivirus transfection method reduced cardiac injury while restoring IFITM3 expression reversed the protective effect of IFITM3 RNA interference. To determine the direct impact of IFITM3, the rat ventricular cell line, H9c2, was treated with palmitic acid which causes apoptosis in these cells. Suppressing IFITM3 expression protects H9c2 cells while overexpressing IFITM3 enhances cell injury. JAK inhibitors reduced IFITM3-mediated myocardial cell injury. In conclusion, IFITM3 may mediate myocardial injury in EAM rats and palmitic acid-induced damage to H9c2 cells through the JAK2/STAT3 pathway.
Collapse
Affiliation(s)
- Chunming Xiong
- Department of Cardiology, the fourth affiliated hospital of Harbin Medical University, Harbin, Heilongjiang 150001 China
| | - Bohan Li
- Harbin Medical University, Harbin, Heilongjiang 150001 China
| | - Renxing Song
- Department of Cardiology, the fourth affiliated hospital of Harbin Medical University, Harbin, Heilongjiang 150001 China
| | - Zizhe Ma
- Department of Cardiology, the fourth affiliated hospital of Harbin Medical University, Harbin, Heilongjiang 150001 China
| | - Sally A Huber
- Department of Pathology and Laboratory Medicine, University of Vermont, Colchester, VT 05446 United States
| | - Wei Liu
- Department of Cardiology, the fourth affiliated hospital of Harbin Medical University, Harbin, Heilongjiang 150001 China; Harbin Medical University, Harbin, Heilongjiang 150001 China; Department of Geriatric Cardiovascular Division, Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080 China.
| |
Collapse
|
4
|
He Y, Yu H, Dai S, He M, Ma L, Xu Z, Luo F, Wang L. Immune checkpoint inhibitors break whose heart? Perspectives from cardio-immuno-oncology. Genes Dis 2024; 11:807-818. [PMID: 37692505 PMCID: PMC10491874 DOI: 10.1016/j.gendis.2023.01.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 01/12/2023] [Indexed: 03/30/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) are monoclonal antibody antagonists, which can block cytotoxic T lymphocyte antigen-4 (CTLA-4), programmed death-1/ligand-1 (PD-1/PD-L1) pathways, and other molecules exploited by tumor cells to evade T cell-mediated immune response. ICIs have transformed the treatment landscape for various cancers due to their amazing efficacy. Many anti-tumor therapies, including targeted therapy, radiotherapy, and chemotherapy, combine ICIs to make the treatment more effective. However, the off-target immune activation caused by ICIs may lead to a broad spectrum of immune-related adverse events (irAEs) affecting multiple organ systems. Among irAEs, cardiotoxicity induced by ICIs, uncommon but fatal, has greatly offset survival benefits from ICIs, which is heartbreaking for both patients and clinicians. Consequently, such cardiotoxicity requires special vigilance, and it has become a common challenge both for patients and clinicians. This article reviewed the clinical manifestations and influence of cardiotoxicity from the view of patients and clinicians, elaborated on the underlying mechanisms in conjunction with animal studies, and then attempted to propose management strategies from a cardio-immuno-oncology multidisciplinary perspective.
Collapse
Affiliation(s)
- Yingying He
- Oncology Department, Deyang People's Hospital, Deyang, Sichuan 618000, China
- Department of Medical Oncology, Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610000, China
| | - Hui Yu
- Cardiovascular Department, Mianyang Central Hospital, Mianyang, Sichuan 621000, China
| | - Shuang Dai
- Department of Medical Oncology, Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610000, China
| | - Miao He
- Oncology Department, Deyang People's Hospital, Deyang, Sichuan 618000, China
- Department of Medical Oncology, Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610000, China
| | - Ling Ma
- Department of Rheumatology and Immunology, Deyang People's Hospital, Deyang, Sichuan 618000, China
| | - Zihan Xu
- Department of Medical Oncology, Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610000, China
| | - Feng Luo
- Department of Medical Oncology, Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610000, China
| | - Li Wang
- Department of Medical Oncology, Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610000, China
| |
Collapse
|
5
|
Yang P, Li F, Tang J, Tian Q, Zheng Z. ET-1 receptor type B (ETBR) overexpression associated with ICAM-1 downregulation leads to inflammatory attenuation in experimental autoimmune myocarditis. PeerJ 2023; 11:e16320. [PMID: 37901475 PMCID: PMC10607261 DOI: 10.7717/peerj.16320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/29/2023] [Indexed: 10/31/2023] Open
Abstract
Background An experimental autoimmune myocarditis rat model was established by subcutaneous injection of porcine myocardial myosin (PCM). The effect of ET-1 receptor type B (ETBR) overexpression on autoimmune myocarditis was observed via tail vein injection of ETBR overexpression lentivirus in rats. We further investigated the mechanisms involved in the regulation of autoimmune myocarditis by ETBR overexpression. Methods Six rats were randomly selected from 24 male Lewis rats as the NC group, and the remaining 18 rats were injected with PCM on Day 0 and Day 7, to establish the experimental autoimmune myocarditis (EAM) rat model. The 18 rats initially immunized were randomly divided into three groups: the EAM group, ETBR-oe group, and GFP group. On Day 21 after the initial immunization of rats, cardiac echocardiography and serum brain natriuretic peptide (BNP) analysis were performed to evaluate cardiac function, myocardial tissue HE staining was performed to assess myocardial tissue inflammatory infiltration and the myocarditis score, and mRNA expression of IFN-γ, IL-12, and IL-17 was detected by qRT-PCR. Subsequently, immunohistochemical analysis was performed to detect the localization and expression of the ETBR and ICAM-1 proteins, and the expression of ETBR and ICAM-1 was verified by qRT-PCR and western blotting methods. Results On Day 21 after initial immunization, left ventricular end-diastolic diameter (LVEDd), left ventricular end-systolic diameter (LVEDs), and serum BNP concentrations increased in the hearts of rats in the EAM group compared with the NC group (P < 0.01), and ejection fraction (EF) and fractional shortening (FS) decreased compared with those of the normal control (NC) group (P < 0.01). LVEDd, LVEDs, and serum BNP concentrations decreased in the ETBR-oe group compared with the EAM group, while EF and FS increased significantly (P < 0.01). HE staining showed that a large number of inflammatory cell infiltrates, mainly lymphocytes, were observed in the EAM group, and the myocarditis score was significantly higher than that of the NC group (P < 0.01). Compared with that of the EAM group, myocardial tissue inflammatory cell infiltration was significantly reduced in the ETBR-oe group, and the myocarditis scores were significantly lower (P < 0.01). The mRNAs of the inflammatory factors IFN-γ, IL-12 and IL-17 in myocardial tissue of rats in the EAM group exhibited elevated levels compared with those of the NC group (P < 0.01) while the mRNAs of IFN-γ, IL-12 and IL-17 were significantly decreased in the ETBR-oe group compared with the EAM group (P < 0.01). Immunohistochemistry showed that the staining depth of ETBR protein in myocardial tissue was greater in the EAM group than in the NC group, and significantly greater in the ETBR-oe group than in the EAM group, while the staining depth of ICAM-1 was significantly greater in the EAM group than in the NC group, and significantly lower in the ETBR-oe group than in the EAM group. The ICAM-1 expression level was significantly higher in the EAM group than in the NC group (P < 0.01), and was significantly lower in the ETBR-oe groupthan in the EAM group (P < 0.01).
Collapse
Affiliation(s)
- Peng Yang
- Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fangfei Li
- Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jiangfeng Tang
- Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qingshan Tian
- Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhenzhong Zheng
- Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Cardiology, Shenzhen Third People's Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
6
|
Yao T, Sun B, Li Y, Sun Y, Zhang G, Yue G, Li C. Integrating network pharmacology and experimental validation to decipher the mechanism of action of Jingfang Granule in the treatment of viral myocarditis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2151-2163. [PMID: 36961551 DOI: 10.1007/s00210-023-02464-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/13/2023] [Indexed: 03/25/2023]
Abstract
This study investigated the mechanisms of Jingfang Granule (JFG) in viral myocarditis (VMC) treatment via network pharmacology-based approach combined with molecular docking and validated the results through in vitro and in vivo experiments. The chemical composition of JFG and its therapeutic targets was queried in Traditional Chinese Medicine Systems Pharmacology (TCMSP) database. The targets related to VMC were retrieved from the disease database, and the overlapping targets were screened. Based on the STRING database, a protein-protein interaction network was constructed. Cytoscape software was used to construct the "component-target-disease" interaction network for visualization. GO and KEGG pathway enrichment analyses were performed using Metascape data. Molecular docking was performed using PyMoL2.3.0 and AutoDock Vina software programs. The target genes were further verified in vitro and in vivo. JFG contains 88 active components. The main biological targets of JFG in VMC include quercetin, luteolin, and kaempferol. The molecular docking results showed that the three key targets showed strong binding properties with both the height components of the molecular docking interaction energies. The results of experimental verification results showed that JFG may be used to treat VMC mainly by down-regulating inflammatory factors TNF-α and NF-κB and inhibiting myocardial apoptosis. The results support the network pharmacological data. JFG reduces myocardial inflammation and myocardial cell apoptosis in VMC and protects myocardial tissue.
