1
|
Fankhauser RG, Johnson DB, Moslehi JJ, Balko JM. Preclinical mouse models of immune checkpoint inhibitor-associated myocarditis. NATURE CARDIOVASCULAR RESEARCH 2025; 4:526-538. [PMID: 40335724 DOI: 10.1038/s44161-025-00640-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 03/10/2025] [Indexed: 05/09/2025]
Abstract
In this Review, we present a comprehensive analysis of preclinical models used to study immune checkpoint inhibitor-associated myocarditis (hereafter ICI-myocarditis), a potentially lethal immune-related adverse event. We begin by providing an overview of immune checkpoint inhibitors, highlighting how their efficacy in cancer treatment is counterbalanced by their predisposition to cause immune-related adverse events. Next, we draw from human data to identify disease features that an effective mouse model should ideally mimic. After that, we present a critical evaluation of a wide variety of existing mouse models including genetic, pharmacological and humanized models. We summarize insights gathered about the underlying mechanisms of ICI-myocarditis and the role of mouse models in these discoveries. We conclude with a perspective on the future of preclinical models, highlighting potential model improvements and research directions that could strengthen our understanding of ICI-myocarditis, ultimately improving patient outcomes.
Collapse
Grants
- 5R01HL156021-04 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 5R01HL155990-04 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 5R01HL141466-05 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- NIH P01 HL141084 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- NIH R01 HL160688 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 5R01CA227481-05 U.S. Department of Health & Human Services | NIH | National Cancer Institute (NCI)
- 5P30CA068485-29 U.S. Department of Health & Human Services | NIH | National Cancer Institute (NCI)
- T32GM007347 U.S. Department of Health & Human Services | NIH | National Institute of General Medical Sciences (NIGMS)
- 25PRE1375723 American Heart Association (American Heart Association, Inc.)
Collapse
Affiliation(s)
- Reilly G Fankhauser
- Medical Scientist Training Program, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Douglas B Johnson
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Javid J Moslehi
- Section of Cardio-Oncology and Immunology, Cardiovascular Research Institute, University of California San Francisco School of Medicine, San Francisco, CA, USA
| | - Justin M Balko
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.
- Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Nashville, TN, USA.
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
2
|
Myers JM, Sandel C, Alvarez K, Garman L, Wiley G, Montgomery C, Gaffney P, Stavrakis S, Fairweather D, Bruno KA, Zhao YD, Cooper LT, Cunningham MW. Cardiac autoantibodies promote a fibrotic transcriptome and reduced ventricular recovery in human myocarditis. Front Immunol 2025; 16:1500909. [PMID: 40181955 PMCID: PMC11965655 DOI: 10.3389/fimmu.2025.1500909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
Myocarditis leads to dilated cardiomyopathy (DCM) with one-third failing to recover normal ejection fraction (EF 50%). Our previous studies have supported a Th17 autoimmune pathogenesis where IL17A and IL-6 are elevated in myocarditis patients who do not recover normal EF. In the non-recovered group, autoantibody mechanisms of pathogenesis in myocardial injury and systolic dysfunction are not fully understood. Furthermore, in our myocarditis cohort, cardiac myosin (CM) autoantibodies (AAbs) were elevated and cross-reactive with the β-adrenergic receptor (βAR). Here we studied cross-reactive CM/βAR serum AAbs and human myocarditis-derived monoclonal antibodies (mAbs) to define their potential pathogenic mechanisms and to identify unique human CM epitopes associated with non-recovery in a longitudinal (n=41) cohort. Elevated CM IgG AAbs in the non-recovered phenotype correlated with reduced EF and poor outcomes. Human CM epitopes unique to the non-recovered phenotype shared strong amino acid sequence homology with extracellular loops of βARs and supported molecular mimicry and cross-reactivity between CM and βAR. Myocarditis-derived IgG and human mAb 2C.4 activated protein kinase A (PKA) in an IgG, CM, and βAR-dependent manner in H9c2 heart myoblast cell line, and transcriptomic analysis revealed mAb 2C.4 induced fibrosis pathways which were highly similar pathways seen with isoproterenol, a beta receptor agonist. Our data translate into new mechanistic insights from our small longitudinal group of myocarditis/DCM patients and into potential therapeutic targets and biomarkers for future studies.
Collapse
Affiliation(s)
- Jennifer M. Myers
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Clayton Sandel
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Kathy Alvarez
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Lori Garman
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Graham Wiley
- Clinical Genomics Core, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Courtney Montgomery
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Patrick Gaffney
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Stavros Stavrakis
- Department of Cardiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - DeLisa Fairweather
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Katelyn A. Bruno
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Yan Daniel Zhao
- Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Leslie T. Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Madeleine W. Cunningham
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
3
|
Zhao Q, Li Z, Wang DW, Li F, Zhang L, Jiang J. Lactate Dehydrogenase Indicates Development From Fulminant Myocarditis to Chronic Persistent Myocarditis: A Multicenter Retrospective Cohort Study in China. Int J Gen Med 2025; 18:1333-1344. [PMID: 40078751 PMCID: PMC11897539 DOI: 10.2147/ijgm.s509291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Background Fulminant myocarditis (FM) is a critical manifestation of myocarditis. However, the clinical features and risk factors associated with its adverse outcomes are not fully understood. Given the high mortality and potential for long-term complications, it is crucial to identify factors that could predict the progression of FM to chronic persistent myocarditis. We hypothesize some clinical or laboratory markers may be predictive of this progression. This study aims to identify clinical factors that may help predict the progress of FM to chronic persistent myocarditis. Methods A total of 82 patients with FM treated based on Chinese protocol were included. Kaplan-Meier curve and regression analysis were used to determine the clinical features and prognostic predictors of chronic persistent myocarditis in patients with 24 months of follow-up. Results Chronic persistent myocarditis was observed in 20 patients during the follow-up. ROC curve showed that the critical value for chronic persistent myocarditis caused by lactate dehydrogenase (LDH) was 577.00U/L (sensitivity 75.0%, specificity 74.2%). Time from onset to admission over 6 days and LDH > 577.00U/L were identified as risk factors for chronic persistent myocarditis in patients with FM in both univariate and multivariate cox analysis. The hazard ratio and 95% Confidence intervals were 3.35 (1.32-8.50) (p = 0.011) and 6.11 (2.02-18.48) (p < 0.001), respectively. The per standard deviation of increment in LDH was associated with the 55% (1.55, 1.11-2.18) in HR and 95% CI of the occurrence of chronic persistent myocarditis. Conclusion About 24.4% of the patients with FM treated based on the life support measures proposed in the consensus of Chinese Society of Cardiology have been observed chronic persistent myocarditis. Time from onset to admission over 6 days and LDH levels >577.00 U/L at admission may serve as risk factors for the progression from FM to chronic persistent myocarditis.
Collapse
Affiliation(s)
- Qu Zhao
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, People’s Republic of China
| | - Zeping Li
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, People’s Republic of China
| | - Dao Wen Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, People’s Republic of China
| | - Fan Li
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, People’s Republic of China
| | - Li Zhang
- Division of Cardiology, Minda Hospital of Hubei Minzu University, Enshi, 445099, People’s Republic of China
| | - Jiangang Jiang
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, People’s Republic of China
| |
Collapse
|
4
|
Di Florio DN, Weigel GJ, Gorelov DJ, McCabe EJ, Beetler DJ, Shapiro KA, Bruno KA, Chekuri I, Jain A, Whelan ER, Salomon GR, Khatib S, Bonvie-Hill NE, Fliess JJ, Giresi PG, Hamilton C, Hartmoyer CJ, Balamurugan V, Darakjian AA, Edenfield BH, Kocsis SC, McLeod CJ, Cooper LT, Audet-Walsh É, Coronado MJ, Sin J, Fairweather D. Sex differences in mitochondrial gene expression during viral myocarditis. Biol Sex Differ 2024; 15:104. [PMID: 39696682 PMCID: PMC11657264 DOI: 10.1186/s13293-024-00678-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 11/20/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Myocarditis is an inflammation of the heart muscle most often caused by viral infections. Sex differences in the immune response during myocarditis have been well described but upstream mechanisms in the heart that might influence sex differences in disease are not completely understood. METHODS Male and female BALB/c wild type mice received an intraperitoneal injection of heart-passaged coxsackievirus B3 (CVB3) or vehicle control. Bulk-tissue RNA-sequencing was conducted to better understand sex differences in CVB3 myocarditis. We performed enrichment analysis and functional validation to understand sex differences in the transcriptional landscape of myocarditis and identify factors that might drive sex differences in myocarditis. RESULTS As expected, the hearts of male and female mice with myocarditis were significantly enriched for pathways related to an innate and adaptive immune response compared to uninfected controls. Unique to this study, we found that males were enriched for inflammatory pathways and gene changes that suggested worse mitochondrial electron transport function while females were enriched for pathways related to mitochondrial homeostasis. Mitochondria isolated from the heart of males were confirmed to have worse mitochondrial respiration than females during myocarditis. Unbiased TRANSFAC analysis identified estrogen-related receptor alpha (ERRα) as a transcription factor that may mediate sex differences in mitochondrial function during myocarditis. Transcript and protein levels of ERRα were confirmed as elevated in females with myocarditis compared to males. Differential binding analysis from chromatin immunoprecipitation (ChIP) sequencing confirmed that ERRα bound highly to select predicted respiratory chain genes in females more than males during myocarditis. CONCLUSIONS Females with viral myocarditis regulate mitochondrial homeostasis by upregulating master regulators of mitochondrial transcription including ERRα.
Collapse
Affiliation(s)
- Damian N Di Florio
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Gabriel J Weigel
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - David J Gorelov
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Elizabeth J McCabe
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Danielle J Beetler
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Katie A Shapiro
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Katelyn A Bruno
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
- Division of Cardiovascular Medicine, Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Isha Chekuri
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Angita Jain
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Emily R Whelan
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Gary R Salomon
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Sami Khatib
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | | | - Jessica J Fliess
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Presley G Giresi
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Charwan Hamilton
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | | | | | - Ashley A Darakjian
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Brandy H Edenfield
- Department of Cancer Biology, Mayo Clinic Jacksonville, Jacksonville, FL, USA
| | - S Christian Kocsis
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | | | - Leslie T Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Étienne Audet-Walsh
- Endocrinology - Nephrology Research Division, CHU de Québec - Université Laval Research Center, Québec, QC, Canada
| | | | - Jon Sin
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, USA
| | - DeLisa Fairweather
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA.
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, USA.
- Department of Immunology, Mayo Clinic, Jacksonville, FL, USA.
- Department of Medicine, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
5
|
da Silva Cabral T, Cayuela NC, Carvalho KGB, Pimenta TS, Rodrigues APD, Diniz DG, Quaresma JAS, de Almeida Medeiros DB, Prazeres ITE, da Silva SP, Araújo TP, da Costa Vasconcelos PF, Diniz CWP, Diniz JAP. Juruaça virus taxonomy, tolerance and resistance to infection, and inflammatory response modulation in murine model. NPJ VIRUSES 2024; 2:46. [PMID: 40295833 PMCID: PMC11721108 DOI: 10.1038/s44298-024-00056-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 09/05/2024] [Indexed: 04/30/2025]
Abstract
Juruaça virus (JUAV), previously unclassified, was isolated from bats and administered to neonatal and adult BALB/c mice to investigate acute and chronic disease progression. In this study, we conducted genomic sequencing to achieve taxonomic classification and utilized these models to explore the inflammatory response and sickness behavior in both neonatal and adult mice. Neonates received a single intranasal instillation of infected brain homogenate (20 µL), whereas 31-day-old mice were given the same volume intranasally for three consecutive days. Control groups were administered equal volumes of uninfected brain homogenate. Our findings reveal that intranasal JUAV infection-induced acute meningoencephalitis and death in neonates, while adult mice exhibited chronic infection with variable clinical signs, inflammatory mediator production, histopathological changes, and neuropathological features. Interestingly, only some adult mice showed sickness behavior post-infection, and among these, a subset continued to decline and die. The differential tissue damage observed in mice with and without overt disease symptoms suggests mechanisms of resistance or tolerance, where exceeding tolerance capacity resulted in pathological outcomes, including chronic dysfunction or death. This study provides the first evidence of JUAV's capability to infect mammals, demonstrating its distinct impact on bats and variable effects in neonatal and adult mice. We provisionally classified JUAV as closely related to the clade containing tombus-like virus 6 found in mute swan feces. Our research highlights the importance of understanding viral-host interactions and the inflammatory responses that contribute to disease variability, offering insights into tolerance and resistance mechanisms based on inflammatory response modulation.