Collapse
Affiliation(s)
- Tian Yao
- Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, Shandong, China
| | - Bowen Sun
- Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, Shandong, China
| | - Yunlun Li
- Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, Shandong, China
| | - Ying Sun
- Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, Shandong, China
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, 276006, China
| | - Guimin Zhang
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, 276006, China.
| | - Guihua Yue
- Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, Shandong, China.
| | - Chao Li
- Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, Shandong, China.
| |
Collapse
|
7
|
Root-Bernstein R, Huber J, Ziehl A, Pietrowicz M. SARS-CoV-2 and Its Bacterial Co- or Super-Infections Synergize to Trigger COVID-19 Autoimmune Cardiopathies. Int J Mol Sci 2023; 24:12177. [PMID: 37569555 PMCID: PMC10418384 DOI: 10.3390/ijms241512177] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 07/20/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Autoimmune cardiopathies (AC) following COVID-19 and vaccination against SARS-CoV-2 occur at significant rates but are of unknown etiology. This study investigated the possible roles of viral and bacterial mimicry, as well as viral-bacterial co-infections, as possible inducers of COVID-19 AC using proteomic methods and enzyme-linked immunoadsorption assays. BLAST and LALIGN results of this study demonstrate that SARS-CoV-2 shares a significantly greater number of high quality similarities to some cardiac protein compared with other viruses; that bacteria such as Streptococci, Staphylococci and Enterococci also display very significant similarities to cardiac proteins but to a different set than SARS-CoV-2; that the importance of these similarities is largely validated by ELISA experiments demonstrating that polyclonal antibodies against SARS-CoV-2 and COVID-19-associated bacteria recognize cardiac proteins with high affinity; that to account for the range of cardiac proteins targeted by autoantibodies in COVID-19-associated autoimmune myocarditis, both viral and bacterial triggers are probably required; that the targets of the viral and bacterial antibodies are often molecularly complementary antigens such as actin and myosin, laminin and collagen, or creatine kinase and pyruvate kinase, that are known to bind to each other; and that the corresponding viral and bacterial antibodies recognizing these complementary antigens also bind to each other with high affinity as if they have an idiotype-anti-idiotype relationship. These results suggest that AC results from SARS-CoV-2 infections or vaccination complicated by bacterial infections. Vaccination against some of these bacterial infections, such as Streptococci and Haemophilus, may therefore decrease AC risk, as may the appropriate and timely use of antibiotics among COVID-19 patients and careful screening of vaccinees for signs of infection such as fever, diarrhea, infected wounds, gum disease, etc.
Collapse
Affiliation(s)
- Robert Root-Bernstein
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA; (J.H.); (A.Z.); (M.P.)
| | | | | | | |
Collapse
|
8
|
Frasca L, Ocone G, Palazzo R. Safety of COVID-19 Vaccines in Patients with Autoimmune Diseases, in Patients with Cardiac Issues, and in the Healthy Population. Pathogens 2023; 12:pathogens12020233. [PMID: 36839505 PMCID: PMC9964607 DOI: 10.3390/pathogens12020233] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/25/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) has been a challenge for the whole world since the beginning of 2020, and COVID-19 vaccines were considered crucial for disease eradication. Instead of producing classic vaccines, some companies pointed to develop products that mainly function by inducing, into the host, the production of the antigenic protein of SARS-CoV-2 called Spike, injecting an instruction based on RNA or a DNA sequence. Here, we aim to give an overview of the safety profile and the actual known adverse effects of these products in relationship with their mechanism of action. We discuss the use and safety of these products in at-risk people, especially those with autoimmune diseases or with previously reported myocarditis, but also in the general population. We debate the real necessity of administering these products with unclear long-term effects to at-risk people with autoimmune conditions, as well as to healthy people, at the time of omicron variants. This, considering the existence of therapeutic interventions, much more clearly assessed at present compared to the past, and the relatively lower aggressive nature of the new viral variants.
Collapse
|
9
|
Sun L, Yuan H, Zhao G. IL-37 alleviates Coxsackievirus B3-induced viral myocarditis via inhibiting NLRP3 inflammasome-mediated pyroptosis. Sci Rep 2022; 12:20077. [PMID: 36418383 PMCID: PMC9684492 DOI: 10.1038/s41598-022-22617-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 10/17/2022] [Indexed: 11/24/2022] Open
Abstract
Our study aims to verify the potential effects and underlying mechanisms of IL-37 in Coxsackievirus B3 (CVB3)-induced viral myocarditis (VMC). VMC model was established by intraperitoneal injection of CVB3 into 6-week-old male balb-c mice on day 0. Each mouse of the IL-37-control group and IL-37-VMC CVB3 groups was intraperitoneally injected with IL-37 on day 4 and day 7. The cardiac function was evaluated by transthoracic echocardiography including LVEF, LVFE, IVSs and IVSd. Myocardial injury was measured by Elisa for serum cTnI. The inflammation infiltration and fibrosis were evaluated by hematoxylin and eosin (HE) staining and Masson staining. The expression levels of NLRP3 inflammasome components in pyroptosis were determined by western blot, Elisa, and immunofluorescent analysis. We also detected the expression of IL-37-IL-1R8 in PBMCs by immunofluorescence after injection with CVB3 and IL-37. Compared with the VMC group, mice received CVB3 and IL-37 have improved cardiac function, reduced inflammation infiltration and fibrosis, and with lower expression of cTnI, IL-1β, IL-18 and NLRP3 inflammasome component. IL-37 weakened the upregulation of GSDMD and phosphorylation of NF-κB p65 induced by CVB3. Exogenous addition of IL-37 with CVB3 further increases the production of IL-37-IL-1R8 -IL-18RA complex in vitro. Our findings indicate that IL-37 alleviates CVB3-induced VMC, which may be produced by inhibiting NLRP3 inflammasome-mediated pyroptosis, NF-κB signaling pathway, and IL-37-IL-1R8 -IL-18RA complex.
Collapse
Affiliation(s)
- Lin Sun
- grid.27255.370000 0004 1761 1174Department of Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021 China ,grid.506261.60000 0001 0706 7839Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037 China
| | - Haitao Yuan
- grid.27255.370000 0004 1761 1174Department of Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021 China ,grid.460018.b0000 0004 1769 9639Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021 China
| | - Gang Zhao
- grid.27255.370000 0004 1761 1174Department of Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021 China ,grid.460018.b0000 0004 1769 9639Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021 China
| |
Collapse
|
10
|
Viruses in the Heart: Direct and Indirect Routes to Myocarditis and Heart Failure. Viruses 2021; 13:v13101924. [PMID: 34696354 PMCID: PMC8537553 DOI: 10.3390/v13101924] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/09/2021] [Accepted: 09/21/2021] [Indexed: 01/01/2023] Open
Abstract
Viruses are an underappreciated cause of heart failure. Indeed, several types of viral infections carry cardiovascular risks. Understanding shared and unique mechanisms by which each virus compromises heart function is critical to inform on therapeutic interventions. This review describes how the key viruses known to lead to cardiac dysfunction operate. Both direct host-damaging mechanisms and indirect actions on the immune systems are discussed. As viral myocarditis is a key pathologic driver of heart failure in infected individuals, this review also highlights the role of cytokine storms and inflammation in virus-induced cardiomyopathy.
Collapse
|
11
|
Eftekhar SP, Yazdanpanah N, Rezaei N. Immune checkpoint inhibitors and cardiotoxicity: possible mechanisms, manifestations, diagnosis and management. Expert Rev Anticancer Ther 2021; 21:1211-1228. [PMID: 34511008 DOI: 10.1080/14737140.2021.1979396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) are a new class of anticancer drugs that enhance the immune system function and activate T cells against cancerous cells. Although cardiac complications are not common, they could be accompanied with high morbidity and mortality. AREAS COVERED Regarding the importance of cardiac complications and their subsequent burden on individuals and the healthcare system, this review attempts to discuss the mechanism, diagnosis, and management of myocarditis, besides recapitulating the possible mechanism of other cardiac adverse events. Moreover, we briefly discuss the concurrent administration of other chemotherapeutic agents. EXPERT OPINION Due to insufficient knowledge concerning the physiopathology of immune-related adverse events (irAEs) and their potential further complications, cardiovascular complications in particular and in the context of this paper's focus, cooperation of oncologists, immunologists, and cardiologists is necessary for the management of patients. Experimental approaches such as using corticosteroids are becoming a part of guidelines for managing cardiac irAEs. However, a unique algorithm for diagnosis and management is necessary, especially in myocarditis cases. Furthermore, more studies are required to resolve current challenges, including prevention of myocarditis, concurrent administration of other chemotherapeutic agents, and re-introducing patients with ICIs.