Collapse
Affiliation(s)
- Tatyane da Silva Cabral
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Avenida Almirante Barroso, 492, Bairro do Marco, CEP 66.093-020, Belém, Pará, Brasil
| | - Natalie Chaves Cayuela
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Avenida Almirante Barroso, 492, Bairro do Marco, CEP 66.093-020, Belém, Pará, Brasil
| | - Karina Glazianne Barbosa Carvalho
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Avenida Almirante Barroso, 492, Bairro do Marco, CEP 66.093-020, Belém, Pará, Brasil
| | - Tamirys Simão Pimenta
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Avenida Almirante Barroso, 492, Bairro do Marco, CEP 66.093-020, Belém, Pará, Brasil
| | - Ana Paula Drummond Rodrigues
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Avenida Almirante Barroso, 492, Bairro do Marco, CEP 66.093-020, Belém, Pará, Brasil
| | - Daniel Guerreiro Diniz
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Avenida Almirante Barroso, 492, Bairro do Marco, CEP 66.093-020, Belém, Pará, Brasil
- Laboratório de Investigações em Neurodegeneração e Infecção, Universidade Federal do Pará, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Rua dos Mundurucus, 4487, Guamá, CEP: 66.073-005, Belém, Pará, Brasil
- Universidade Federal do Pará, Hospital Universitário João de Barros Barreto, Núcleo de Pesquisas em Oncologia, Rua dos Mundurucus, 4487, Guamá, CEP: 66.073-005, Belém, Pará, Brasil
| | - Juarez Antônio Simões Quaresma
- Departamento de Patologia, Universidade do Estado do Pará, Centro de Ciências Biológicas e da Saúde, Belém, Pará, Brasil, Rua do Una, 156, Telégrafo, CEP: 66.050-540, Belém, Pará, Brasil
- Universidade Federal do Pará, Núcleo de Medicina Tropical, Av. Generalíssimo Deodoro, 92 - Umarizal, CEP: 66.055-240, Belém, Pará, Brasil
| | - Daniele Barbosa de Almeida Medeiros
- Instituto Evandro Chagas, Seção de Arbovirologia e Febres Hemorrágicas, Rodovia BR-316 km 7 s/n - Levilândia, CEP: 67.030-000, Ananindeua, Pará, Brasil
| | - Ivy Tsuya Essashika Prazeres
- Instituto Evandro Chagas, Seção de Arbovirologia e Febres Hemorrágicas, Rodovia BR-316 km 7 s/n - Levilândia, CEP: 67.030-000, Ananindeua, Pará, Brasil
| | - Sandro Patroca da Silva
- Instituto Evandro Chagas, Seção de Arbovirologia e Febres Hemorrágicas, Rodovia BR-316 km 7 s/n - Levilândia, CEP: 67.030-000, Ananindeua, Pará, Brasil
| | - Taís Pinheiro Araújo
- Instituto Evandro Chagas, Seção de Arbovirologia e Febres Hemorrágicas, Rodovia BR-316 km 7 s/n - Levilândia, CEP: 67.030-000, Ananindeua, Pará, Brasil
| | - Pedro Fernando da Costa Vasconcelos
- Departamento de Patologia, Universidade do Estado do Pará, Centro de Ciências Biológicas e da Saúde, Belém, Pará, Brasil, Rua do Una, 156, Telégrafo, CEP: 66.050-540, Belém, Pará, Brasil
- Instituto Evandro Chagas, Seção de Arbovirologia e Febres Hemorrágicas, Rodovia BR-316 km 7 s/n - Levilândia, CEP: 67.030-000, Ananindeua, Pará, Brasil
| | - Cristovam Wanderley Picanço Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Universidade Federal do Pará, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Rua dos Mundurucus, 4487, Guamá, CEP: 66.073-005, Belém, Pará, Brasil
| | - José Antonio Picanço Diniz
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Avenida Almirante Barroso, 492, Bairro do Marco, CEP 66.093-020, Belém, Pará, Brasil.
| |
Collapse
|
6
|
Ye J, Xu S, Liu X, Zhang Q, Li X, Zhang H, Ma J, Leng L, Zhang S. Effect and mechanism of T lymphocytes on human induced pluripotent stem cell-derived cardiomyocytes via Proteomics. Stem Cell Res Ther 2024; 15:236. [PMID: 39075540 PMCID: PMC11288085 DOI: 10.1186/s13287-024-03791-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 06/08/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Abnormalities in T cell activation play an important role in the pathogenesis of myocarditis, and persistent T cell responses can lead to autoimmunity and chronic cardiac inflammation, as well as even dilated cardiomyopathy. Although previous work has examined the role of T cells in myocarditis in animal models, the specific mechanism for human cardiomyocytes has not been investigated. METHODS In this study, we constructed the human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and established the T cell-mediated cardiac injury model by co-culturing with activated CD4 + T or CD8 + T cells that were isolated from peripheral mononuclear blood to elucidate the pathogenesis of myocardial cell injury caused by inflammation. RESULTS By combination of quantitative proteomics with tissue and cell immunofluorescence examination, we established a proteome profile of inflammatory myocardia from hiPSC-CMs with obvious cardiomyocyte injury and increased levels of lactate dehydrogenase content, creatine kinase isoenzyme MB and cardiac troponin. A series of molecular dysfunctions of hiPSC-CMs was observed and indicated that CD4 + cells could produce direct cardiomyocyte injury by activating the NOD-like receptor signals pathway. CONCLUSIONS The data presented in our study established a proteome map of inflammatory myocardial based on hiPSC-CMs injury model. These results can provide guidance in the discovery of improved clinical treatments for myocarditis.
Collapse
Affiliation(s)
- Jin Ye
- Stem cell and Regenerative Medicine Lab, Institute of Clinical Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Sichi Xu
- Stem cell and Regenerative Medicine Lab, Institute of Clinical Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Xiaoqing Liu
- Stem cell and Regenerative Medicine Lab, Institute of Clinical Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
- College of Life Sciences, Hebei University, Baoding, 071002, China
| | - Qiyu Zhang
- Stem cell and Regenerative Medicine Lab, Institute of Clinical Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Xiao Li
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Hui Zhang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jie Ma
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China.
| | - Ling Leng
- Stem cell and Regenerative Medicine Lab, Institute of Clinical Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Shuyang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
7
|
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
|
8
|
Di Florio DN, Beetler DJ, McCabe EJ, Sin J, Ikezu T, Fairweather D. Mitochondrial extracellular vesicles, autoimmunity and myocarditis. Front Immunol 2024; 15:1374796. [PMID: 38550582 PMCID: PMC10972887 DOI: 10.3389/fimmu.2024.1374796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 02/28/2024] [Indexed: 04/02/2024] Open
Abstract
For many decades viral infections have been suspected as 'triggers' of autoimmune disease, but mechanisms for how this could occur have been difficult to establish. Recent studies have shown that viral infections that are commonly associated with viral myocarditis and other autoimmune diseases such as coxsackievirus B3 (CVB3) and SARS-CoV-2 target mitochondria and are released from cells in mitochondrial vesicles that are able to activate the innate immune response. Studies have shown that Toll-like receptor (TLR)4 and the inflammasome pathway are activated by mitochondrial components. Autoreactivity against cardiac myosin and heart-specific immune responses that occur after infection with viruses where the heart is not the primary site of infection (e.g., CVB3, SARS-CoV-2) may occur because the heart has the highest density of mitochondria in the body. Evidence exists for autoantibodies against mitochondrial antigens in patients with myocarditis and dilated cardiomyopathy. Defects in tolerance mechanisms like autoimmune regulator gene (AIRE) may further increase the likelihood of autoreactivity against mitochondrial antigens leading to autoimmune disease. The focus of this review is to summarize current literature regarding the role of viral infection in the production of extracellular vesicles containing mitochondria and virus and the development of myocarditis.
Collapse
Affiliation(s)
- Damian N. Di Florio
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Danielle J. Beetler
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Elizabeth J. McCabe
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Jon Sin
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
| | - Tsuneya Ikezu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| | - DeLisa Fairweather
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, United States
- Department of Immunology, Mayo Clinic, Jacksonville, FL, United States
- Department of Medicine, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
9
|
Musigk N, Suwalski P, Golpour A, Fairweather D, Klingel K, Martin P, Frustaci A, Cooper LT, Lüscher TF, Landmesser U, Heidecker B. The inflammatory spectrum of cardiomyopathies. Front Cardiovasc Med 2024; 11:1251780. [PMID: 38464847 PMCID: PMC10921946 DOI: 10.3389/fcvm.2024.1251780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 01/29/2024] [Indexed: 03/12/2024] Open
Abstract
Infiltration of the myocardium with various cell types, cytokines and chemokines plays a crucial role in the pathogenesis of cardiomyopathies including inflammatory cardiomyopathies and myocarditis. A more comprehensive understanding of the precise immune mechanisms involved in acute and chronic myocarditis is essential to develop novel therapeutic approaches. This review offers a comprehensive overview of the current knowledge of the immune landscape in cardiomyopathies based on etiology. It identifies gaps in our knowledge about cardiac inflammation and emphasizes the need for new translational approaches to improve our understanding thus enabling development of novel early detection methods and more effective treatments.
Collapse
Affiliation(s)
- Nicolas Musigk
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
| | - Phillip Suwalski
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
| | - Ainoosh Golpour
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
| | - DeLisa Fairweather
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Department of Environmental Health Sciences and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, United States
| | - Karin Klingel
- Cardiopathology Institute for Pathology, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Pilar Martin
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Centro de Investigación Biomédica en Red Cardiovascular (CIBER-CV, ISCIII), Madrid, Spain
| | | | - Leslie T. Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Thomas F. Lüscher
- GZO-Zurich Regional Health Centre, Wetzikon & Cardioimmunology, Centre for Molecular Cardiology, University of Zurich, Zurich, Switzerland
- Royal Brompton & Harefield Hospitals and National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Ulf Landmesser
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
| | - Bettina Heidecker
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
| |
Collapse
|
10
|
Kanuri SH, Sirrkay PJ. Adjuvants in COVID-19 vaccines: innocent bystanders or culpable abettors for stirring up COVID-heart syndrome. Ther Adv Vaccines Immunother 2024; 12:25151355241228439. [PMID: 38322819 PMCID: PMC10846003 DOI: 10.1177/25151355241228439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 01/05/2024] [Indexed: 02/08/2024] Open
Abstract
COVID-19 infection is a multi-system clinical disorder that was associated with increased morbidity and mortality. Even though antiviral therapies such as Remdesvir offered modest efficacy in reducing the mortality and morbidity, they were not efficacious in reducing the risk of future infections. So, FDA approved COVID-19 vaccines which are widely administered in the general population worldwide. These COVID-19 vaccines offered a safety net against future infections and re-infections. Most of these vaccines contain inactivated virus or spike protein mRNA that are primarily responsible for inducing innate and adaptive immunity. These vaccines were also formulated to contain supplementary adjuvants that are beneficial in boosting the immune response. During the pandemic, clinicians all over the world witnessed an uprise in the incidence and prevalence of cardiovascular diseases (COVID-Heart Syndrome) in patients with and without cardiovascular risk factors. Clinical researchers were not certain about the underlying reason for the upsurge of cardiovascular disorders with some blaming them on COVID-19 infections while others blaming them on COVID-19 vaccines. Based on the literature review, we hypothesize that adjuvants included in the COVID-19 vaccines are the real culprits for causation of cardiovascular disorders. Operation of various pathological signaling events under the influence of these adjuvants including autoimmunity, bystander effect, direct toxicity, anti-phospholipid syndrome (APS), anaphylaxis, hypersensitivity, genetic susceptibility, epitope spreading, and anti-idiotypic antibodies were partially responsible for stirring up the onset of cardiovascular disorders. With these mechanisms in place, a minor contribution from COVID-19 virus itself cannot be ruled out. With that being said, we strongly advocate for careful selection of vaccine adjuvants included in COVID-19 vaccines so that future adverse cardiac disorders can be averted.
Collapse
Affiliation(s)
- Sri Harsha Kanuri
- Research Fellow, Stark Neurosciences Institute, Indiana University School of Medicine, 320 W 15 ST, Indianapolis, IN 46202, USA
| | | |
Collapse
|
11
|
Liu K, Han B. Role of immune cells in the pathogenesis of myocarditis. J Leukoc Biol 2024; 115:253-275. [PMID: 37949833 DOI: 10.1093/jleuko/qiad143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/15/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023] Open
Abstract
Myocarditis is an inflammatory heart disease that mostly affects young people. Myocarditis involves a complex immune network; however, its detailed pathogenesis is currently unclear. The diversity and plasticity of immune cells, either in the peripheral blood or in the heart, have been partially revealed in a number of previous studies involving patients and several kinds of animal models with myocarditis. It is the complexity of immune cells, rather than one cell type that is the culprit. Thus, recognizing the individual intricacies within immune cells in the context of myocarditis pathogenesis and finding the key intersection of the immune network may help in the diagnosis and treatment of this condition. With the vast amount of cell data gained on myocarditis and the recent application of single-cell sequencing, we summarize the multiple functions of currently recognized key immune cells in the pathogenesis of myocarditis to provide an immune background for subsequent investigations.
Collapse
Affiliation(s)
- Keyu Liu
- Department of Pediatric Cardiology, Shandong Provincial Hospital, Shandong University, Cheeloo Colledge of Medicine, No. 324 Jingwu Road, 250021, Jinan, China
| | - Bo Han
- Department of Pediatric Cardiology, Shandong Provincial Hospital, Shandong University, Cheeloo Colledge of Medicine, No. 324 Jingwu Road, 250021, Jinan, China
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 Jingwu Road, 250021, Jinan, China
- Shandong Provincial Hospital, Shandong Provincial Clinical Research Center for Children' s Health and Disease office, No. 324 Jingwu Road, 250021, Jinan, China
| |
Collapse
|
12
|
Stoffers B, Wolf H, Bacmeister L, Kupsch S, Vico T, Marchini T, Brehm MA, Yan I, Becher PM, Ardeshirdavani A, Escher F, Kim SV, Klingel K, Kirchhof P, Blankenberg S, Zeller T, Wolf D, Hilgendorf I, Westermann D, Lindner D. GPR15-mediated T cell recruitment during acute viral myocarditis facilitated virus elimination and improved outcome. NATURE CARDIOVASCULAR RESEARCH 2024; 3:76-93. [PMID: 39195892 PMCID: PMC11357984 DOI: 10.1038/s44161-023-00401-z] [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: 09/07/2022] [Accepted: 11/27/2023] [Indexed: 08/29/2024]
Abstract
Viral myocarditis is characterized by infiltration of mononuclear cells essential for virus elimination. GPR15 has been identified as a homing receptor for regulatory T cells in inflammatory intestine diseases, but its role in inflammatory heart diseases is still elusive. Here we show that GPR15 deficiency impairs coxsackievirus B3 elimination, leading to adverse cardiac remodeling and dysfunction. Delayed recruitment of regulatory T cells in GPR15-deficient mice was accompanied by prolonged persistence of cytotoxic and regulatory T cells. In addition, RNA sequencing revealed prolonged inflammatory response and altered chemotaxis in knockout mice. In line, we identified GPR15 and its ligand GPR15L as an important chemokine receptor-ligand pair for the recruitment of regulatory and cytotoxic T cells. In summary, the insufficient virus elimination might be caused by a delayed recruitment of T cells as well as delayed interferon-γ expression, resulting in a prolonged inflammatory response and an adverse outcome in GPR15-deficient mice.
Collapse
MESH Headings
- Animals
- Myocarditis/immunology
- Myocarditis/metabolism
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/deficiency
- Receptors, G-Protein-Coupled/immunology
- Mice, Knockout
- Coxsackievirus Infections/immunology
- Coxsackievirus Infections/genetics
- Disease Models, Animal
- Enterovirus B, Human/immunology
- Mice, Inbred C57BL
- T-Lymphocytes, Regulatory/immunology
- Acute Disease
- Interferon-gamma/metabolism
- Mice
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- Male
- Chemotaxis, Leukocyte/genetics
- Chemotaxis, Leukocyte/immunology
- Myocardium/metabolism
- Myocardium/immunology
- Myocardium/pathology
- Signal Transduction
Collapse
Affiliation(s)
- Bastian Stoffers
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Hanna Wolf
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Lucas Bacmeister
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Svenja Kupsch
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tamara Vico
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Timoteo Marchini
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maria A Brehm
- Department Digital Health Sciences and Biomedicine, School of Life Sciences, University of Siegen, Siegen, Germany
| | - Isabell Yan
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Department of Cardiology, University Heart & Vascular Centre Hamburg, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - P Moritz Becher
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Department of Cardiology, University Heart & Vascular Centre Hamburg, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Armin Ardeshirdavani
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Felicitas Escher
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Institute for Cardiac Diagnostics and Therapy, Berlin, Germany
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Campus Virchow Klinikum, Berlin, Germany
| | - Sangwon V Kim
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Karin Klingel
- Cardiopathology, Institute of Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Paulus Kirchhof
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Department of Cardiology, University Heart & Vascular Centre Hamburg, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Blankenberg
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Department of Cardiology, University Heart & Vascular Centre Hamburg, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Tanja Zeller
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Department of Cardiology, University Heart & Vascular Centre Hamburg, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Dennis Wolf
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ingo Hilgendorf
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dirk Westermann
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Diana Lindner
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| |
Collapse
|
13
|
Di Florio D, Gorelov D, McCabe E, Beetler D, Shapiro K, Bruno K, Chekuri I, Jain A, Whelan E, Salomon G, Khatib S, Bonvie-Hill N, Giresi P, Balamurugan V, Weigel G, Fliess J, Darakjian A, Edenfield B, Kocsis C, McLeod C, Cooper L, Audet-Walsh E, Coronado M, Sin J, Fairweather D. Sex differences in mitochondrial gene expression during viral myocarditis. RESEARCH SQUARE 2023:rs.3.rs-3716881. [PMID: 38196574 PMCID: PMC10775395 DOI: 10.21203/rs.3.rs-3716881/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Background Myocarditis is an inflammation of the heart muscle most often caused by an immune response to viral infections. Sex differences in the immune response during myocarditis have been well described but upstream mechanisms in the heart that might influence sex differences in disease are not completely understood. Methods Male and female BALB/c wild type mice received an intraperitoneal injection of heart-passaged coxsackievirus B3 (CVB3) or vehicle control. Bulk-tissue RNA-sequencing was conducted to better understand sex differences in CVB3 myocarditis. We performed enrichment analysis to understand sex differences in the transcriptional landscape of myocarditis and identify candidate transcription factors that might drive sex differences in myocarditis. Results The hearts of male and female mice with myocarditis were significantly enriched for pathways related to an innate and adaptive immune response compared to uninfected controls. When comparing females to males with myocarditis, males were enriched for inflammatory pathways and gene changes that suggested worse mitochondrial transcriptional support (e.g., mitochondrial electron transport genes). In contrast, females were enriched for pathways related to mitochondrial respiration and bioenergetics, which were confirmed by higher transcript levels of master regulators of mitochondrial function including peroxisome proliferator-activated receptor gamma coactivator 1 (PGC1α), nuclear respiratory factor 1 (NRF1) and estrogen-related receptor alpha (ERRα). TRANSFAC analysis identified ERRa as a transcription factor that may mediate sex differences in mitochondrial function during myocarditis. Conclusions Master regulators of mitochondrial function were elevated in females with myocarditis compared to males and may promote sex differences in mitochondrial respiratory transcript expression during viral myocarditis resulting in less severe myocarditis in females following viral infection.
Collapse
|
14
|
Di Florio DN, Macomb LP, Giresi PG, Beetler DJ, Bonvie-Hill NE, Shapiro KA, Naser ARN, Khatib S, Whelan ER, Weigel GJ, Edenfield BH, Balamurugan V, Burris SK, Rich LJ, Bruno KA, Cooper LT, McLeod CJ, Yamani MH, Fairweather D. Sex differences in left-ventricular strain in a murine model of coxsackievirus B3 myocarditis. iScience 2023; 26:108493. [PMID: 38146431 PMCID: PMC10749256 DOI: 10.1016/j.isci.2023.108493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 10/02/2023] [Accepted: 11/16/2023] [Indexed: 12/27/2023] Open
Abstract
Myocarditis is typically caused by viral infections, but most cases are thought to be subclinical. Echocardiography is often used for initial assessment of myocarditis patients but is poor at detecting subtle changes in cardiac dysfunction. Cardiac strain, such as global longitudinal strain (GLS) and global circumferential strain (GCS), represents an increasingly used set of measurements which can detect these subtle changes. Using a murine model of coxsackievirus B3 myocarditis, we characterized functional changes in the heart using echocardiography during myocarditis and by sex. We found that 2D GLS, 4D mode, and 4D strains detected a significant reduction in ejection fraction and GLS during myocarditis compared to baseline and in males compared to females. Furthermore, worse GLS correlated to increased levels of CD45+, CD11b+, and CD3+ immune cells. Our findings closely resemble published reports of GLS in patients with myocarditis indicating the usefulness of this animal model for translational studies of myocarditis.
Collapse
Affiliation(s)
- Damian N. Di Florio
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, USA
| | - Logan P. Macomb
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Presley G. Giresi
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Danielle J. Beetler
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, USA
| | | | - Katie A. Shapiro
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | | | - Sami Khatib
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Emily R. Whelan
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, USA
| | - Gabriel J. Weigel
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | | | | | | | | | - Katelyn A. Bruno
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
- Division of Cardiovascular Medicine, Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Leslie T. Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Chris J. McLeod
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Mohamad H. Yamani
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - DeLisa Fairweather
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, USA
- Department of Immunology, Mayo Clinic, Jacksonville, FL, USA
- Department of Medicine, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
15
|
Mone K, Reddy J. The knowns and unknowns of cardiac autoimmunity in viral myocarditis. Rev Med Virol 2023; 33:e2478. [PMID: 37658748 DOI: 10.1002/rmv.2478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/17/2023] [Accepted: 08/23/2023] [Indexed: 09/05/2023]
Abstract
Myocarditis can result from various infectious and non-infectious causes that can lead to dilated cardiomyopathy (DCM) and heart failure. Among the infectious causes, viruses are commonly suspected. But the challenge is our inability to demonstrate infectious viral particles during clinical presentations, partly because by that point, the viruses would have damaged the tissues and be cleared by the immune system. Therefore, viral signatures such as viral nucleic acids and virus-reactive antibodies may be the only readouts pointing to viruses as potential primary triggers of DCM. Thus, it becomes hard to explain persistent inflammatory infiltrates that might occur in individuals affected with chronic myocarditis/DCM manifesting myocardial dysfunctions. In these circumstances, autoimmunity is suspected, and antibodies to various autoantigens have been demonstrated, suggesting that immune therapies to suppress the autoimmune responses may be necessary. From this perspective, we endeavoured to determine whether or not the known viral causes are associated with development of autoimmune responses to cardiac antigens that include both cardiotropic and non-cardiotropic viruses. If so, what their nature and significance are in developing chronic myocarditis resulting from viruses as primary triggers.
Collapse
Affiliation(s)
- Kiruthiga Mone
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Jay Reddy
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| |
Collapse
|
16
|
Toprak K, Inanır M, Memioğlu T, Kaplangoray M, Palice A, Tascanov MB. Could Zonulin and Presepsin Be Biomarkers and Therapeutic Targets for Acute Myocarditis? Arq Bras Cardiol 2023; 120:e20230017. [PMID: 37556677 PMCID: PMC10464857 DOI: 10.36660/abc.20230017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/28/2023] [Accepted: 05/17/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND The diagnosis of acute myocarditis is usually made with clinical and laboratory parameters. This can sometimes be mixed up with diseases that have similar clinical features, making the diagnosis difficult. Therefore, the use of more specific biomarkers, in addition to the classically used biomarkers such as troponin, will accelerate the diagnosis. In addition, these biomarkers may help us to understand the mechanism of myocarditis development and thus predict unpredictable clinical outcomes. OBJECTIVE This study aims to reveal the possible relationship between intestinal permeability and acute myocarditis. METHODS In this study, we wanted to evaluate serum levels of zonulin and presepsin in 138 consecutive subjects, including 68 patients with myocarditis and another 70 as the control group, matched for age, gender, and cardiovascular risk factors. P-values <0.05 were considered to be statistically significant. RESULTS Compared to the control group, zonulin and presepsin were significantly higher in the patient group with myocarditis (p < 0.001, for all). Zonulin levels were positively correlated with presepsin, peak CK-MB, and peak troponin levels (r = 0.461, p < 0.001; r = 0.744, p < 0.001; r = 0.627, p < 0.001; respectively). In regression analysis, presepsin and zonulin were determined as independent predictors for myocarditis (OR 1.002, 95% CI 1.001-1.003, p = 0.025; OR 12.331, 95% CI 4.261-35.689; p < 0.001; respectively). The predictive value of acute myocarditis of presepsin and zonulin in ROC curve analysis was statistically significant (p < 0.001, for both). CONCLUSION This study showed that zonulin and presepsin could be biomarkers that can be used in the diagnosis of myocarditis, and they can also be therapeutic targets by shedding light on the developmental mechanism of myocarditis.
Collapse
Affiliation(s)
- Kenan Toprak
- Harran UniversityFaculty of MedicineDepartment of CardiologySanliurfaTurquiaHarran University Faculty of Medicine – Department of Cardiology, Sanliurfa – Turquia
| | - Mehmet Inanır
- Abant Izzet Baysal University HospitalBoluTurquiaAbant Izzet Baysal University Hospital – Cardiology, Bolu – Turquia
| | - Tolga Memioğlu
- Abant Izzet Baysal University HospitalBoluTurquiaAbant Izzet Baysal University Hospital – Cardiology, Bolu – Turquia
| | - Mustafa Kaplangoray
- Sanliurfa Mehmet Akif Inan Training and ResearchSanliurfaTurquiaSanliurfa Mehmet Akif Inan Training and Research – Cardiology, Sanliurfa – Turquia
| | - Ali Palice
- Sanliurfa Mehmet Akif Inan Training and ResearchSanliurfaTurquiaSanliurfa Mehmet Akif Inan Training and Research – Cardiology, Sanliurfa – Turquia
| | - Mustafa Begenc Tascanov
- Harran UniversityFaculty of MedicineDepartment of CardiologySanliurfaTurquiaHarran University Faculty of Medicine – Department of Cardiology, Sanliurfa – Turquia
| |
Collapse
|
17
|
Fang Y, Wu H, Liu G, Li Z, Wang D, Ning Y, Pan S, Hu Y. Secondary immunoreaction in patients with neurosyphilis and its relevance to clinical outcomes. Front Neurol 2023; 14:1201452. [PMID: 37346161 PMCID: PMC10281193 DOI: 10.3389/fneur.2023.1201452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 05/16/2023] [Indexed: 06/23/2023] Open
Abstract
Background and purpose Several reported cases of autoimmune conditions such as anti-NMDAR encephalitis and neuromyelitis optica (AQP4) have been considered to be potentially secondary to Treponema pallidum infection. Since the role of immune impairment in neurosyphilis is unclear, in this retrospective study, we examined the correlation of the immune impairment in patients with neurosyphilis with their clinical characteristics and outcomes. Methods Clinical information was collected from patients with neurosyphilis in our center from January 2019 to December 2021. Cerebrospinal fluid (CSF) samples were subjected to indirect immunofluorescence tissue-based assay (IIF-TBA) on mouse brain sections and cell-based assay (CBA). The clinical characteristics and treatment outcomes of TBA-positive and-negative patients were compared. Results A total number of 81 patients diagnosed with neurosyphilis were included. The results of the CBA tests showed that three cases had anti-NMDAR, AQP4, or GAD65 antibodies, respectively. By TBA test, 38 patients (38/81, 46.9%) had positive immunostains, including staining of neuronal cells in 21 cases (21/38, 55.3%), glial cells in 11 cases (11/38, 28.9%), and neuronal and glial cells in six cases (6/38, 15.8%). We then compared the clinical characteristics and treatment outcomes between the TBA-positive and-negative patients and found that TBA-positive staining was significantly correlated with syphilis antibody titers (p = 0.027 for serum and p = 0.006 for CSF) and head MRI abnormalities (p < 0.001 for parenchymal abnormalities and p = 0.013 for white matter lesions). The cognitive prognosis of TBA-positive neurosyphilis patients was significantly worse than that of TBA-negative patients (p < 0.001). Conclusion The correlation between the TBA results and clinical data of our neurosyphilis patients imply the presence of secondary immune damage, which affected their prognosis. Therefore, TBA can be used as an additional biomarker for neurosyphilis patient prognosis.