Collapse
Affiliation(s)
- Seyed Parsa Eftekhar
- School of Medicine, Babol University of Medical Sciences, Babol, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (Niima), Universal Scientific Education and Research Network (Usern), Babol, Iran
| | - Niloufar Yazdanpanah
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (Niima), Universal Scientific Education and Research Network (Usern), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (Niima), Universal Scientific Education and Research Network (Usern), Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Wu Y, Zheng Z, Cao X, Yang Q, Norton V, Adini A, Maiti AK, Adini I, Wu H. RIP1/RIP3/MLKL Mediates Myocardial Function Through Necroptosis in Experimental Autoimmune Myocarditis. Front Cardiovasc Med 2021; 8:696362. [PMID: 34497836 PMCID: PMC8419468 DOI: 10.3389/fcvm.2021.696362] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 07/22/2021] [Indexed: 12/23/2022] Open
Abstract
Cardiomyopathy often leads to dilated cardiomyopathy (DCM) when caused by viral myocarditis. Apoptosis is long considered as the principal process of cell death in cardiomyocytes, but programmed necrosis or necroptosis is recently believed to play an important role in cardiomyocyte cell death. We investigated the role of necroptosis and its interdependency with other processes of cell death, autophagy, and apoptosis in a rat system of experimental autoimmune myocarditis (EAM). We successfully created a rat model system of EAM by injecting porcine cardiac myosin (PCM) and showed that in EAM, all three forms of cell death increase considerably, resulting in the deterioration of cardiac conditions with an increase in inflammatory infiltration in cardiomyocytes. To explore whether necroptosis occurs in EAM rats independent of autophagy, we treated EAM rats with a RIP1/RIP3/MLKL kinase-mediated necroptosis inhibitor, Necrostatin-1 (Nec-1). In Nec-1 treated rats, cell death proceeds through apoptosis but has no significant effect on autophagy. In contrast, autophagy inhibitor 3-Methyl Adenine (3-MA) increases necroptosis, implying that blockage of autophagy must be compensated through necroptosis. Caspase 8 inhibitor zVAD-fmk blocks apoptosis but increases both necroptosis and autophagy. However, all necroptosis, apoptosis, and autophagy inhibitors independently reduce inflammatory infiltration in cardiomyocytes and improve cardiac conditions. Since apoptosis or autophagy is involved in many important cellular aspects, instead of suppressing these two major cell death processes, Nec1 can be developed as a potential therapeutic target for inflammatory myocarditis.
Collapse
Affiliation(s)
- Yujing Wu
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China.,Department of Cardiology, Jiangxi Hypertension Research Institute, Nanchang, China.,Department of Emergency, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhenzhong Zheng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China.,Department of Cardiology, Jiangxi Hypertension Research Institute, Nanchang, China
| | - Xiantong Cao
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qing Yang
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China.,Department of Cardiology, Jiangxi Hypertension Research Institute, Nanchang, China
| | - Vikram Norton
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Avner Adini
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Amit K Maiti
- Mydnavar, Department of Genetics and Genomics, Troy, MI, United States
| | - Irit Adini
- Center for Engineering in Medicine, Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States
| | - Hao Wu
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| |
Collapse
|
13
|
Kogan EA, Kukleva AD, Berezovskiy YS, Blagova OV, Zharkov NV, Ainetdinova DK, Demyashkin GA. [Clinical and morphological characteristics of SARS-CoV-2-related myocarditis proven by the presence of viral RNA and proteins in myocardial tissue]. Arkh Patol 2021; 83:5-13. [PMID: 34278755 DOI: 10.17116/patol2021830415] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To investigate the clinical and morphological features of SARS-CoV-2-related myocarditis, by determining the presence of viral RNA and proteins in myocardial tissue. MATERIAL AND METHODS The study was conducted to examine the material of 32 autopsies with a confirmed diagnosis of myocarditis. There were data of a morphological study, including a standard histological study, as well as immunohistochemical determination of the surface markers CD45, CD3, CD20, and CD68 cells of an inflammatory infiltrate and virus proteins (SARS-CoV-2 nucleocapsid protein and spike protein). Positive and negative control tests were carried out. In addition, coronavirus RNA was detected in the myocardium using a polymerase chain reaction. RESULTS Polymerase chain reaction (PCR) revealed viral RNA in myocardial tissue. Viral proteins were identified in the macrophages of an inflammatory infiltrate and cardiomyocytes. CONCLUSION The findings may suggest that the virus persists in the myocardium and chronic myocarditis develops.
Collapse
Affiliation(s)
- E A Kogan
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of Russia, Moscow, Russia
| | - A D Kukleva
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of Russia, Moscow, Russia
| | | | - O V Blagova
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of Russia, Moscow, Russia
| | - N V Zharkov
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of Russia, Moscow, Russia
| | - D Kh Ainetdinova
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of Russia, Moscow, Russia
| | - G A Demyashkin
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of Russia, Moscow, Russia
| |
Collapse
|
14
|
Colombo CSSDS, Leitão MB, Avanza AC, Borges SF, da Silveira AD, Braga F, Camarozano AC, Kopiler DA, Lazzoli JK, de Freitas OGA, Grossman GB, Milani M, Nunes MB, Ritt LEF, Sellera CAC, Ghorayeb N. Position Statement on Post-COVID-19 Cardiovascular Preparticipation Screening: Guidance for Returning to Physical Exercise and Sports - 2020. Arq Bras Cardiol 2021; 116:1213-1226. [PMID: 34133609 PMCID: PMC8288531 DOI: 10.36660/abc.20210368] [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: 12/23/2022] Open
Affiliation(s)
- Cléa Simone Sabino de Souza Colombo
- Faculdade de Medicina São Leopoldo MandicCampinasSPBrasilFaculdade de Medicina São Leopoldo Mandic – Campinas, SP – Brasil
- Sportscardio Clínica CardiológicaValinhosSPBrasilSportscardio Clínica Cardiológica – Valinhos, SP – Brasil
- Hospital do CoraçãoSão PauloSPBrasilHospital do Coração (HCor), São Paulo, SP – Brasil
| | - Marcelo Bichels Leitão
- CEFIT – Centro de Estudos de Fisiologia do Exercício e TreinamentoCuritibaPRBrasilCEFIT – Centro de Estudos de Fisiologia do Exercício e Treinamento, Curitiba, PR – Brasil
- Conselho Federal de MedicinaCâmara técnica de Medicina DesportivaRio de JaneiroRJBrasilConselho Federal de Medicina, Câmara técnica de Medicina Desportiva, Rio de Janeiro, RJ – Brasil
- Comissão de Autorização para Uso Terapêutico (CAUT) da Autoridade Brasileira de Controle de DopagemBrasilComissão de Autorização para Uso Terapêutico (CAUT) da Autoridade Brasileira de Controle de Dopagem (ABCD)
| | - Antônio Carlos Avanza
- Universidade Vila VelhaESBrasilUniversidade Vila Velha, ES – Brasil
- Clínica CentrocorVitóriaESBrasilClínica Centrocor, Vitória, ES – Brasil
| | - Serafim Ferreira Borges
- Clube de Regatas do FlamengoRio de JaneiroRJBrasilClube de Regatas do Flamengo, Rio de Janeiro, RJ – Brasil
- Instituto Estadual de Cardiologia Aloysio de CastroRio de JaneiroRJBrasilInstituto Estadual de Cardiologia Aloysio de Castro, Rio de Janeiro, RJ – Brasil
- Imagecor Medicina Diagnóstica e do ExercícioRio de JaneiroRJBrasilImagecor Medicina Diagnóstica e do Exercício, Rio de Janeiro, RJ – Brasil
- Conselho Federal de MedicinaCâmara técnica de Medicina DesportivaRio de JaneiroRJBrasilConselho Federal de Medicina, Câmara técnica de Medicina Desportiva, Rio de Janeiro, RJ – Brasil
| | - Anderson Donelli da Silveira
- Hospital de Clínicas de Porto AlegrePorto AlegreRSBrasilHospital de Clínicas de Porto Alegre, Porto Alegre, RS – Brasil
- Universidade Federal do Rio Grande do SulPorto AlegreRSBrasilUniversidade Federal do Rio Grande do Sul, Porto Alegre, RS – Brasil
| | - Fabrício Braga
- Laboratório de Performance HumanaRio de JaneiroRJBrasilLaboratório de Performance Humana, Rio de Janeiro, RJ – Brasil
- Casa de Saúde São JoséRio de JaneiroRJBrasilCasa de Saúde São José, Rio de Janeiro, RJ – Brasil
- Confederação Brasileira de TriathlonRio de JaneiroRJBrasilConfederação Brasileira de Triathlon, Rio de Janeiro, RJ – Brasil
| | - Ana Cristina Camarozano
- Universidade Federal do ParanáCuritibaPRBrasilUniversidade Federal do Paraná, Curitiba, PR – Brasil
| | - Daniel Arkader Kopiler
- Instituto Nacional de CardiologiaRio de JaneiroRJBrasilInstituto Nacional de Cardiologia (INC), Rio de Janeiro, RJ – Brasil
- Confederação Pan-Americana de Medicina do EsporteBrasilConfederação Pan-Americana de Medicina do Esporte
| | - José Kawazoe Lazzoli
- Instituto Biomédico da Universidade Federal FluminenseNiteróiRJBrasilInstituto Biomédico da Universidade Federal Fluminense (UFF), Niterói, RJ – Brasil