Collapse
Affiliation(s)
- Yaxiu Fang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Neurology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hong Wu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guanghui Liu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ziang Li
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dongmei Wang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuping Ning
- Department of Neurology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Suyue Pan
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yafang Hu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
18
|
Matsumori A. Myocarditis and Autoimmunity. Expert Rev Cardiovasc Ther 2023. [PMID: 37243585 DOI: 10.1080/14779072.2023.2219895] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 05/20/2023] [Accepted: 05/26/2023] [Indexed: 05/29/2023]
Abstract
INTRODUCTION Autoimmune myocarditis may develop due to heterogeneous causes. Myocarditis is often caused by viral infections, but it can also be caused by systemic autoimmune diseases. Immune checkpoint inhibitors and virus vaccines induce immune activation, and they can cause the development of myocarditis, as well as several immune-related adverse events. The development of myocarditis is dependent on the genetic factors of the host, and the major histocompatibility complex (MHC) may be an important determinant of the type and severity of the disease. However, non-MHC immunoregulatory genes may also play a role in determining susceptibility. AREA COVERED This review summarizes the current knowledge of the etiology, pathogenesis, diagnosis and treatment of autoimmune myocarditis with a particular focus on viral infection and autoimmunity, and biomarkers of myocarditis. EXPERT OPINION An endomyocardial biopsy may not be the gold standard for the diagnosis of myocarditis. Cardiac magnetic resonance imaging is useful in diagnosing autoimmune myocarditis. Recently identified biomarkers of inflammation and myocyte injury are promising for the diagnosis of myocarditis when measured simultaneously. Future treatments should focus on the appropriate diagnosis of the etiologic agent, as well as on the specific stage of the evolution of immune and inflammatory processes.
Collapse
Affiliation(s)
- Akira Matsumori
- Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto 612-8555, Japan
| |
Collapse
|
19
|
Abstract
Viral infections are a leading cause of myocarditis and pericarditis worldwide, conditions that frequently coexist. Myocarditis and pericarditis were some of the early comorbidities associated with SARS-CoV-2 infection and COVID-19. Many epidemiologic studies have been conducted since that time concluding that SARS-CoV-2 increased the incidence of myocarditis/pericarditis at least 15× over pre-COVID levels although the condition remains rare. The incidence of myocarditis pre-COVID was reported at 1 to 10 cases/100 000 individuals and with COVID ranging from 150 to 4000 cases/100 000 individuals. Before COVID-19, some vaccines were reported to cause myocarditis and pericarditis in rare cases, but the use of novel mRNA platforms led to a higher number of reported cases than with previous platforms providing new insight into potential pathogenic mechanisms. The incidence of COVID-19 vaccine-associated myocarditis/pericarditis covers a large range depending on the vaccine platform, age, and sex examined. Importantly, the findings highlight that myocarditis occurs predominantly in male patients aged 12 to 40 years regardless of whether the cause was due to a virus-like SARS-CoV-2 or associated with a vaccine-a demographic that has been reported before COVID-19. This review discusses findings from COVID-19 and COVID-19 vaccine-associated myocarditis and pericarditis considering the known symptoms, diagnosis, management, treatment, and pathogenesis of disease that has been gleaned from clinical research and animal models. Sex differences in the immune response to COVID-19 are discussed, and theories for how mRNA vaccines could lead to myocarditis/pericarditis are proposed. Additionally, gaps in our understanding that need further research are raised.
Collapse
Affiliation(s)
- DeLisa Fairweather
- Department of Cardiovascular Medicine (D.F., D.J.B., D.N.D., L.T.C.), Mayo Clinic, Jacksonville, FL
- Department of Environmental Health Sciences and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (D.F.,)
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN (D.F., D.J.B., D.N.D.)
| | - Danielle J. Beetler
- Department of Cardiovascular Medicine (D.F., D.J.B., D.N.D., L.T.C.), Mayo Clinic, Jacksonville, FL
- Mayo Clinic Graduate School of Biomedical Sciences (D.J.B., D.N.D.), Mayo Clinic, Jacksonville, FL
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN (D.F., D.J.B., D.N.D.)
| | - Damian N. Di Florio
- Department of Cardiovascular Medicine (D.F., D.J.B., D.N.D., L.T.C.), Mayo Clinic, Jacksonville, FL
- Mayo Clinic Graduate School of Biomedical Sciences (D.J.B., D.N.D.), Mayo Clinic, Jacksonville, FL
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN (D.F., D.J.B., D.N.D.)
| | - Nicolas Musigk
- Deutsches Herzzentrum der Charité, Berlin, Germany (N.M., B.H.)
| | | | - Leslie T. Cooper
- Department of Cardiovascular Medicine (D.F., D.J.B., D.N.D., L.T.C.), Mayo Clinic, Jacksonville, FL
| |
Collapse
|
20
|
Johnson D, Jiang W. Infectious diseases, autoantibodies, and autoimmunity. J Autoimmun 2023; 137:102962. [PMID: 36470769 PMCID: PMC10235211 DOI: 10.1016/j.jaut.2022.102962] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 11/27/2022] [Indexed: 12/04/2022]
Abstract
Infections are known to trigger flares of autoimmune diseases in humans and serve as an inciting cause of autoimmunity in animals. Evidence suggests a causative role of infections in triggering antigen-specific autoimmunity, previous thought mainly through antigen mimicry. However, an infection can induce bystander autoreactive T and B cell polyclonal activation, believed to result in non-pathogenic and pathogenic autoimmune responses. Lastly, epitope spreading in autoimmunity is a mechanism of epitope changes of autoreactive cells induced by infection, promoting the targeting of additional self-epitopes. This review highlights recent research findings, emphasizes infection-mediated autoimmune responses, and discusses the possible mechanisms involved.
Collapse
Affiliation(s)
- Douglas Johnson
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave., Charleston, SC, USA; Ralph H. Johnson VA Medical Center, Charleston, SC, USA
| | - Wei Jiang
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave., Charleston, SC, USA; Ralph H. Johnson VA Medical Center, Charleston, SC, USA; Divison of Infectious Disease, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
21
|
Cohen CD, Rousseau ST, Bermea KC, Bhalodia A, Lovell JP, Dina Zita M, Čiháková D, Adamo L. Myocardial Immune Cells: The Basis of Cardiac Immunology. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1198-1207. [PMID: 37068299 PMCID: PMC10111214 DOI: 10.4049/jimmunol.2200924] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 01/14/2023] [Indexed: 04/19/2023]
Abstract
The mammalian heart is characterized by the presence of striated myocytes, which allow continuous rhythmic contraction from early embryonic development until the last moments of life. However, the myocardium contains a significant contingent of leukocytes from every major class. This leukocyte pool includes both resident and nonresident immune cells. Over recent decades, it has become increasingly apparent that the heart is intimately sensitive to immune signaling and that myocardial leukocytes exhibit an array of critical functions, both in homeostasis and in the context of cardiac adaptation to injury. Here, we systematically review current knowledge of all major leukocyte classes in the heart, discussing their functions in health and disease. We also highlight the connection between the myocardium, immune cells, lymphoid organs, and both local and systemic immune responses.
Collapse
Affiliation(s)
- Charles D. Cohen
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Sylvie T. Rousseau
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Kevin C. Bermea
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Aashik Bhalodia
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Jana P. Lovell
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Marcelle Dina Zita
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Daniela Čiháková
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - Luigi Adamo
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| |
Collapse
|
22
|
Giardino G, Romano R, Lougaris V, Castagnoli R, Cillo F, Leonardi L, La Torre F, Soresina A, Federici S, Cancrini C, Pacillo L, Toriello E, Cinicola BL, Corrente S, Volpi S, Marseglia GL, Pignata C, Cardinale F. Immune tolerance breakdown in inborn errors of immunity: Paving the way to novel therapeutic approaches. Clin Immunol 2023; 251:109302. [PMID: 36967025 DOI: 10.1016/j.clim.2023.109302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 03/06/2023] [Accepted: 03/22/2023] [Indexed: 05/12/2023]
Abstract
Up to 25% of the patients with inborn errors of immunity (IEI) also exhibit immunodysregulatory features. The association of immune dysregulation and immunodeficiency may be explained by different mechanisms. The understanding of mechanisms underlying immune dysregulation in IEI has paved the way for the development of targeted treatments. In this review article, we will summarize the mechanisms of immune tolerance breakdown and the targeted therapeutic approaches to immune dysregulation in IEI.
Collapse
Affiliation(s)
- Giuliana Giardino
- Pediatric Section, Department of Translational Medical Sciences, Federico II University, Naples, Italy.
| | - Roberta Romano
- Pediatric Section, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Vassilios Lougaris
- Department of Clinical and Experimental Sciences, Pediatrics Clinic and Institute for Molecular Medicine A. Nocivelli, University of Brescia and ASST-Spedali Civili di Brescia, Brescia, Italy
| | - Riccardo Castagnoli
- Department of Pediatrics, Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Francesca Cillo
- Pediatric Section, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Lucia Leonardi
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Francesco La Torre
- Department of Pediatrics, Giovanni XXIII Pediatric Hospital, University of Bari, Bari, Italy
| | - Annarosa Soresina
- Unit of Pediatric Immunology, Pediatrics Clinic, University of Brescia, ASST Spedali Civili Brescia, Brescia, Italy
| | - Silvia Federici
- Division of Rheumatology, IRCCS, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Caterina Cancrini
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Research Unit of Primary Immunodeficiencies, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Lucia Pacillo
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Research Unit of Primary Immunodeficiencies, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Elisabetta Toriello
- Pediatric Section, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Bianca Laura Cinicola
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | | | - Stefano Volpi
- Center for Autoinflammatory Diseases and Immunodeficiency, IRCCS Istituto Giannina Gaslini, Università degli Studi di Genova, Genoa, Italy
| | - Gian Luigi Marseglia
- Department of Pediatrics, Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Claudio Pignata
- Pediatric Section, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Fabio Cardinale
- Department of Pediatrics, Giovanni XXIII Pediatric Hospital, University of Bari, Bari, Italy
| |
Collapse
|
23
|
Fairweather D, Beetler DJ, Musigk N, Heidecker B, Lyle MA, Cooper LT, Bruno KA. Sex and gender differences in myocarditis and dilated cardiomyopathy: An update. Front Cardiovasc Med 2023; 10:1129348. [PMID: 36937911 PMCID: PMC10017519 DOI: 10.3389/fcvm.2023.1129348] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/06/2023] [Indexed: 03/06/2023] Open
Abstract
In the past decade there has been a growing interest in understanding sex and gender differences in myocarditis and dilated cardiomyopathy (DCM), and the purpose of this review is to provide an update on this topic including epidemiology, pathogenesis and clinical presentation, diagnosis and management. Recently, many clinical studies have been conducted examining sex differences in myocarditis. Studies consistently report that myocarditis occurs more often in men than women with a sex ratio ranging from 1:2-4 female to male. Studies reveal that DCM also has a sex ratio of around 1:3 women to men and this is also true for familial/genetic forms of DCM. Animal models have demonstrated that DCM develops after myocarditis in susceptible mouse strains and evidence exists for this progress clinically as well. A consistent finding is that myocarditis occurs primarily in men under 50 years of age, but in women after age 50 or post-menopause. In contrast, DCM typically occurs after age 50, although the age that post-myocarditis DCM occurs has not been investigated. In a small study, more men with myocarditis presented with symptoms of chest pain while women presented with dyspnea. Men with myocarditis have been found to have higher levels of heart failure biomarkers soluble ST2, creatine kinase, myoglobin and T helper 17-associated cytokines while women develop a better regulatory immune response. Studies of the pathogenesis of disease have found that Toll-like receptor (TLR)2 and TLR4 signaling pathways play a central role in increasing inflammation during myocarditis and in promoting remodeling and fibrosis that leads to DCM, and all of these pathways are elevated in males. Management of myocarditis follows heart failure guidelines and there are currently no disease-specific therapies. Research on standard heart failure medications reveal important sex differences. Overall, many advances in our understanding of the effect of biologic sex on myocarditis and DCM have occurred over the past decade, but many gaps in our understanding remain. A better understanding of sex and gender effects are needed to develop disease-targeted and individualized medicine approaches in the future.
Collapse
Affiliation(s)
- DeLisa Fairweather
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Department of Environmental Health Sciences and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, United States
| | - Danielle J. Beetler
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, United States
| | - Nicolas Musigk
- Department of Cardiology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Bettina Heidecker
- Department of Cardiology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Melissa A. Lyle
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Leslie T. Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Katelyn A. Bruno
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Division of Cardiovascular Medicine, Department of Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
24
|
Petrov N, Stoyanova M, Stoyanova A, Nikolova I, Grozdanov P, Galabov A. Gene silencing of VP1 gene of coxsackievirus B3 neurotropic strain Nancy by dsRNAs and siRNAs. BIOTECHNOL BIOTEC EQ 2022. [DOI: 10.1080/13102818.2022.2082320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Affiliation(s)
- Nikolay Petrov
- Laboratory of Virology, Department of Natural Sciences, New Bulgarian University, Sofia, Bulgaria
| | - Mariya Stoyanova
- Department of Plant Protection, Institute of Soil Science, Agrotechnologies and Plant Protection “N. Pushkarov”, Agricultural Academy, Sofia, Bulgaria
| | - Adelina Stoyanova
- Department of Virology, The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Ivanka Nikolova
- Department of Virology, The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Petar Grozdanov
- Department of Virology, The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Angel Galabov
- Department of Virology, The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| |
Collapse
|
25
|
Matsumori A, Auda ME, Bruno KA, Shapiro KA, Kato T, Nakamura T, Hasegawa K, Saleh A, Abdelrazek S, Negm H, Karunawan NH, Cooper LT, Fairweather D. Cardiovascular Factors Associated with COVID-19 from an International Registry of Primarily Japanese Patients. Diagnostics (Basel) 2022; 12:2350. [PMID: 36292038 PMCID: PMC9600010 DOI: 10.3390/diagnostics12102350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 09/17/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022] Open
Abstract
Aims: We developed an international registry to examine cardiovascular complications of COVID-19. Methods: A REDCap form was created in March 2020 at Mayo Clinic in collaboration with the International Society of Cardiomyopathy, Myocarditis and Heart Failure (ISCMF) and data were entered from April 2020 through April 2021. Results: Of the 696 patients in the COVID-19 Registry, 411 (59.2%) were male and 283 (40.8%) were female, with a sex ratio of 1.5:1 male to female. In total, 95.5% of the patients were from Japan. The average age was 52 years with 31.5% being >65 years of age. COVID-19 patients with a history of cardiovascular disease (CVD) had more pre-existing conditions including type II diabetes (p < 0.0001), cancer (p = 0.0003), obesity (p = 0.001), and kidney disease (p = 0.001). They also had a greater mortality of 10.1% compared to 1.7% in those without a history of CVD (p < 0.0001). The most common cardiovascular conditions in patients with a history of CVD were hypertension (33.7%), stroke (5.7%) and arrhythmias (5.1%). We found that troponin T, troponin I, brain natriuretic peptide (BNP), N-terminal pro-BNP (NT-proBNP), C-reactive protein (CRP), IL-6 and lambda immunoglobulin free light chains (Ig FLC) were elevated above reference levels in patients with COVID-19. Myocarditis is known to occur mainly in adults under the age of 50, and when we examined biomarkers in patients that were ≤50 years of age and had no history of CVD we found that a majority of patients had elevated levels of troponin T (71.4%), IL-6 (59.5%), creatine kinase/CK-MB (57.1%), D-dimer (57.8%), kappa Ig FLC (75.0%), and lambda Ig FLC (71.4%) suggesting myocardial injury and possible myocarditis. Conclusions: We report the first findings to our knowledge of cardiovascular complications from COVID-19 in the first year of the pandemic in a predominantly Japanese population. Mortality was increased by a history of CVD and pre-existing conditions including type II diabetes, cancer, obesity, and kidney disease. Our findings indicate that even in cases where no abnormalities are found in ECG or ultrasound cardiography that myocardial damage may occur, and cardiovascular and inflammatory biomarkers may be useful for the diagnosis.
Collapse
Affiliation(s)
- Akira Matsumori
- Clinical Research Center, Kyoto Medical Center, Kyoto 612-8555, Japan
| | - Matthew E. Auda
- Department of Cardiovascular Medicine, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Katelyn A. Bruno
- Department of Cardiovascular Medicine, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
- Center of Clinical and Translational Science, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
- Department of Immunology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
- Division of Cardiovascular Medicine, Department of Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Katie A. Shapiro
- Department of Cardiovascular Medicine, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Toru Kato
- Department of Clinical Research, National Hospital Organization, Tochigi Medical Center, Utsunomiya 320-8580, Japan
| | - Toshihiro Nakamura
- Department of Cardiology, National Hospital Organization, Kyushu Medical Center, Fukuoka 810-8563, Japan
| | - Koji Hasegawa
- Division of Translational Research, National Hospital Organization, Kyoto Medical Center, Kyoto 602-8566, Japan
| | - Ahmed Saleh
- Klinikum Braunschweig, Academic Hospital of Hannover Medical School, 30625 Braunschweig, Germany
| | - Sherif Abdelrazek
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, 79189 Bad Krozingen, Germany
| | - Hany Negm
- Cardiology and Ultrasonography Unit, Research Institute of Ophthalmology (RIO), Cairo 11261, Egypt
| | - Niyata Hananta Karunawan
- Department of Cardiology and Vascular Medicine, Tugurejo General Hospital, Semarang 50185, Central Java, Indonesia
| | - Leslie T. Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - DeLisa Fairweather
- Department of Cardiovascular Medicine, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
- Center of Clinical and Translational Science, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
- Department of Immunology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, USA
| |
Collapse
|
26
|
Vlachiotis S, Abolhassani H. Transcriptional regulation of B cell class-switch recombination: the role in development of noninfectious complications. Expert Rev Clin Immunol 2022; 18:1145-1154. [DOI: 10.1080/1744666x.2022.2123795] [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]
Affiliation(s)
- Stelios Vlachiotis
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Hassan Abolhassani
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
Liu M, Lin Y, Xu H, Li L, Ding T. Combination of Sophora flavescens alkaloids and Panax quinquefolium saponins modulates different stages of experimental autoimmune myocarditis via the NF‑κB and TGF‑β1 pathways. Exp Ther Med 2022; 24:570. [PMID: 36034755 PMCID: PMC9400131 DOI: 10.3892/etm.2022.11507] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/29/2022] [Indexed: 12/04/2022] Open
Abstract
Chronic cardiac inflammation and fibrosis can progress into severe forms of cardiomyopathy. Sophora flavescens alkaloids (KuShen) have been previously reported to exert anti-inflammatory effects, whereas Panax quinquefolium saponins (XiYangShen) has been shown to alleviate cardiac fibrosis. Therefore, the potential effects of their combination (KX) on different stages of autoimmune myocarditis were investigated in the present study. Mice were randomly divided into the following four groups: Control; experimental autoimmune myocarditis (EAM); KX-High (275 mg/kg); and KX-Low (138 mg/kg). A 21-day and a 60-day EAM model was established through multi-site subcutaneous injections of cardiac myosin mixed with complete Freund's adjuvant on days 0, 7, 21 and 42. Mice in the High and Low KX groups were treated by gavage (10 ml/kg) daily from day 0 (1 day before treatment) until sacrifice (day 21 or 60). Mice in the control and EAM groups received an equivalent volume of distilled water. The levels of lactate dehydrogenase (LDH), creatine kinase-myocardial band (CK-MB), cardiac troponin I (cTn-I), IL-1β, IL-6, TNF-α, TGF-β1, collagen type I (Col Ⅰ) and collagen type III (Col Ⅲ) were measured by ELISA in the mouse myocardial tissues or serum. Myocardial tissue structure and extent of fibrosis were visualized using H&E and Masson's staining. Western blotting and immunohistochemistry were used to measure the expression levels NF-κB and TGF-β1 pathway proteins in the myocardial tissues. The degree of inflammation in the 21-day EAM model was found to be significantly higher compared with that in the 60-day EAM model. KX significantly reduced the inflammatory response at 21 days by decreasing the expression levels of CK-MB, LDH, cTn-I, IL-1β, IL-6, TNF-α and TGF-β-activated kinase 1-binding protein 1/NF-κB pathway proteins. Myocardial fibrosis in the 60-day EAM model was also significantly worse compared with that in the 21-day EAM model. However, fibrosis was significantly delayed by treatment with KX. In addition, KX significantly decreased the expression levels of TGF-β1, Smad2, Smad4, Col I and Col III. Therefore, these data suggest that KX is beneficial for treating myocarditis by targeting multiple pathways.
Collapse
Affiliation(s)
- Menghui Liu
- Department of Traditional Chinese Medicine, Changchun University of Traditional Chinese Medicine, Changchun, Jilin 130000, P.R. China
| | - Yue Lin
- Department of Traditional Chinese Medicine, Changchun University of Traditional Chinese Medicine, Changchun, Jilin 130000, P.R. China
| | - Huibo Xu
- Pharmacodynamic and Toxicological Evaluation Center, Jilin Academy of Traditional Chinese Medicine, Changchun, Jilin 130000, P.R. China
| | - Lixin Li
- Department of Pediatrics, Jilin Academy of Traditional Chinese Medicine, Changchun, Jilin 130000, P.R. China
| | - Tao Ding
- Pharmacodynamic and Toxicological Evaluation Center, Jilin Academy of Traditional Chinese Medicine, Changchun, Jilin 130000, P.R. China
| |
Collapse
|
28
|
Oh JH, Kim GB, Seok H. Implication of microRNA as a potential biomarker of myocarditis. Clin Exp Pediatr 2022; 65:230-238. [PMID: 35240034 PMCID: PMC9082251 DOI: 10.3345/cep.2021.01802] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 01/29/2022] [Indexed: 12/15/2022] Open
Abstract
Myocarditis was previously attributed to an epidemic viral infection. Additional harmful reagents, in addition to viruses, play a role in its etiology. Coronavirus disease 2019 (COVID-19) vaccine-induced myocarditis has recently been described, drawing attention to vaccine-induced myocarditis in children and adolescents. Its pathology is based on a series of complex immune responses, including initial innate immune responses in response to viral entry, adaptive immune responses leading to the development of antigen-specific antibodies, and autoimmune responses to cellular injury caused by cardiomyocyte rupture that releases antigens. Chronic inflammation and fibrosis in the myocardium eventually result in cardiac failure. Recent advancements in molecular biology have remarkably increased our understanding of myocarditis. In particular, microRNAs (miRNAs) are a hot topic in terms of the role of new biomarkers and the pathophysiology of myocarditis. Myocarditis has been linked with microRNA-221/222 (miR-221/222), miR-155, miR-10a*, and miR-590. Despite the lack of clinical trials of miRNA intervention in myocarditis yet, multiple clinical trials of miRNAs in other cardiac diseases have been aggressively conducted to help pave the way for future research, which is bolstered by the success of recently U.S. Food and Drug Administration-approved small-RNA medications. This review presents basic information and recent research that focuses on myocarditis and related miRNAs as a potential novel biomarker and the therapeutics.
Collapse
Affiliation(s)
- Jin-Hee Oh
- Department of Pediatrics, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Gi Beom Kim
- Department of Pediatrics, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Heeyoung Seok
- Department of Transdisciplinary Research and Collaboration, Genomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
29
|
Pourgholaminejad A, Pahlavanneshan S, Basiri M. COVID-19 immunopathology with emphasis on Th17 response and cell-based immunomodulation therapy: Potential targets and challenges. Scand J Immunol 2021; 95:e13131. [PMID: 34936112 DOI: 10.1111/sji.13131] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 12/04/2021] [Accepted: 12/15/2021] [Indexed: 12/27/2022]
Abstract
The role of the immune system against coronavirus disease 2019 (COVID-19) is unknown in many aspects, and the protective or pathologic mechanisms of the immune response are poorly understood. Pro-inflammatory cytokine release and a consequent cytokine storm can lead to acute respiratory distress syndrome (ARDS) and result in multi-organ failure. There are many T cell subsets during anti-viral immunity. The Th17-associated response, as a pro-inflammatory pathway, and its consequent outcomes in many autoimmune disorders play a fundamental role in progression of systemic hyper-inflammation during COVID-19. Therapeutic strategies based on immunomodulation therapy could be helpful for targeting hyper-inflammatory immune responses in COVID-19, especially Th17-related inflammation and hyper-cytokinemia. Cell-based immunotherapeutic approaches including mesenchymal stem cells (MSCs), tolerogenic dendritic cells (tolDCs) and regulatory T cells (Tregs) seem to be promising strategies as orchestrators of the immune response against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In this review, we highlight Th17-related immunopathology of SARS-CoV-2 infection and discuss cell-based immunomodulatory strategies and their mechanisms for regulation of the hyper-inflammation during COVID-19.
Collapse
Affiliation(s)
- Arash Pourgholaminejad
- Department of Immunology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Saghar Pahlavanneshan
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Basiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
30
|
Sex-Specific Effects of Plastic Caging in Murine Viral Myocarditis. Int J Mol Sci 2021; 22:ijms22168834. [PMID: 34445539 PMCID: PMC8396197 DOI: 10.3390/ijms22168834] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/29/2021] [Accepted: 08/12/2021] [Indexed: 02/07/2023] Open
Abstract
Background: Myocarditis is an inflammatory heart disease caused by viral infections that can lead to heart failure, and occurs more often in men than women. Since animal studies have shown that myocarditis is influenced by sex hormones, we hypothesized that endocrine disruptors, which interfere with natural hormones, may play a role in the progression of the disease. The human population is exposed to the endocrine disruptor bisphenol A (BPA) from plastics, such as water bottles and plastic food containers. Methods: Male and female adult BALB/c mice were housed in plastic versus glass caging, or exposed to BPA in drinking water versus control water. Myocarditis was induced with coxsackievirus B3 on day 0, and the endpoints were assessed on day 10 post infection. Results: We found that male BALB/c mice that were exposed to plastic caging had increased myocarditis due to complement activation and elevated numbers of macrophages and neutrophils, whereas females had elevated mast cell activation and fibrosis. Conclusions: These findings show that housing mice in traditional plastic caging increases viral myocarditis in males and females, but using sex-specific immune mechanisms.