- Hospital Santa Teresa/ACSCPetrópolisRJBrasilHospital Santa Teresa/ACSC, Petrópolis, RJ – Brasil
- Confederação Pan-Americana de Medicina do EsporteBrasilConfederação Pan-Americana de Medicina do Esporte (COPAMEDE)
- Federação Internacional de Medicina do EsporteBrasilFederação Internacional de Medicina do Esporte (FIMS)
- Comissão de Autorização para Uso Terapêutico (CAUT) da Autoridade Brasileira de Controle de DopagemBrasilComissão de Autorização para Uso Terapêutico (CAUT) da Autoridade Brasileira de Controle de Dopagem (ABCD)
| | | | - Gabriel Blacher Grossman
- Hospital Moinhos de VentoPorto AlegreRSBrasilHospital Moinhos de Vento, Porto Alegre, RS – Brasil
- Clínica CardionuclearPorto AlegreRSBrasilClínica Cardionuclear, Porto Alegre, RS – Brasil
| | - Mauricio Milani
- Fitcordis Medicina do ExercícioBrasíliaDFBrasilFitcordis Medicina do Exercício, Brasília, DF – Brasil
| | - Mauricio B. Nunes
- Hospital PortuguêsSalvadorBABrasilHospital Português, Salvador, BA – Brasil
| | - Luiz Eduardo Fonteles Ritt
- Hospital Cárdio PulmonarSalvadorBABrasilHospital Cárdio Pulmonar, Salvador, BA – Brasil
- Escola Bahiana de Medicina e Saúde PúblicaSalvadorBABrasilEscola Bahiana de Medicina e Saúde Pública, – Salvador, BA – Brasil
| | - Carlos Alberto Cyrillo Sellera
- Santa Casa de SantosSantosSPBrasilSanta Casa de Santos, Santos, SP – Brasil
- Universidade Metropolitana de SantosSantosSPBrasilUniversidade Metropolitana de Santos, Santos, SP – Brasil
| | - Nabil Ghorayeb
- Instituto Dante Pazzanese de CardiologiaSão PauloSPBrasilInstituto Dante Pazzanese de Cardiologia, São Paulo, SP – Brasil
- Hospital do CoraçãoSão PauloSPBrasilHospital do Coração (HCor), São Paulo, SP – Brasil
| |
Collapse
|
15
|
COVID-19 and severe ENT infections in pediatric patients. IS there a relationship? Int J Pediatr Otorhinolaryngol 2021; 145:110714. [PMID: 33894522 PMCID: PMC8051009 DOI: 10.1016/j.ijporl.2021.110714] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 02/21/2021] [Accepted: 04/08/2021] [Indexed: 12/01/2022]
Abstract
BACKGROUND Preliminary evidence suggests that children are just as likely to become infected with SARS-CoV-2 as adults but are less prone to developing severe clinical conditions. However, there are pediatric inflammatory conditions that have also been encountered. The aim of this report is to determine whether there is a relationship between COVID-19 and severe infections in the ear, nose, throat, and deep cervical area (ENT) in pediatric populations. MATERIALS AND METHODS A compilation was made of all the cases of ENT area infections in the pediatric population per month attended to at the Niño Jesús University Children's Hospital from January 2010 to June 2020. Endemic channels and dispersion analysis were designed to analyze the incidence presented in the year 2020, compared to what was expected based on historical data from 2010 to 2019. Then, an epidemiological interview was conducted of the close contacts of COVID-19 of the children who presented a severe ENT infection in 2020. Finally, a serological test of IgG antibodies was performed on all of them to find out if they had overcome the COVID- 19. RESULTS 620 patients from 1022 were eligible for the study. We observed a significant outbreak in the incidence of complicated mastoiditis and deep cervical infections with complications in the year 2020 (13 patients) linked to the COVID-19 pandemic. From these patients, 54% had been confirmed or had high suspicion of close contact with COVID-19.15.4% of children were positive in serological tests for IgG antibodies. CONCLUSION There has been a significant increase in mastoiditis and deep cervical infections with complications in the first four months of 2020, which constitutes an outbreak. A considerable number (54%) of these complicated infections were related to close contact with COVID-19. Still, only 15.4% were positive in serological tests for IgG antibodies, so we cannot establish a direct categorical relationship. The limitations in primary care due to a shortage of human resources in dealing with the pandemic's initial onslaught and changes in help-seeking behavior could explain increased complicated infections.
Collapse
|
16
|
Moslehi J, Lichtman AH, Sharpe AH, Galluzzi L, Kitsis RN. Immune checkpoint inhibitor-associated myocarditis: manifestations and mechanisms. J Clin Invest 2021; 131:145186. [PMID: 33645548 DOI: 10.1172/jci145186] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have transformed the treatment of various cancers, including malignancies once considered untreatable. These agents, however, are associated with inflammation and tissue damage in multiple organs. Myocarditis has emerged as a serious ICI-associated toxicity, because, while seemingly infrequent, it is often fulminant and lethal. The underlying basis of ICI-associated myocarditis is not completely understood. While the importance of T cells is clear, the inciting antigens, why they are recognized, and the mechanisms leading to cardiac cell injury remain poorly characterized. These issues underscore the need for basic and clinical studies to define pathogenesis, identify predictive biomarkers, improve diagnostic strategies, and develop effective treatments. An improved understanding of ICI-associated myocarditis will provide insights into the equilibrium between the immune and cardiovascular systems.
Collapse
Affiliation(s)
- Javid Moslehi
- Division of Cardiovascular Medicine and Division of Oncology, Cardio-Oncology Program, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Andrew H Lichtman
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Arlene H Sharpe
- Department of Immunology and Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA.,Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Sandra and Edward Meyer Cancer Center, Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medical College, New York, New York, USA.,Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA.,Université de Paris, Paris, France
| | - Richard N Kitsis
- Departments of Medicine and Cell Biology, Wilf Family Cardiovascular Research Institute, and Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
17
|
Jaiswal V, Sarfraz Z, Sarfraz A, Mukherjee D, Batra N, Hitawala G, Yaqoob S, Patel A, Agarwala P, Ruchika, Sarfraz M, Bano S, Azeem N, Naz S, Jaiswal A, Sharma P, Chaudhary G. COVID-19 Infection and Myocarditis: A State-of-the-Art Systematic Review. J Prim Care Community Health 2021; 12:21501327211056800. [PMID: 34854348 PMCID: PMC8647231 DOI: 10.1177/21501327211056800] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/13/2021] [Accepted: 10/13/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND COVID-19 was initially considered to be a respiratory illness, but current findings suggest that SARS-CoV-2 is increasingly expressed in cardiac myocytes as well. COVID-19 may lead to cardiovascular injuries, resulting in myocarditis, with inflammation of the heart muscle. OBJECTIVE This systematic review collates current evidence about demographics, symptomatology, diagnostic, and clinical outcomes of COVID-19 infected patients with myocarditis. METHODS In accordance with PRISMA 2020 guidelines, a systematic search was conducted using PubMed, Cochrane Central, Web of Science and Google Scholar until August, 2021. A combination of the following keywords was used: SARS-CoV-2, COVID-19, myocarditis. Cohorts and case reports that comprised of patients with confirmed myocarditis due to COVID-19 infection, aged >18 years were included. The findings were tabulated and subsequently synthesized. RESULTS In total, 54 case reports and 5 cohorts were identified comprising 215 patients. Hypertension (51.7%), diabetes mellitus type 2 (46.4%), cardiac comorbidities (14.6%) were the 3 most reported comorbidities. Majority of the patients presented with cough (61.9%), fever (60.4%), shortness of breath (53.2%), and chest pain (43.9%). Inflammatory markers were raised in 97.8% patients, whereas cardiac markers were elevated in 94.8% of the included patients. On noting radiographic findings, cardiomegaly (32.5%) was the most common finding. Electrocardiography testing obtained ST segment elevation among 44.8% patients and T wave inversion in 7.3% of the sample. Cardiovascular magnetic resonance imaging yielded 83.3% patients with myocardial edema, with late gadolinium enhancement in 63.9% patients. In hospital management consisted of azithromycin (25.5%), methylprednisolone/steroids (8.5%), and other standard care treatments for COVID-19. The most common in-hospital complication included acute respiratory distress syndrome (66.4%) and cardiogenic shock (14%). On last follow up, 64.7% of the patients survived, whereas 31.8% patients did not survive, and 3.5% were in the critical care unit. CONCLUSION It is essential to demarcate COVID-19 infection and myocarditis presentations due to the heightened risk of death among patients contracting both myocardial inflammation and ARDS. With a multitude of diagnostic and treatment options available for COVID-19 and myocarditis, patients that are under high risk of suspicion for COVID-19 induced myocarditis must be appropriately diagnosed and treated to curb co-infections.