Collapse
|
31
|
Favere K, Bosman M, Klingel K, Heymans S, Van Linthout S, Delputte PL, De Sutter J, Heidbuchel H, Guns PJ. Toll-Like Receptors: Are They Taking a Toll on the Heart in Viral Myocarditis? Viruses 2021; 13:v13061003. [PMID: 34072044 PMCID: PMC8227433 DOI: 10.3390/v13061003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/23/2021] [Accepted: 05/24/2021] [Indexed: 12/30/2022] Open
Abstract
Myocarditis is an inflammatory disease of the heart with viral infections being the most common aetiology. Its complex biology remains poorly understood and its clinical management is one of the most challenging in the field of cardiology. Toll-like receptors (TLRs), a family of evolutionarily conserved pattern recognition receptors, are increasingly known to be implicated in the pathophysiology of viral myocarditis. Their central role in innate and adaptive immune responses, and in the inflammatory reaction that ensues, indeed makes them prime candidates to profoundly affect every stage of the disease process. This review describes the pathogenesis and pathophysiology of viral myocarditis, and scrutinises the role of TLRs in every phase. We conclude with directions for future research in this field.
Collapse
Affiliation(s)
- Kasper Favere
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, 2610 Antwerp, Belgium; (M.B.); (P.-J.G.)
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, 2610 Antwerp, Belgium;
- Department of Cardiology, Antwerp University Hospital, 2650 Antwerp, Belgium
- Department of Internal Medicine, Ghent University, 9000 Ghent, Belgium;
- Correspondence:
| | - Matthias Bosman
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, 2610 Antwerp, Belgium; (M.B.); (P.-J.G.)
| | - Karin Klingel
- Cardiopathology, Institute for Pathology, University Hospital Tuebingen, 72076 Tuebingen, Germany;
| | - Stephane Heymans
- Department of Cardiology, Maastricht University, 6229 ER Maastricht, The Netherlands;
- Centre for Molecular and Vascular Biology, KU Leuven, 3000 Leuven, Belgium
| | - Sophie Van Linthout
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité, Universitätsmedizin Berlin, 10117 Berlin, Germany;
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, 10785 Berlin, Germany
| | - Peter L. Delputte
- Laboratory of Microbiology, Parasitology and Hygiene, University of Antwerp, 2610 Antwerp, Belgium;
| | - Johan De Sutter
- Department of Internal Medicine, Ghent University, 9000 Ghent, Belgium;
| | - Hein Heidbuchel
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, 2610 Antwerp, Belgium;
- Department of Cardiology, Antwerp University Hospital, 2650 Antwerp, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, 2610 Antwerp, Belgium; (M.B.); (P.-J.G.)
| |
Collapse
|
32
|
Golpour A, Patriki D, Hanson PJ, McManus B, Heidecker B. Epidemiological Impact of Myocarditis. J Clin Med 2021; 10:603. [PMID: 33562759 PMCID: PMC7915005 DOI: 10.3390/jcm10040603] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 01/24/2021] [Accepted: 02/01/2021] [Indexed: 02/06/2023] Open
Abstract
Myocarditis is an inflammatory disease of the heart muscle with a wide range of potential etiological factors and consequently varying clinical patterns across the world. In this review, we address the epidemiology of myocarditis. Myocarditis was considered a rare disease until intensified research efforts in recent decades revealed its true epidemiological importance. While it remains a challenge to determine the true prevalence of myocarditis, studies are underway to obtain better approximations of the proportions of this disease. Nowadays, the prevalence of myocarditis has been reported from 10.2 to 105.6 per 100,000 worldwide, and its annual occurrence is estimated at about 1.8 million cases. This wide range of reported cases reflects the uncertainty surrounding the true prevalence and a potential underdiagnosis of this disease. Since myocarditis continues to be a significant public health issue, particularly in young adults in whom myocarditis is among the most common causes of sudden cardiac death, improved diagnostic and therapeutic procedures are necessary. This manuscript aims to summarize the current knowledge on the epidemiology of myocarditis, new diagnostic approaches and the current epidemiological impact of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Ainoosh Golpour
- Campus Benjamin Franklin, Charite Universitätsmedizin Berlin, 12203 Berlin, Germany;
| | - Dimitri Patriki
- Department of Medicine, Cantonal Hospital of Baden, 15005 Baden, Switzerland;
| | - Paul J. Hanson
- Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC V5K0A1, Canada; (P.J.H.); (B.M.)
| | - Bruce McManus
- Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC V5K0A1, Canada; (P.J.H.); (B.M.)
| | - Bettina Heidecker
- Campus Benjamin Franklin, Charite Universitätsmedizin Berlin, 12203 Berlin, Germany;
| |
Collapse
|
33
|
Heidecker B, Williams SH, Jain K, Oleynik A, Patriki D, Kottwitz J, Berg J, Garcia JA, Baltensperger N, Lovrinovic M, Baltensweiler A, Mishra N, Briese T, Hanson PJ, Lauten A, Poller W, Leistner DM, Landmesser U, Enseleit F, McManus B, Lüscher TF, Lipkin WI. Virome Sequencing in Patients With Myocarditis. Circ Heart Fail 2020; 13:e007103. [PMID: 32586108 DOI: 10.1161/circheartfailure.120.007103] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Polymerase chain reaction analyses of cardiac tissues have detected viral sequences in up to 67% of cases of myocarditis. However, viruses have not been implicated in giant cell myocarditis (GCM). Furthermore, efforts to detect viruses implicated in myocarditis have been unsuccessful in more accessible samples such as peripheral blood. METHODS We used Virome Capture Sequencing for Vertbrate Viruses (VirCapSeq-VERT), a method that simultaneously screens for all known vertebrate viruses, to investigate viruses in 33 patients with myocarditis. We investigated peripheral blood mononuclear cells (n=24), plasma (n=27), endomyocardial biopsies (n=2), and cardiac tissue samples from explanted hearts (n=13). RESULTS Nine patients (27%) had GCM and 4 patients (13%) had fulminant myocarditis. We found the following viruses in the blood of patients with myocarditis: Epstein Barr virus (n=11, 41%), human pegivirus (n=1, 4%), human endogenous retrovirus K (n=27, 100%), and anellovirus (n=15, 56%). All tissue samples from fulminant myocarditis (n=2) and GCM (n=13) contained human endogenous retrovirus K. CONCLUSIONS No nucleic acids from viruses previously implicated in myocarditis or other human illnesses were detected in relevant amounts in cardiac tissue samples from GCM or in blood samples from other types of myocarditis. These findings do not exclude a role for viral infection in GCM but do suggest that if viruses are implicated, the mechanism is likely to be indirect rather than due to cytotoxic infection of myocardium.
Collapse
Affiliation(s)
- Bettina Heidecker
- Department of Cardiology, Charite University Hospital Berlin; Berlin Institute of Health (BIH), Berlin, Germany (B.H., A.L., W.P., D.L., U.L.).,Center for Infection and Immunity, Mailman School of Public Health, Columbia University, NY (B.H., S.H.W., K.J., A.O., J.A.G., N.M., T.B., W.I.L.).,University Hospital Zurich, Zurich, Switzerland (B.H., D.P., J.K, J.B., N.B., M.L., A.B., F.E.)
| | - Simon H Williams
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, NY (B.H., S.H.W., K.J., A.O., J.A.G., N.M., T.B., W.I.L.)
| | - Komal Jain
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, NY (B.H., S.H.W., K.J., A.O., J.A.G., N.M., T.B., W.I.L.)
| | - Alexandra Oleynik
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, NY (B.H., S.H.W., K.J., A.O., J.A.G., N.M., T.B., W.I.L.)
| | - Dimitri Patriki
- University Hospital Zurich, Zurich, Switzerland (B.H., D.P., J.K, J.B., N.B., M.L., A.B., F.E.)
| | - Jan Kottwitz
- University Hospital Zurich, Zurich, Switzerland (B.H., D.P., J.K, J.B., N.B., M.L., A.B., F.E.)
| | - Jan Berg
- University Hospital Zurich, Zurich, Switzerland (B.H., D.P., J.K, J.B., N.B., M.L., A.B., F.E.)
| | - Joel A Garcia
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, NY (B.H., S.H.W., K.J., A.O., J.A.G., N.M., T.B., W.I.L.)
| | - Nora Baltensperger
- University Hospital Zurich, Zurich, Switzerland (B.H., D.P., J.K, J.B., N.B., M.L., A.B., F.E.)
| | - Marina Lovrinovic
- University Hospital Zurich, Zurich, Switzerland (B.H., D.P., J.K, J.B., N.B., M.L., A.B., F.E.)
| | - Andrea Baltensweiler
- University Hospital Zurich, Zurich, Switzerland (B.H., D.P., J.K, J.B., N.B., M.L., A.B., F.E.)
| | - Nishay Mishra
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, NY (B.H., S.H.W., K.J., A.O., J.A.G., N.M., T.B., W.I.L.)
| | - Thomas Briese
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, NY (B.H., S.H.W., K.J., A.O., J.A.G., N.M., T.B., W.I.L.)
| | - Paul J Hanson
- University of British Columbia, Vancouver, Canada (P.J.H., B.M.)
| | - Alexander Lauten
- Department of Cardiology, Charite University Hospital Berlin; Berlin Institute of Health (BIH), Berlin, Germany (B.H., A.L., W.P., D.L., U.L.)
| | - Wolfgang Poller
- Department of Cardiology, Charite University Hospital Berlin; Berlin Institute of Health (BIH), Berlin, Germany (B.H., A.L., W.P., D.L., U.L.)
| | - David M Leistner
- Department of Cardiology, Charite University Hospital Berlin; Berlin Institute of Health (BIH), Berlin, Germany (B.H., A.L., W.P., D.L., U.L.)
| | - Ulf Landmesser
- Department of Cardiology, Charite University Hospital Berlin; Berlin Institute of Health (BIH), Berlin, Germany (B.H., A.L., W.P., D.L., U.L.)
| | - Frank Enseleit
- University Hospital Zurich, Zurich, Switzerland (B.H., D.P., J.K, J.B., N.B., M.L., A.B., F.E.)
| | - Bruce McManus
- University of British Columbia, Vancouver, Canada (P.J.H., B.M.)
| | - Thomas F Lüscher
- University Hospital Zurich, Zurich, Switzerland (B.H., D.P., J.K, J.B., N.B., M.L., A.B., F.E.)
| | - W Ian Lipkin
- Royal Brompton and Harefield Hospitals and Imperial College, London, United Kingdom (T.F.L.).,University of Zurich, Center for Molecular Cardiology, Switzerland (T.F.L.)
| |
Collapse
|
34
|
Abstract
Myocarditis (MCD) is a type of inflammatory disease in which inflammatory cells infiltrate the myocardium, leading to cardiac dysfunction, myocardial necrosis, and fibrosis. Although it has been reported that MCD is mediated by T cells, the immune system is complex and includes many types of immune cells that interact with one another. Through investigations of the inflammatory responses in MCD including myocardial necrosis, fibrosis, and arrhythmia, we have gained further insight into the pathogenesis of MCD. This article aims to discuss the diversity and the roles of immune cells involved in the pathogenesis of MCD. Moreover, immunotherapy for the treatment of MCD remains controversial, and further investigation is required to identify accurate immunotherapies for special cell types.
Collapse
|
35
|
Di Florio DN, Sin J, Coronado MJ, Atwal PS, Fairweather D. Sex differences in inflammation, redox biology, mitochondria and autoimmunity. Redox Biol 2020; 31:101482. [PMID: 32197947 PMCID: PMC7212489 DOI: 10.1016/j.redox.2020.101482] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 02/19/2020] [Accepted: 02/27/2020] [Indexed: 02/07/2023] Open
Abstract
Autoimmune diseases are characterized by circulating antibodies and immune complexes directed against self-tissues that result in both systemic and organ-specific inflammation and pathology. Most autoimmune diseases occur more often in women than men. One exception is myocarditis, which is an inflammation of the myocardium that is typically caused by viral infections. Sex differences in the immune response and the role of the sex hormones estrogen and testosterone are well established based on animal models of autoimmune viral myocarditis as well as in mitochondrial function leading to reactive oxygen species production. RNA viruses like coxsackievirus B3, the primary cause of myocarditis in the US, activate the inflammasome through mitochondrial antiviral signaling protein located on the mitochondrial outer membrane. Toll-like receptor 4 and the inflammasome are the primary signaling pathways that increase inflammation during myocarditis, which is increased by testosterone. This review describes what is known about sex differences in inflammation, redox biology and mitochondrial function in the male-dominant autoimmune disease myocarditis and highlights gaps in the literature and future directions.