Collapse
Affiliation(s)
| | | | | | | | - Nitya Batra
- Maulana Azad Medical College, New
Delhi, India
| | | | - Sadia Yaqoob
- Jinnah Medical and Dental College,
Karachi, Pakistan
| | | | | | - Ruchika
- JJM Medical college, Davangere,
India
| | | | - Shehar Bano
- Fatima Jinnah Medical University,
Lahore, Pakistan
| | | | - Sidra Naz
- Harvard Medical School, Boston, MA,
USA
| | - Akash Jaiswal
- All India Institute of Medical
Science, New Delhi, India
| | | | | |
Collapse
|
18
|
Mirtajaddini M, Khajali Z, Naderi N, Rezaeian N. Does myocardial injury rapidly progress in marfan syndrome following COVID-19? Res Cardiovasc Med 2021. [DOI: 10.4103/rcm.rcm_10_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
19
|
Chen X, Zhao MZ, Miao BP, Liu ZQ, Yang G, Liu JQ, Yang PC, Song JP. Inhibition of Bcl2L12 Attenuates Eosinophilia-Related Inflammation in the Heart. Front Immunol 2020; 11:1955. [PMID: 33013849 PMCID: PMC7516035 DOI: 10.3389/fimmu.2020.01955] [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: 03/05/2020] [Accepted: 07/20/2020] [Indexed: 11/25/2022] Open
Abstract
Background: The eosinophilic inflammation plays a critical role in myocarditis (Mcd); its underlying mechanism remains to be further elucidated. This study aims to investigate the role of Bcl2-like protein 12 (Bcl2L12) in inducing the defects of apoptosis in eosinophils (Eos) of the heart tissues. Methods: Human explant heart samples were collected. Eosinophilia and myocarditis (Mcd)-like inflammation were induced in the mouse heart by immunizing with murine cardiac α-myosin heavy chain (MyHCα) peptides. Results: Markedly more Eos were observed in heart tissues from patients with Mcd than those from patients with dilated cardiomyopathy. Eos isolated from Mcd hearts showed the signs of apoptosis defects. The Eo counts in the Mcd heart tissues were positively correlated with the Bcl2L12 expression in Eos isolated from the heart tissues. Exposure to interleukin 5 in the culture induced the expression of Bcl2L12 in Eos. Bcl2L12 bound c-Myc, the transcription factor of Fas ligand (FasL), to prevent c-Myc from binding to the FasL promoter, to restrict the FasL gene transcription in Eos. Inhibition of Bcl2L12 prevented the induction of eosinophilia and Mcd-like inflammation in the mouse heart. Conclusions: The Bcl2L12 expression contributes to apoptosis defects in Eos of the Mcd heart. Blocking Bcl2L12 prevents the eosinophilia induction and alleviates Mcd-like inflammation in mice.
Collapse
Affiliation(s)
- Xiao Chen
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mei-Zhen Zhao
- Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Bei-Ping Miao
- Department of Otolaryngology, First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Zhi-Qiang Liu
- Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Gui Yang
- Department of Otolaryngology, Longgang Central Hospital, Shenzhen, China
| | - Jiang-Qi Liu
- Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Ping-Chang Yang
- Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China.,Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen, China
| | - Jiang-Ping Song
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
20
|
Pirzada A, Mokhtar AT, Moeller AD. COVID-19 and Myocarditis: What Do We Know So Far? CJC Open 2020; 2:278-285. [PMID: 32691024 PMCID: PMC7254016 DOI: 10.1016/j.cjco.2020.05.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 05/20/2020] [Indexed: 02/06/2023] Open
Abstract
COVID-19 has been declared a global pandemic by the World Health Organization and is responsible for hundreds of thousands of deaths worldwide. COVID-19 is caused by SARS-CoV-2, and common clinical symptoms include fever, cough, sore throat, headache, and fatigue. Myocardial injury is relatively common in patients with COVID-19, accounting for 7%-23% of cases, and is associated with a higher rate of morbidity and mortality. There is a discrepancy in the literature about myocarditis as the etiology of myocardial injury in patients with COVID-19; although many anecdotal reports of myocarditis have been noted, there are only a handful of case reports in the literature about myocarditis related to COVID-19. In this review we summarize the most up to date literature around the association between COVID-19 and myocarditis and provide clinicians a practical framework about the clinical manifestations, diagnostic tools, and treatment options currently available. Importantly, this review will heighten suspicion for myocarditis as an etiology of myocardial injury in COVID-19 patients, therefore improving clinical outcomes and encouraging shared clinical decision-making. This will also open the door for further research to build around this review. Emergent treatment options for COVID-19 are in clinical trials and might be of benefit to COVID-19 patients with myocarditis in addition to current guideline-based recommendations.
Collapse
Affiliation(s)
- Ashar Pirzada
- Division of Cardiology, Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Ahmed T. Mokhtar
- Division of Cardiology, Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Andrew D. Moeller
- Division of Cardiology, Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
21
|
Grabie N, Lichtman AH, Padera R. T cell checkpoint regulators in the heart. Cardiovasc Res 2020; 115:869-877. [PMID: 30721928 DOI: 10.1093/cvr/cvz025] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 01/07/2019] [Accepted: 02/04/2019] [Indexed: 12/27/2022] Open
Abstract
T lymphocyte-mediated immune responses in the heart are potentially dangerous because they can interfere with the electromechanical function. Furthermore, the myocardium has limited regenerative capacity to repair damage caused by effector T cells. Myocardial T cell responses are normally suppressed by multiple mechanisms of central and peripheral tolerance. T cell inhibitory molecules, so called immune checkpoints, limit the activation and effector function of heart antigen-reactive T cells that escape deletion during development in the thymus. Programmed cell protein death-1 (PD-1) and cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) are checkpoint molecules homologous to the costimulatory receptor CD28, and they work to block activating signals from the T cell antigen receptor and CD28. Nonetheless, PD-1 and CTLA-4 function in different ways and at different steps in a T cell response to antigen. Studies in mice have established that genetic deficiencies of checkpoint molecules, including PD-1, PD-L1, CTLA-4, and lymphocyte activation gene-3, result in enhanced risk of autoimmune T cell-mediated myocarditis and increased pathogenicity of heart antigen-specific effector T cells. The PD-1/PD-L1 pathway appears to be particularly important in cardiac protection from T cells. PD-L1 is markedly up-regulated on myocardial cells by interferon-gamma secreted by T cells and PD-1 or PD-L1 deficiency synergizes with other defects in immune regulation in promoting myocarditis. Consistent with these studies, myocarditis has emerged as a serious adverse reaction of cancer therapies that target checkpoint molecules to enhance anti-tumour T cell responses. Histopathology and immunohistochemical analyses of myocardial tissue from immune checkpoint blockade (ICB)-treated patients echoes findings in checkpoint-deficient mice. Many questions about myocarditis in the setting of cancer immunotherapy still need to be answered, including the nature of the target antigens, genetic risk factors, and variations in the disease with combined therapies. Addressing these questions will require further immunological analyses of blood and heart tissue from patients treated with ICB.
Collapse
Affiliation(s)
- Nir Grabie
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, NRB Room 752N, 77 Avenue Louis Pasteur, Boston, MA, USA
| | - Andrew H Lichtman
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, NRB Room 752N, 77 Avenue Louis Pasteur, Boston, MA, USA
| | - Robert Padera
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, NRB Room 752N, 77 Avenue Louis Pasteur, Boston, MA, USA
| |
Collapse
|
22
|
Fulminant myocarditis due to the influenza B virus in adults: Report of two cases and literature review. BIOMEDICA 2019; 39:11-19. [PMID: 31529829 DOI: 10.7705/biomedica.v39i3.4645] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Indexed: 11/21/2022]
Abstract
La miocarditis es una enfermedad inflamatoria del miocardio. Las infecciones virales son la causa más común, aunque también puede deberse a reacciones de hipersensibilidad y de etiología autoinmunitaria, entre otras. El espectro clínico de la enfermedad es variado y comprende desde un curso asintomático, seguido de dolor torácico, arritmias y falla cardiaca aguda, hasta un cuadro fulminante. El término 'fulminante' se refiere al desarrollo de un shock cardiogénico con necesidad de soporte vasopresor e inotrópico o dispositivos de asistencia circulatoria, ya sea oxigenación por membrana extracorpórea o balón de contrapulsación intraaórtico. Cerca del 10 % de los casos de falla cardiaca por miocarditis corresponde a miocarditis fulminante. La miocarditis por influenza se considera una condición infrecuente; no obstante, su incidencia ha aumentado desde el 2009 a raíz de la pandemia de influenza por el virus AH1N1. Por su parte, la miocarditis por influenza de tipo B sigue siendo una condición infrecuente. Se describen aquí dos casos confirmados de miocarditis fulminante por el virus de la influenza B atendidos en un centro cardiovascular, que requirieron dispositivos de asistencia circulatoria mecánica.
Collapse
|
23
|
Ammirati E, Veronese G, Cipriani M, Moroni F, Garascia A, Brambatti M, Adler ED, Frigerio M. Acute and Fulminant Myocarditis: a Pragmatic Clinical Approach to Diagnosis and Treatment. Curr Cardiol Rep 2018; 20:114. [PMID: 30259175 DOI: 10.1007/s11886-018-1054-z] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW To review the clinical features of acute myocarditis, including its fulminant presentation, and present a pragmatic approach to the diagnosis and treatment, considering indications of American and European Scientific Statements and recent data derived by large contemporary registries. RECENT FINDINGS Patients presenting with acute uncomplicated myocarditis (i.e., without left ventricular dysfunction, heart failure, or ventricular arrhythmias) have a favorable short- and long-term prognosis: these findings do not support the indication to endomyocardial biopsy in this clinical scenario. Conversely, patients with complicated presentations, especially those with fulminant myocarditis, require an aggressive and comprehensive management, including endomyocardial biopsy and availability of advanced therapies for circulatory support. Although several immunomodulatory or immunosuppressive therapies have been studied and are actually prescribed in the real-world practice, their effectiveness has not been clearly demonstrated. Patients with specific histological subtypes of acute myocarditis (i.e., giant cell and eosinophilic myocarditis) or those affected by sarcoidosis or systemic autoimmune disorders seem to benefit most from immunosuppression. On the other hand, no clear evidence supports the use of immunosuppressive agents in patients with lymphocytic acute myocarditis, even though small series suggest a potential benefit. Acute myocarditis is a heterogeneous condition with distinct pathophysiological pathways. Further research is mandatory to identify factors and mechanisms that may trigger/maintain or counteract/repair the myocardial damage, in order to provide a rational for future evidence-based treatment of patients affected by this condition.