Collapse
Affiliation(s)
- Damian N Di Florio
- Center for Clinical and Translational Science, Mayo Clinic, Jacksonville, FL, USA.
| | - Jon Sin
- Cedars-Sinai Medical Center, Heart Institute, Los Angeles, CA, USA.
| | | | | | - DeLisa Fairweather
- Center for Clinical and Translational Science, Mayo Clinic, Jacksonville, FL, USA; Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA; Department of Immunology, Mayo Clinic, Jacksonville, FL, USA; Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
36
|
Myoglobin for Detection of High-Risk Patients with Acute Myocarditis. J Cardiovasc Transl Res 2020; 13:853-863. [PMID: 32006209 PMCID: PMC7541375 DOI: 10.1007/s12265-020-09957-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 01/14/2020] [Indexed: 12/12/2022]
Abstract
There is an unmet need for accurate and practical screening to detect myocarditis. We sought to test the hypothesis that the extent of acute myocarditis, measured by late gadolinium enhancement (LGE) on cardiac magnetic resonance imaging (CMR), can be estimated based on routine blood markers. A total of 44 patients were diagnosed with acute myocarditis and included in this study. There was strong correlation between myoglobin and LGE (rs = 0.73 [95% CI 0.51; 0.87], p < 0.001), while correlation was weak between LGE and TnT-hs (rs = 0.37 [95% CI 0.09; 0.61], p = 0.01). Receiver operating curve (ROC) analysis determined myoglobin ≥ 87 μg/L as cutoff to identify myocarditis (92% sensitivity, 80% specificity). The data were reproduced in an established model of coxsackievirus B3 myocarditis in mice (n = 26). These data suggest that myoglobin is an accurate marker of acute myocarditis. Graphical Abstract Receiver operating curve analysis determined myoglobin ≥ 87 μg/L as cutoff to identify myocarditis and these data were reproduced in an established model of coxsackievirus B3 myocarditis in mice: CMRI, cardiac magnetic resonance imaging; Mb, myoglobin; LGE, late gadolinium enhancement; ROC, receiver operating curve analysis.
Collapse
|
37
|
Abstract
The observation that heart failure with reduced ejection fraction is associated with elevated circulating levels of pro-inflammatory cytokines opened a new area of research that has revealed a potentially important role for the immune system in the pathogenesis of heart failure. However, until the publication in 2019 of the CANTOS trial findings on heart failure outcomes, all attempts to target inflammation in the heart failure setting in phase III clinical trials resulted in neutral effects or worsening of clinical outcomes. This lack of positive results in turn prompted questions on whether inflammation is a cause or consequence of heart failure. This Review summarizes the latest developments in our understanding of the role of the innate and adaptive immune systems in the pathogenesis of heart failure, and highlights the results of phase III clinical trials of therapies targeting inflammatory processes in the heart failure setting, such as anti-inflammatory and immunomodulatory strategies. The most recent of these studies, the CANTOS trial, raises the exciting possibility that, in the foreseeable future, we might be able to identify those patients with heart failure who have a cardio-inflammatory phenotype and will thus benefit from therapies targeting inflammation.
Collapse
|
38
|
Razzano D, Fallon JT. Myocarditis: somethings old and something new. Cardiovasc Pathol 2019; 44:107155. [PMID: 31760237 DOI: 10.1016/j.carpath.2019.107155] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 09/16/2019] [Accepted: 09/17/2019] [Indexed: 11/29/2022] Open
Abstract
"Since the pathological conditions take place at the cellular level, viral myocarditis and postinfectious autoimmunity can be suggested but not diagnosed clinically. All clinical methods including imaging techniques are misleading if infectious agents are involved. Accurate diagnosis demands simultaneous histologic, immunohistochemical, and molecular biological workup of the tissue. If the primary infectious or immune-mediated causes of the disease are carefully defined by clinical and biopsy-based tools, specific antiviral treatment options in addition to basic symptomatic therapy are available under certain conditions. These may allow a tailored cause-specific treatment that improves symptoms and prognosis of patients with acute and chronic disease." Uwe Kühl; Heinz-Peter SchultheissViral myocarditis.Swiss Medical Weekly. 144():w14010, JAN 2014 DOI:10.4414/smw.2014.14010.
Collapse
Affiliation(s)
- Dana Razzano
- New York Medical College at Westchester Medical Center, Valhalla, NY, 10595, USA.
| | | |
Collapse
|
39
|
Gonciarz W, Matusiak A, Rudnicka K, Rechciński T, Chałubiński M, Czkwianianc E, Broncel M, Gajewski A, Chmiela M. Autoantibodies to a specific peptide epitope of human Hsp60 (ATVLA) with homology to Helicobacter pylori HspB in H. pylori-infected patients. APMIS 2019; 127:139-149. [PMID: 30746790 DOI: 10.1111/apm.12925] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 12/27/2018] [Indexed: 02/06/2023]
Abstract
Helicobacter pylori (Hp) may initiate autoimmunity as a result of molecular mimicry. The aim of this study was to compare the level of IgG antibodies to a specific epitope (P1 peptide) of human heat shock protein (Hsp)60 homologous to Hp Hsp60 (HspB) in the sera of healthy donors (HD), patients with Hp-related gastritis or coronary heart disease (CHD), uninfected or with Hp infection confirmed by rapid urease test, histological examination (dyspeptic patients) the 13 C urea breath test (13 C UBT), and anti-Hp antibodies (healthy donors, CHD patients). The Anti-P1 IgG induction by Hp was verified by adsorption of sera with these bacteria and by experimental immunization of Caviae porcellus with Hp. Cytokine secretion by THP-1Blue™ monocytes in response to P1 was also assessed. Anti-P1 antibodies were detected in patients with gastritis or CHD infected with Hp and they were not found in uninfected individuals or asymptomatic carriers. No antibodies were raised against P2 in any group. Reduced cross-reactivity to P1 was exhibited by sera adsorbed with Hp. Caviae porcellus infected with Hp produced anti-P1 autoantibodies. THP-1XBlue™ monocytes responded to P1 by production of proinflammatory cytokines. Autoantibodies against P1 in Hp-positive patients with gastritis or CHD and upregulation of proinflammatory cytokines by P1 may contribute to the pathogenesis of Hp infection.
Collapse
Affiliation(s)
- Weronika Gonciarz
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Agnieszka Matusiak
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Karolina Rudnicka
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Tomasz Rechciński
- Department of Cardiology Biegański Regional Speciality Hospital, Medical University of Łódź, Łódź, Poland
| | - Maciej Chałubiński
- Clinic of Immunology, Rheumatology and Allergy of the Medical University of Łódź, Łódź, Poland
| | - Elżbieta Czkwianianc
- Department of Gastroenterology and Pediatrics, Polish Mother's Memorial Hospital, Łódź, Poland
| | - Marlena Broncel
- Department of Internal Diseases and Clinical Pharmacology, Laboratory of Tissue Immunopharmacology, Biegański Regional Speciality Hospital, Medical University of Łódź, Łódź, Poland
| | - Adrian Gajewski
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Magdalena Chmiela
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| |
Collapse
|
40
|
Kovacevic MM, Pejnovic N, Mitrovic S, Jovicic N, Petrovic I, Arsenijevic N, Lukic ML, Ljujic B. Galectin-3 deficiency enhances type 2 immune cell-mediated myocarditis in mice. Immunol Res 2018; 66:491-502. [PMID: 30099675 DOI: 10.1007/s12026-018-9013-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Experimental autoimmune myocarditis (EAM) is a mouse model of immune-mediated myocarditis and cardiomyopathy. The role of Galectin-3 (Gal-3), a β-galactoside-binding lectin, in autoimmune myocarditis has not been studied. Therefore, the aim of this study was to delineate the role of Gal-3 in myosin peptide-induced autoimmune myocarditis in mice. EAM was induced in relatively resistant C57BL/6J mice (wild type, WT) and in mice with a targeted deletion of Gal-3 gene (Gal-3KO) by immunization with myosin peptide MyHCα334-352. Gal-3KO mice developed more severe myocarditis and more pronounced heart hypertrophy than WT mice. Increased infiltration of CD45+ leucocytes, CD3+ T cells, F4/80+ macrophages, and eosinophils was observed in hearts of Gal-3KO mice compared to WT mice on day 21 after EAM induction. Moreover, hearts of Gal-3KO mice had more T helper type 2 (Th2) cells, alternatively activated M2 macrophages, higher amounts of IgG deposits, and higher serum levels of IL-4 and IL-33 than WT mice. Ablation of Gal-3 in Th1-dominant C57BL/6J mice that are relatively resistant to EAM resulted in more severe disease characterized by type 2 cardiac inflammation. The complex effects of Gal-3 on EAM progression might be important in the consideration of therapeutic options for the treatment of EAM.
Collapse
Affiliation(s)
- Marina Miletic Kovacevic
- Department of Histology and Embryology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nada Pejnovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Slobodanka Mitrovic
- Department of Pathology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nemanja Jovicic
- Department of Histology and Embryology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Ivica Petrovic
- Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nebojsa Arsenijevic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Miodrag L Lukic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia.
| | - Biljana Ljujic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
41
|
Nana-Leventaki E, Nana M, Poulianitis N, Sampaziotis D, Perrea D, Sanoudou D, Rontogianni D, Malliaras K. Cardiosphere-Derived Cells Attenuate Inflammation, Preserve Systolic Function, and Prevent Adverse Remodeling in Rat Hearts With Experimental Autoimmune Myocarditis. J Cardiovasc Pharmacol Ther 2018; 24:70-77. [DOI: 10.1177/1074248418784287] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Background: Cardiosphere-derived cells (CDCs) have yielded promising efficacy signals in early-phase clinical trials of ischemic and nonischemic cardiomyopathy. The potential efficacy of CDCs in acute myocarditis, an inflammatory cardiomyopathy without effective therapy, remains unexplored. Given that CDCs produce regenerative, cardioprotective, anti-inflammatory, and anti-fibrotic effects (all of which could be beneficial in acute myocarditis), we investigated the efficacy of intracoronary delivery of CDCs in a rat model of experimental autoimmune myocarditis. Methods: Lewis rats underwent induction of experimental autoimmune myocarditis by subcutaneous footpad injection of purified porcine cardiac myosin supplemented with Mycobacterium tuberculosis on days 1 and 7. On day 10, rats were randomly assigned to receive global intracoronary delivery of 500 000 CDCs or vehicle. Global intracoronary delivery was performed by injection of cells or vehicle into the left ventricular (LV) cavity during transient occlusion of the aortic root. Rats were euthanized 18 days after infusion. Cardiac volumes and systolic function were assessed by serial echocardiography, performed on days 1, 10, and 28. Myocardial inflammation, T-cell infiltration, and cardiac fibrosis were evaluated by histology. Results: Experimental autoimmune myocarditis was successfully induced in 14/14 rats that completed follow-up. Left ventricular ejection fraction (LVEF) and volumes were comparable on days 1 and 10 between groups. CDC infusion resulted in increased LVEF (81.5% ± 3% vs 65.4% ± 8%, P < .001) and decreased LV end-systolic volume (43 ± 15 vs 100 ± 24 μL, P < .001) compared to placebo administration at 18 days post-infusion. Cardiosphere-derived cell infusion decreased myocardial inflammation (7.4% ± 7% vs 20.7% ± 4% of myocardium, P = .007), cardiac fibrosis (16.6% ± 13% vs 38.1% ± 3% of myocardium, P = .008), and myocardial T-cell infiltration (30.4 ± 29 vs 125.8 ± 49 cells per field, P = .005) at 18 days post-infusion compared to placebo administration. Conclusion: Intracoronary delivery of CDCs attenuates myocardial inflammation, T-cell infiltration, and fibrosis while preventing myocarditis-induced systolic dysfunction and adverse remodeling in rats with experimental autoimmune myocarditis.
Collapse
Affiliation(s)
- E. Nana-Leventaki
- Third Department of Cardiology, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - M. Nana
- Third Department of Cardiology, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - N. Poulianitis
- Department of Pathology, Evangelismos Hospital, Athens, Greece
| | - D. Sampaziotis
- Department of Pathology, Evangelismos Hospital, Athens, Greece
| | - D. Perrea
- Laboratory for Experimental Surgery and Surgical Research “N.S. Christeas”, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - D. Sanoudou
- Molecular Biology Division, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - D. Rontogianni
- Department of Pathology, Evangelismos Hospital, Athens, Greece
| | - K. Malliaras
- Third Department of Cardiology, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| |
Collapse
|
42
|
Caruso JA, Carruthers N, Shin N, Gill R, Stemmer PM, Rosenspire A. Mercury alters endogenous phosphorylation profiles of SYK in murine B cells. BMC Immunol 2017; 18:37. [PMID: 28716125 PMCID: PMC5514489 DOI: 10.1186/s12865-017-0221-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 07/07/2017] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Epidemiological evidence and animal models suggest that exposure to low and non-neurotoxic concentrations of mercury may contribute to idiosyncratic autoimmune disease. Since defects in function and signaling in B cells are often associated with autoimmunity, we investigated whether mercury exposure might alter B cell responsiveness to self-antigens by interfering with B cell receptor (BCR) signal transduction. In this study we determined the effects of mercury on the protein tyrosine kinase SYK, a critical protein involved in regulation of the BCR signaling pathway. METHODS Phosphorylation sites of murine SYK were mapped before and after treatment of WEHI cell cultures with mercury, or with anti-IgM antibody (positive control) or pervanadate (a potent phosphatase inhibitor). Phosphopeptides were enriched by either titanium dioxide chromatography or anti-phosphotyrosine immunoaffinity, and analyzed by liquid chromatography-mass spectrometry. Select SYK phosphosite cluster regions were profiled for responsiveness to treatments using multiple reaction monitoring (MRM) methodology. RESULTS A total of 23 phosphosites were identified with high probability in endogenous SYK, including 19 tyrosine and 4 serine residues. For 10 of these sites phosphorylation levels were increased following BCR activation. Using MRM to profile changes in phosphorylation status we found that 4 cluster regions, encompassing 8 phosphosites, were activated by mercury and differentially responsive to all 3 treatments. Phosphorylation of tyrosine-342 and -346 residues were most sensitive to mercury exposure. This cluster is known to propagate normal BCR signal transduction by recruiting adaptor proteins such as PLC-γ and Vav-1 to SYK during formation of the BCR signalosome. CONCLUSIONS Our data shows that mercury alters the phosphorylation status of SYK on tyrosine sites known to have a role in promoting BCR signals. Considering the importance of SYK in the BCR signaling pathway, these data suggest that mercury can alter BCR signaling in B cells, which might affect B cell responsiveness to self-antigen and have implications with respect to autoimmunity and autoimmune disease.