Collapse
Affiliation(s)
- Enrico Ammirati
- "De Gasperis" Cardio Center and Transplant Center, Niguarda Hospital, Milan, Italy.
| | - Giacomo Veronese
- "De Gasperis" Cardio Center and Transplant Center, Niguarda Hospital, Milan, Italy.,School of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Manlio Cipriani
- "De Gasperis" Cardio Center and Transplant Center, Niguarda Hospital, Milan, Italy
| | | | - Andrea Garascia
- "De Gasperis" Cardio Center and Transplant Center, Niguarda Hospital, Milan, Italy
| | - Michela Brambatti
- Division of Cardiology, Department of Medicine, University of California San Diego, San Diego, CA, USA
| | - Eric D Adler
- Division of Cardiology, Department of Medicine, University of California San Diego, San Diego, CA, USA
| | - Maria Frigerio
- "De Gasperis" Cardio Center and Transplant Center, Niguarda Hospital, Milan, Italy
| |
Collapse
|
24
|
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: 27] [Impact Index Per Article: 4.5] [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
|
25
|
Role of gene polymorphisms/haplotypes and serum levels of interleukin-17A in susceptibility to viral myocarditis. Exp Mol Pathol 2018. [PMID: 29530464 DOI: 10.1016/j.yexmp.2018.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Interleukin-17A (IL-17A) has been implicated in the pathogenesis of viral myocarditis (VMC). However, the role of IL-17A polymorphisms in susceptibility to VMC has not been reported to date. The aim of this study was to explore the association between IL-17A variants as well as serum IL-17 levels with VMC. Three single-nucleotide polymorphisms (SNPs) (rs2275913, rs3819025, and rs3748067) were analyzed by the polymerase chain reaction-restriction fragment length polymorphism method in 236 VMC patients and 259 controls from China. Serum IL-17A levels were measured by enzyme-linked immunosorbent assay kits. Multivariable logistic regression analysis that the rs2275913 AA genotype and the haplotype -197A/+45G/+1249G (AGG) were associated with an increased risk of VMC (all P < 0.05). Consistent with these findings, the rs2275913 AA genotype was linked to higher serum IL-17A compared to GG/AG genotype (all P < 0.001). We observed no associations between the other two SNPs and risk of VMC. Serum IL-17A levels were significantly higher in the VMC group than controls (P < 0.001) and gradually increased with the increase of New York Heart Association grade in VMC patients (P < 0.05). Spearman correlation test revealed that the serum IL-17A level was correlated with the cardiac damage and left ventricular systolic functions among VMC patients (all P < 0.05). Our study reveals that IL-17A expression may contribute to the development and severity of VMC. The SNP rs2275913 in the IL-17A gene might exert influence on susceptibility to VMC via linking with the serum IL-17A level.
Collapse
|
26
|
Honore PM, Spapen HD. Fulminant myocarditis in children. Continuous renal replacement therapy to the rescue? ACTA ACUST UNITED AC 2018; 63:941-942. [PMID: 29451654 DOI: 10.1590/1806-9282.63.11.941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Patrick M Honore
- MD, PhD, FCCM, Professor of Intensive Care Medicine and Co-director of Research, ICU Department, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Herbert D Spapen
- MD, PhD, FCCM, Professor of Intensive Care Medicine, Head of Unit and Director of Research, ICU Department, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
27
|
Krishnan B, Massilamany C, Basavalingappa RH, Gangaplara A, Rajasekaran RA, Afzal MZ, Khalilzad-Sharghi V, Zhou Y, Riethoven JJ, Nandi SS, Mishra PK, Sobel RA, Strande JL, Steffen D, Reddy J. Epitope Mapping of SERCA2a Identifies an Antigenic Determinant That Induces Mainly Atrial Myocarditis in A/J Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:523-537. [PMID: 29229678 PMCID: PMC5760440 DOI: 10.4049/jimmunol.1701090] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 11/05/2017] [Indexed: 12/20/2022]
Abstract
Sarcoplasmic/endoplasmic reticulum Ca2+ adenosine triphosphatase (SERCA)2a, a critical regulator of calcium homeostasis, is known to be decreased in heart failure. Patients with myocarditis or dilated cardiomyopathy develop autoantibodies to SERCA2a suggesting that they may have pathogenetic significance. In this report, we describe epitope mapping analysis of SERCA2a in A/J mice that leads us to make five observations: 1) SERCA2a contains multiple T cell epitopes that induce varying degrees of myocarditis. One epitope, SERCA2a 971-990, induces widespread atrial inflammation without affecting noncardiac tissues; the cardiac abnormalities could be noninvasively captured by echocardiography, electrocardiography, and magnetic resonance microscopy imaging. 2) SERCA2a 971-990-induced disease was associated with the induction of CD4 T cell responses and the epitope preferentially binds MHC class II/IAk rather than IEk By creating IAk/and IEk/SERCA2a 971-990 dextramers, the T cell responses were determined by flow cytometry to be Ag specific. 3) SERCA2a 971-990-sensitized T cells produce both Th1 and Th17 cytokines. 4) Animals immunized with SERCA2a 971-990 showed Ag-specific Abs with enhanced production of IgG2a and IgG2b isotypes, suggesting that SERCA2a 971-990 can potentially act as a common epitope for both T cells and B cells. 5) Finally, SERCA2a 971-990-sensitized T cells were able to transfer disease to naive recipients. Together, these data indicate that SERCA2a is a critical autoantigen in the mediation of atrial inflammation in mice and that our model may be helpful to study the inflammatory events that underlie the development of conditions such as atrial fibrillation in humans.
Collapse
Affiliation(s)
- Bharathi Krishnan
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583
| | - Chandirasegaran Massilamany
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892
| | - Rakesh H Basavalingappa
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583
| | - Arunakumar Gangaplara
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Rajkumar A Rajasekaran
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583
| | | | - Vahid Khalilzad-Sharghi
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583
| | - You Zhou
- Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE 68588
| | | | - Shyam S Nandi
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198; and
| | - Paras K Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198; and
| | - Raymond A Sobel
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94304
| | | | - David Steffen
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583
| | - Jay Reddy
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583;
| |
Collapse
|
28
|
Green Tea Polyphenol Epigallocatechin-3-gallate-Alleviated Coxsackievirus B3-induced Myocarditis Through Inhibiting Viral Replication but Not Through Inhibiting Inflammatory Responses. J Cardiovasc Pharmacol 2017; 69:41-47. [PMID: 27753702 DOI: 10.1097/fjc.0000000000000439] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Viral myocarditis, which is mainly caused by coxsackievirus B3 (CVB3), affects about 5%-20% of the world population and still lacks efficient treatments. Green tea, a tonic and healthful beverage that was originated in ancient China, has been receiving considerable attention for its protective effect on cardiovascular diseases in recent years. In the present investigation, we aimed to explore the effect of green tea polyphenol epigallocatechin-3-gallate (EGCG) on CVB3-induced myocarditis and its underlying mechanism. Our study showed that EGCG could alleviate CVB3-induced myocarditis as evidenced by less cardiac injury and higher survival rate. Furthermore, we found that EGCG failed to downregulate the expression of inflammatory cytokines but could significantly inhibit the replication of CVB3. Furthermore, we found that EGCG treatment could downregulate the protein expression level of coxsackievirus and adenovirus receptor, the major receptor for CVB3 to infect cardiac myocytes. In conclusion, our data indicated that EGCG could ameliorate CVB3-induced myocarditis through inhibiting viral replication, which might provide a potential novel therapeutic strategy for viral myocarditis.
Collapse
|
29
|
Jiang Y, Zhu Y, Mu Q, Luo H, Zhi Y, Shen X. Oxymatrine provides protection against Coxsackievirus B3-induced myocarditis in BALB/c mice. Antiviral Res 2017; 141:133-139. [PMID: 28115196 DOI: 10.1016/j.antiviral.2017.01.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 12/21/2016] [Accepted: 01/18/2017] [Indexed: 01/25/2023]
Abstract
Oxymatrine is the primary pharmacological component of Sophora flavescens Ait. In the present study, we investigated the protective effect of oxymatrine against Coxsackievirus B3-induced myocarditis in mice. Coxsackievirus B3-infected HeLa cells were treated with oxymatrine and the viral titer, as well as the degree of cellular proliferation were determined. Additionally, BALB/c mice were infected with Coxsackievirus B3 and received differing concentrations of oxymatrine. On days 5 and 12 following treatment, mice were sacrificed, and serum lactate dehydrogenase, creatine kinase-MB isozyme, and tumor necrosis factor-α levels were quantified. The heart index and degree of myocardial tissue inflammation were also assessed. On day 5, the Coxsackievirus B3 TCID50 values of the heart tissue, and the expression of NTR, IFN-γ, and TNF-α genes in the myocardial tissue were measured. Our results showed that oxymatrine exhibits potent antiviral effects on Coxsackievirus B3 as 50% inhibition was achieved at a concentration as low as 0.238 mg/mL. Oxymatrine markedly reduced the viral titer and inhibited cardiac myocyte pathology exhibited in viral myocarditis. Furthermore, oxymatrine treatment reduced the expression of Coxsackievirus B3 NTR and mouse TNF-α genes compared to the controls. Therefore, our findings indicate that oxymatrine is a promising potent antiviral agent against Coxsackievirus B3-induced myocarditis.