Collapse
Affiliation(s)
- Joseph A Caruso
- Institute of Environmental Health Sciences, Center for Urban Responses to Environmental Stressors (CURES), Wayne State University, Detroit, MI, 48201, USA.
| | - Nicholas Carruthers
- Institute of Environmental Health Sciences, Center for Urban Responses to Environmental Stressors (CURES), Wayne State University, Detroit, MI, 48201, USA
| | - Namhee Shin
- Institute of Environmental Health Sciences, Center for Urban Responses to Environmental Stressors (CURES), Wayne State University, Detroit, MI, 48201, USA
| | - Randal Gill
- Department of Immunology and Microbiology, Center for Urban Responses to Environmental Stressors (CURES), Wayne State University, Detroit, MI, 48201, USA
| | - Paul M Stemmer
- Institute of Environmental Health Sciences, Center for Urban Responses to Environmental Stressors (CURES), Wayne State University, Detroit, MI, 48201, USA
| | - Allen Rosenspire
- Department of Immunology and Microbiology, Center for Urban Responses to Environmental Stressors (CURES), Wayne State University, Detroit, MI, 48201, USA
| |
Collapse
|
43
|
Azizi G, Ziaee V, Tavakol M, Alinia T, Yazdai R, Mohammadi H, Abolhassani H, Aghamohammadi A. Approach to the Management of Autoimmunity in Primary Immunodeficiency. Scand J Immunol 2017; 85:13-29. [PMID: 27862144 DOI: 10.1111/sji.12506] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 11/04/2016] [Indexed: 12/14/2022]
Abstract
Primary immunodeficiency diseases (PIDs) consist of a genetically heterogeneous group of immune disorders that affect distinct elements of the immune system. PID patients are more prone to infections and non-infectious complications, particularly autoimmunity. The concomitance of immunodeficiency and autoimmunity appears to be paradoxical and leads to difficulty in the management of autoimmune complications in PID patients. Therefore, management of autoimmunity in patients with PID requires special considerations because dysregulations and dysfunctions of the immune system along with persistent inflammation impair the process of diagnosis and treatment.
Collapse
Affiliation(s)
- G Azizi
- Department of Laboratory Medicine, Imam Hassan Mojtaba Hospital, Alborz University of Medical Sciences, Karaj, Iran.,Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - V Ziaee
- Pediatric Rheumatology Research Group, Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pediatrics, Tehran University of Medical Sciences, Tehran, Iran
| | - M Tavakol
- Department of Allergy and Clinical Immunology, Shahid Bahonar Hospital, Alborz University of Medical Sciences, Karaj, Iran
| | - T Alinia
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - R Yazdai
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - H Mohammadi
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - H Abolhassani
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - A Aghamohammadi
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
44
|
Bracamonte-Baran W, Čiháková D. Cardiac Autoimmunity: Myocarditis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1003:187-221. [PMID: 28667560 DOI: 10.1007/978-3-319-57613-8_10] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Myocarditis is the inflammation of the muscle tissues of the heart (myocardium). After a pathologic cardiac-specific inflammatory process, it may progress to chronic damage and dilated cardiomyopathy. The latter is characterized by systolic dysfunction, whose clinical correlate is heart failure. Nevertheless, other acute complications may arise as consequence of tissue damage and electrophysiologic disturbances. Different etiologies are involved in triggering myocarditis. In some cases, such as giant cell myocarditis or eosinophilic necrotizing myocarditis, it is an autoimmune process. Several factors predispose the development of autoimmune myocarditis such as systemic/local primary autoimmunity, viral infection, HLA and gender bias, exposure of cryptic antigens, mimicry, and deficient thymic training/Treg induction. Once the anti-myocardium autoimmune process is triggered, several components of the immune response orchestrate a sustained attack toward myocardial tissues with particular timing and immunopathogenic features. Innate response mediated by monocytes/macrophages, neutrophils, and eosinophils parallels the adaptive response, playing a final effector role and not only a priming function. Stromal cells like fibroblast are also involved in the process through specific cytokines. Furthermore, adaptive T cell responses have anti-paradigmatic features, as Th17 response is dispensable for acute myocarditis but is the main driver of the process leading to dilated cardiomyopathy. Humoral response, thought to be a bystander, is important in the appearance of late-stage hemodynamic complications. The complexity of that process, as well as the unspecific and variable clinical presentation, had generated difficulties for diagnosis and treatment, which remain suboptimal. In this chapter, we will discuss the most relevant immunopathogenic findings from a basic science and clinical perspective.
Collapse
Affiliation(s)
- William Bracamonte-Baran
- Department of Pathology, Division of Immunology, Johns Hopkins University School of Medicine, 720 Rutland Ave., Baltimore, MD, 21205, USA
| | - Daniela Čiháková
- Division of Immunology, Department of Pathology, Johns Hopkins University School of Medicine, 720 Rutland Ave., Baltimore, MD, 21205, USA. .,W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, 21205, USA.
| |
Collapse
|
45
|
Azizi G, Ahmadi M, Abolhassani H, Yazdani R, Mohammadi H, Mirshafiey A, Rezaei N, Aghamohammadi A. Autoimmunity in Primary Antibody Deficiencies. Int Arch Allergy Immunol 2016; 171:180-193. [DOI: 10.1159/000453263] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
46
|
Azizi G, Pouyani MR, Abolhassani H, Sharifi L, dizaji MZ, Mohammadi J, Mirshafiey A, Aghamohammadi A. Cellular and molecular mechanisms of immune dysregulation and autoimmunity. Cell Immunol 2016; 310:14-26. [DOI: 10.1016/j.cellimm.2016.08.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 08/18/2016] [Accepted: 08/25/2016] [Indexed: 12/22/2022]
|
47
|
Heymans S, Eriksson U, Lehtonen J, Cooper LT. The Quest for New Approaches in Myocarditis and Inflammatory Cardiomyopathy. J Am Coll Cardiol 2016; 68:2348-2364. [PMID: 27884253 DOI: 10.1016/j.jacc.2016.09.937] [Citation(s) in RCA: 243] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 09/22/2016] [Accepted: 09/26/2016] [Indexed: 12/15/2022]
Abstract
Myocarditis is a diverse group of heart-specific immune processes classified by clinical and histopathological manifestations. Up to 40% of dilated cardiomyopathy is associated with inflammation or viral infection. Recent experimental studies revealed complex regulatory roles for several microribonucleic acids and T-cell and macrophage subtypes. Although the prevalence of myocarditis remained stable between 1990 and 2013 at about 22 per 100,000 people, overall mortality from cardiomyopathy and myocarditis has decreased since 2005. The diagnostic and prognostic value of cardiac magnetic resonance has increased with new, higher-sensitivity sequences. Positron emission tomography has emerged as a useful tool for diagnosis of cardiac sarcoidosis. The sensitivity of endomyocardial biopsy may be increased, especially in suspected sarcoidosis, by the use of electrogram guidance to target regions of abnormal signal. Investigational treatments on the basis of mechanistic advances are entering clinical trials. Revised management recommendations regarding athletic participation after acute myocarditis have heightened the importance of early diagnosis.
Collapse
Affiliation(s)
- Stephane Heymans
- Department of Cardiology, CARIM, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Urs Eriksson
- GZO Regional Health Center, Wetzikon & Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | | | - Leslie T Cooper
- Cardiovascular Department, Mayo Clinic, Jacksonville, Florida.
| |
Collapse
|
48
|
Natural killer cells in inflammatory heart disease. Clin Immunol 2016; 175:26-33. [PMID: 27894980 DOI: 10.1016/j.clim.2016.11.010] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 10/09/2016] [Accepted: 11/20/2016] [Indexed: 02/07/2023]
Abstract
Despite of a multitude of excellent studies, the regulatory role of natural killer (NK) cells in the pathogenesis of inflammatory cardiac disease is greatly underappreciated. Clinical abnormalities in the numbers and functions of NK cells are observed in myocarditis and inflammatory dilated cardiomyopathy (DCMi) as well as in cardiac transplant rejection [1-6]. Because treatment of these disorders remains largely symptomatic in nature, patients have little options for targeted therapies [7,8]. However, blockade of NK cells and their receptors can protect against inflammation and damage in animal models of cardiac injury and inflammation. In these models, NK cells suppress the maturation and trafficking of inflammatory cells, alter the local cytokine and chemokine environments, and induce apoptosis in nearby resident and hematopoietic cells [1,9,10]. This review will dissect each protective mechanism employed by NK cells and explore how their properties might be exploited for their therapeutic potential.
Collapse
|
49
|
Azizi G, Abolhassani H, Asgardoon MH, Alinia T, Yazdani R, Mohammadi J, Rezaei N, Ochs HD, Aghamohammadi A. Autoimmunity in common variable immunodeficiency: epidemiology, pathophysiology and management. Expert Rev Clin Immunol 2016; 13:101-115. [DOI: 10.1080/1744666x.2016.1224664] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Gholamreza Azizi
- Department of Laboratory Medicine, Imam Hassan Mojtaba Hospital, Alborz University of Medical Sciences, Karaj, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hassan Abolhassani
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Mohammad Hosein Asgardoon
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Iranian Student Society for Immunodeficiencies, Student’s Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Tina Alinia
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Yazdani
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Javad Mohammadi
- Department of Biomedical Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hans D. Ochs
- Seattle Children’s Research Institute and Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Asghar Aghamohammadi
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
50
|
Lee WS, Erdelyi K, Matyas C, Mukhopadhyay P, Varga ZV, Liaudet L, Hask’ G, ’iháková D, Mechoulam R, Pacher P. Cannabidiol Limits T Cell-Mediated Chronic Autoimmune Myocarditis: Implications to Autoimmune Disorders and Organ Transplantation. Mol Med 2016; 22:136-146. [PMID: 26772776 PMCID: PMC5004721 DOI: 10.2119/molmed.2016.00007] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 01/07/2016] [Indexed: 12/22/2022] Open
Abstract
Myocarditis is a major cause of heart failure and sudden cardiac death in young adults and adolescents. Many cases of myocarditis are associated with autoimmune processes in which cardiac myosin is a major autoantigen. Conventional immunosuppressive therapies often provide unsatisfactory results and are associated with adverse toxicities during the treatment of autoimmune myocarditis. Cannabidiol (CBD) is a nonpsychoactive constituent of marijuana that exerts antiinflammatory effects independent of classical cannabinoid receptors. Recently, 80 clinical trials have investigated the effects of CBD in various diseases from inflammatory bowel disease to graft versus host disease. CBD-based formulations are used for the management of multiple sclerosis in numerous countries, and CBD also received U.S. Food and Drug Administration approval for the treatment of refractory childhood epilepsy and glioblastoma multiforme. Herein, using a well-established mouse model of experimental autoimmune myocarditis (EAM) induced by immunization with cardiac myosin emmulsified in adjuvant resulting in T cell-mediated inflammation, cardiomyocyte cell death, fibrosis and myocardial dysfunction, we studied the potential beneficial effects of CBD. EAM was characterized by marked myocardial T-cell infiltration, profound inflammatory response and fibrosis (measured by quantitative real-time polymerase chain reaction, histology and immunohistochemistry analyses) accompanied by marked attenuation of both systolic and diastolic cardiac functions measured with a pressure-volume conductance catheter technique. Chronic treatment with CBD largely attenuated the CD3+ and CD4+ T cell-mediated inflammatory response and injury, myocardial fibrosis and cardiac dysfunction in mice. In conclusion, CBD may represent a promising novel treatment for managing autoimmune myocarditis and possibly other autoimmune disorders and organ transplantation.
Collapse
Affiliation(s)
- Wen-Shin Lee
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States of America
- Division of General Medicine, Department of Medicine, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Katalin Erdelyi
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Csaba Matyas
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States of America
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Partha Mukhopadhyay
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Zoltan V Varga
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lucas Liaudet
- Department of Intensive Care Medicine, University Hospital Medical Center, Lausanne, Switzerland
| | - György Hask’
- Department of Surgery, Rutgers New Jersey Medical School, Newark, New Jersey, United States of America
| | - Daniela ’iháková
- Department of Pathology and The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University School of Medicine and School of Public Health, Baltimore, Maryland, United States of America
| | - Raphael Mechoulam
- Department for Medicinal Chemistry and Natural Products, Faculty of Medicine, Hebrew University of Jerusalem, EinKerem, Jerusalem, Israel
| | - Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|