Collapse
Affiliation(s)
- Yan Jiang
- Department of Microbiology and Immunology of Guizhou Medical University Affiliated Hospital, Guiyang 550004, China; Clinical Research Center, Guizhou Medical University Affiliated Hospital, Guiyang 550004, China
| | - Yanxin Zhu
- Department of Microbiology and Immunology of Guizhou Medical University Affiliated Hospital, Guiyang 550004, China
| | - Qiuju Mu
- Department of Microbiology and Immunology of Guizhou Medical University Affiliated Hospital, Guiyang 550004, China
| | - Hong Luo
- Department of Microbiology and Immunology of Guizhou Medical University Affiliated Hospital, Guiyang 550004, China
| | - Yan Zhi
- Department of Microbiology and Immunology of Guizhou Medical University Affiliated Hospital, Guiyang 550004, China
| | - Xiangchun Shen
- Key Laboratory of Optimal Utilization of Natural Medicinal Resources, Guizhou Medical University, Guiyang 550004, China.
| |
Collapse
|
30
|
Li M, Su Y, Yu Y, Yu Y, Wang X, Zou Y, Ge J, Chen R. Dual roles of calpain in facilitating Coxsackievirus B3 replication and prompting inflammation in acute myocarditis. Int J Cardiol 2016; 221:1123-31. [PMID: 27472894 PMCID: PMC7114300 DOI: 10.1016/j.ijcard.2016.07.121] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 06/17/2016] [Accepted: 07/08/2016] [Indexed: 01/14/2023]
Abstract
Background Viral myocarditis (VMC) treatment has long been lacking of effective methods. Our former studies indicated roles of calpain in VMC pathogenesis. This study aimed at verifying the potential of calpain in Coxsackievirus B3 (CVB3)-induced myocarditis treatment. Methods A transgenic mouse overexpressing the endogenous calpain inhibitor, calpastatin, was introduced in the study. VMC mouse model was established via intraperitoneal injection of CVB3 in transgenic and wild mouse respectively. Myocardial injury was assayed histologically (HE staining and pathology grading) and serologically (myocardial damage markers of CK-MB and cTnI). CVB3 replication was observed in vivo and in vitro via the capsid protein VP1 detection or virus titration. Inflammation/fibrotic factors of MPO, perforin, IFNγ, IL17, Smad3 and MMP2 were evaluated using western blot or immunohistology stain. Role of calpain in regulating fibroblast migration was studied in scratch assays. Results Calpastatin overexpression ameliorated myocardial injury induced by CVB3 infection significantly in transgenic mouse indicated by reduced peripheral CK-MB and cTnI levels and improved histology injury. Comparing with CVB3-infected wild type mouse, the transgenic mouse heart tissue carried lower virus load. The inflammation factors of MPO, perforin, IFNγ and IL17 were down-regulated accompanied with fibrotic agents of Smad3 and MMP2 inhibition. And calpain participated in the migration of fibroblasts in vitro, which further proves its role in regulating fibrosis. Conclusion Calpain plays dual roles of facilitating CVB3 replication and inflammation promotion. Calpain inhibition in CVB3-induced myocarditis showed significant treatment effect. Calpain might be a novel target for VMC treatment in clinical practices. Calpain is involved in virus replication in myocarditis. Calpain mediates inflammation infiltration in myocarditis. Calpain might be a candidate for viral myocarditis treatment.
Collapse
Affiliation(s)
- Minghui Li
- Department of Cardiovascular Diseases, Key Laboratory of Viral Heart Diseases, Ministry of Public Health, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yangang Su
- Department of Cardiovascular Diseases, Key Laboratory of Viral Heart Diseases, Ministry of Public Health, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yong Yu
- Department of Cardiovascular Diseases, Key Laboratory of Viral Heart Diseases, Ministry of Public Health, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ying Yu
- Department of Cardiovascular Diseases, Key Laboratory of Viral Heart Diseases, Ministry of Public Health, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xinggang Wang
- Department of Cardiovascular Diseases, Key Laboratory of Viral Heart Diseases, Ministry of Public Health, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yunzeng Zou
- Department of Cardiovascular Diseases, Key Laboratory of Viral Heart Diseases, Ministry of Public Health, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Junbo Ge
- Department of Cardiovascular Diseases, Key Laboratory of Viral Heart Diseases, Ministry of Public Health, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Ruizhen Chen
- Department of Cardiovascular Diseases, Key Laboratory of Viral Heart Diseases, Ministry of Public Health, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
31
|
Zhao Z, Cai TZ, Lu Y, Liu WJ, Cheng ML, Ji YQ. Coxsackievirus B3 induces viral myocarditis by upregulating toll-like receptor 4 expression. BIOCHEMISTRY (MOSCOW) 2016; 80:455-62. [PMID: 25869363 DOI: 10.1134/s0006297915040094] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
In the present study, we investigated the potential pathogenesis of coxsackievirus B3 (CVB3)-induced viral myocarditis and the promising protective effect of silencing RNA (small interfering RNA, siRNA). One hundred and twenty mice were included in the study, and 30 mice were intraperitoneally inoculated with CVB3 to establish an acute viral myocarditis model. The survival rate was observed for the CVB3-infected mouse model (MOD), and myocardial injury was examined by HE (hematoxylin and eosin) staining assay. Real-time PCR (RT-PCR) and Western blot assay were selected to detect the toll-like receptor 4 (TLR4) expression in myocardial tissues. The TLR4 gene was silenced for the MOD mice, and the effects of this treatment were observed. The results indicate that the expression of TLR4 mRNA and the protein significantly and persistently increased during the progression of CVB3-induced myocarditis. The activities of cardiac enzymes including CK, LDH, AST, and CK-MB were also enhanced in CVB3-induced myocardial tissues. Interestingly, when the TLR4 gene was silenced, the CVB3-induced TLR4 production was significantly decreased and the severity of myocarditis was significantly lessened. In conclusion, CVB3 may induce viral myocarditis by upregulating toll-like receptor 4 expression. The viral myocarditis can be ameliorated by silencing the TLR4 gene in the CVB3 viral myocarditis model, which may be a feasible therapeutic method for treatment of viral myocarditis.
Collapse
Affiliation(s)
- Zhao Zhao
- Department of Cardiovascular Medicine, First Hospital of Xi'an, Xi'an, 710002, China.
| | | | | | | | | | | |
Collapse
|
32
|
Zhu H, Lou C, Liu P. Interleukin-27 ameliorates coxsackievirus-B3-induced viral myocarditis by inhibiting Th17 cells. Virol J 2015; 12:189. [PMID: 26578236 PMCID: PMC4650905 DOI: 10.1186/s12985-015-0418-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 11/04/2015] [Indexed: 11/22/2022] Open
Abstract
Background Interleukin (IL)-27, which has both pro and anti- inflammatory properties, is a new discovered heterodimeric cytokine that belongs to IL-12 family. However, the expression pattern and functional role of IL-27 in viral myocarditis (VMC) has not been investigated. Methods BALB/c mice were intraperitoneally (i.p) infected with Coxsackie virus B3 (CVB3) for establishing VMC models. Mice were then injected i.p. with Anti-Mouse IL-27 p28Ab or recombinant IL-27 for neutralization and overexpression of IL-27. The survival rates of mice were recorded and the kinetics of IL-27 expression, the frequencies of Th17 cells and the expression of inflammatory cytokine in CVB3-infected mice were determined by ELISA, real-time PCR and flow cytometry. Results The IL-27 expression in heart tissues and serum in coxsackievirus B3 (CVB3)-induced myocarditis mice peaked on day 4 but then rapidly decreased during the late infectious stage of CVB3, high IL-27 levels were negatively correlated with bodyweight loss (r = −0.71, P = 0.021) and myocardial pathological score (r = −0.85, P = 0.0018). Additionally, neutralization of IL-27 with Anti-IL-27 Ab accelerated, whereas systemic administration of recombinant mouse IL-27 ameliorated CVB3-induced myocarditis. The protective role of IL-27 in VMC was reflected by an improved survival rate, increased bodyweights, and reduced pathological scores in Anti-IL-27 group compared with IgG control group. Mechanistic investigations showed that IL-27 inhibited Th17 cells frequencies and IL-17 production, as well as the Th17-related proinflammatory cytokines in heart tissues. Conclusions Our results demonstrate that that IL-27 effectively protects the myocardium from the pathogenesis of CVB3 induced myocarditis, which may be attributable to reduced Th17 production. IL-27 might serve as a novel therapeutic treatment for VMC. Electronic supplementary material The online version of this article (doi:10.1186/s12985-015-0418-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hengshan Zhu
- Department of Cardiovascular Surgery, Xianyang Hospital of Yan'an University, Xianyang, 712000, Shaanxi, China.
| | - Chuang Lou
- Department of Cardiology, AnKang Hospital of Traditional Chinese Medicine, Ankang, 725000, Shaanxi, China.
| | - Ping Liu
- Department of Cardiology, Xianyang Hospital of Yan'an University, No 38 Wenlin Raod, Xianyang, 712000, Shaanxi, China.
| |
Collapse
|
33
|
Inhibition of autoimmune Chagas-like heart disease by bone marrow transplantation. PLoS Negl Trop Dis 2014; 8:e3384. [PMID: 25521296 PMCID: PMC4270743 DOI: 10.1371/journal.pntd.0003384] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 10/30/2014] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Infection with the protozoan Trypanosoma cruzi manifests in mammals as Chagas heart disease. The treatment available for chagasic cardiomyopathy is unsatisfactory. METHODS/PRINCIPAL FINDINGS To study the disease pathology and its inhibition, we employed a syngeneic chicken model refractory to T. cruzi in which chickens hatched from T. cruzi inoculated eggs retained parasite kDNA (1.4 kb) minicircles. Southern blotting with EcoRI genomic DNA digests revealed main 18 and 20 kb bands by hybridization with a radiolabeled minicircle sequence. Breeding these chickens generated kDNA-mutated F1, F2, and F3 progeny. A targeted-primer TAIL-PCR (tpTAIL-PCR) technique was employed to detect the kDNA integrations. Histocompatible reporter heart grafts were used to detect ongoing inflammatory cardiomyopathy in kDNA-mutated chickens. Fluorochromes were used to label bone marrow CD3+, CD28+, and CD45+ precursors of the thymus-dependent CD8α+ and CD8β+ effector cells that expressed TCRγδ, vβ1 and vβ2 receptors, which infiltrated the adult hearts and the reporter heart grafts. CONCLUSIONS/SIGNIFICANCE Genome modifications in kDNA-mutated chickens can be associated with disruption of immune tolerance to compatible heart grafts and with rejection of the adult host's heart and reporter graft, as well as tissue destruction by effector lymphocytes. Autoimmune heart rejection was largely observed in chickens with kDNA mutations in retrotransposons and in coding genes with roles in cell structure, metabolism, growth, and differentiation. Moreover, killing the sick kDNA-mutated bone marrow cells with cytostatic and anti-folate drugs and transplanting healthy marrow cells inhibited heart rejection. We report here for the first time that healthy bone marrow cells inhibited heart pathology in kDNA+ chickens and thus prevented the genetically driven clinical manifestations of the disease.
Collapse
|
34
|
Lymphocytes Infiltrate the Quadriceps Muscle in Lymphocytic Myocarditis Patients: A Potential New Diagnostic Tool. Can J Cardiol 2014; 30:1547-54. [DOI: 10.1016/j.cjca.2014.07.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 07/17/2014] [Accepted: 07/17/2014] [Indexed: 11/21/2022] Open
|
35
|
Fett JD. Peripartum cardiomyopathy: A puzzle closer to solution. World J Cardiol 2014; 6:87-99. [PMID: 24669290 PMCID: PMC3964190 DOI: 10.4330/wjc.v6.i3.87] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 12/28/2013] [Accepted: 02/18/2014] [Indexed: 02/06/2023] Open
Abstract
Peripartum cardiomyopathy (PPCM) represents new heart failure in a previously heart-healthy peripartum patient. It is necessary to rule out all other known causes of heart failure before accepting a diagnosis of PPCM. The modern era for PPCM in the United States and beyond began with the report of the National Institutes of Health PPCM Workshop in 2000, clarifying all then-currently known aspects of the disease. Since then, hundreds of publications have appeared, an indication of how devastating this disease can be to young mothers and their families and the urgent desire to find solutions for its cause and better treatment. The purpose of this review is to highlight the important advances that have brought us nearer to the solution of this puzzle, focusing on what we have learned about PPCM since 2000; and what still remains unanswered. Despite many improvements in outcome, we still do not know the actual triggers that initiate the pathological process; but realize that cardiac angiogenic imbalances resulting from complex pregnancy-related immune system and hormonal changes play a key role.
Collapse
|
36
|
|
37
|
Root-Bernstein R. Rethinking Molecular Mimicry in Rheumatic Heart Disease and Autoimmune Myocarditis: Laminin, Collagen IV, CAR, and B1AR as Initial Targets of Disease. Front Pediatr 2014; 2:85. [PMID: 25191648 PMCID: PMC4137453 DOI: 10.3389/fped.2014.00085] [Citation(s) in RCA: 28] [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: 05/30/2014] [Accepted: 07/24/2014] [Indexed: 01/05/2023] Open
Abstract
RATIONALE Molecular mimicry theory (MMT) suggests that epitope mimicry between pathogens and human proteins can activate autoimmune disease. Group A streptococci (GAS) mimics human cardiac myosin in rheumatic heart disease (RHD) and coxsackie viruses (CX) mimic actin in autoimmune myocarditis (AM). But myosin and actin are immunologically inaccessible and unlikely initial targets. Extracellular cardiac proteins that mimic GAS and CX would be more likely. OBJECTIVES To determine whether extracellular cardiac proteins such as coxsackie and adenovirus receptor (CAR), beta 1 adrenergic receptor (B1AR), CD55/DAF, laminin, and collagen IV mimic GAS, CX, and/or cardiac myosin or actin. METHODS BLAST 2.0 and LALIGN searches of the UniProt protein database were employed to identify potential molecular mimics. Quantitative enzyme-linked immunosorbent assay was used to measure antibody cross-reactivity. MEASUREMENTS Similarities were considered to be significant if a sequence contained at least 5 identical amino acids in 10. Antibodies were considered to be cross-reactive if the binding constant had a K d less than 10(-9) M. MAIN RESULTS Group A streptococci mimics laminin, CAR, and myosin. CX mimics actin and collagen IV and B1AR. The similarity search results are mirrored by antibody cross-reactivities. Additionally, antibodies against laminin recognize antibodies against collagen IV; antibodies against actin recognize antibodies against myosin, and antibodies against GAS recognize antibodies against CX. Thus, there is both mimicry of extracellular proteins and antigenic complementarity between GAS-CX in RHD/AM. CONCLUSION Rheumatic heart disease/AM may be due to combined infections of GAS with CX localized at cardiomyocytes that may produce a synergistic, hyperinflammatory response that cross-reacts with laminin, collagen IV, CAR, and/or B1AR. Epitope drift shifts the immune response to myosin and actin after cardiomyocytes become damaged.
Collapse
|
38
|
The role of sex differences in autophagy in the heart during coxsackievirus B3-induced myocarditis. J Cardiovasc Transl Res 2013; 7:182-91. [PMID: 24323874 DOI: 10.1007/s12265-013-9525-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 11/20/2013] [Indexed: 12/28/2022]
Abstract
Under normal conditions, autophagy maintains cardiomyocyte health and integrity through turnover of organelles. During stress, oxygen and nutrient deprivation, or microbial infection, autophagy prolongs cardiomyocyte survival. Sex differences in induction of cell death may to some extent explain the disparity between the sexes in many human diseases. However, sex differences in gene expression, which regulate cell death and autophagy, were so far not taken in consideration to explain the sex bias of viral myocarditis. Coxsackievirus B3 (CVB3)-induced myocarditis is a sex-biased disease, with females being substantially less susceptible than males and sex hormones largely determine this bias. CVB3 was shown to induce and subvert the autophagosome for its optimal viral RNA replication. Gene expression analysis on mouse and human, healthy and CVB3-infected, cardiac samples of both sexes, suggests sex differences in autophagy-related gene expression. This review discusses the aspects of sex bias in autophagy induction in cardiomyocytes.
Collapse
|
39
|
Lecuit M, Eloit M. The human virome: new tools and concepts. Trends Microbiol 2013; 21:510-5. [PMID: 23906500 PMCID: PMC7172527 DOI: 10.1016/j.tim.2013.07.001] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 06/15/2013] [Accepted: 07/01/2013] [Indexed: 02/07/2023]
Abstract
New sequencing technologies increase our knowledge regarding the composition of the human virome. There are beneficial and detrimental viruses. The human virome can impact upon health at body surfaces (skin or gut) and within tissues. Some animal viruses are transmitted via the oral route by consumption of food.
The human virome is the viral component of the microbiome. Its composition, and interindividual and temporal variability are not precisely known. Its impact on human health has received less attention than that of the bacterial microbiome, but is likely to be equally important, both in homeostasis and disease. Here we review the recent advances in this field and the questions that arise in the context of our rapidly increasing knowledge regarding the composition and function of the human virome. With the ever-extending use of next-generation sequencing (NGS) on a variety of clinical samples, rapid progress on the composition of the human virome and its impact upon human health are to be expected in the coming years.
Collapse
Affiliation(s)
- Marc Lecuit
- Institut Pasteur, Biology of Infection Unit, 75724 Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 1117, Paris, France; Paris Descartes University, Sorbonne Paris Cité, Institut Imagine, Paris, France; Necker-Enfants Malades University Hospital, Division of Infectious Diseases and Tropical Medicine, Paris, France
| | | |
Collapse
|