1
|
Jain L, Kaur D, Khalil S, Pradhan P, Malik M, Dogra S, Kaur K, Mathew JL. Efficacy and Safety of Intravenous Immunoglobulin (IVIg) in Acute Viral Myocarditis in Children: A Systematic Review of Randomized Controlled Trials. Indian Pediatr 2025; 62:56-62. [PMID: 39754433 DOI: 10.1007/s13312-025-3359-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 11/13/2024] [Indexed: 01/06/2025]
Abstract
CONTEXT Acute myocarditis is a rare but potentially life-threatening condition in infants and children. While immunosuppressive agents have shown limited effectiveness, intravenous immunoglobulin (IVIg) holds promise as a treatment option. OBJECTIVE To study the efficacy and safety of intravenous immunoglobulin (IVIg) in treating acute viral myocarditis in children. EVIDENCE ACQUISITION We searched five databases including PubMed, EMBASE, the Cochrane Library, Scopus, and Web of Science; and four trial registries for published studies on the topic. Grey literature was searched through ProQuest and Open Grey databases. The studies eligible for this review were randomized controlled trials in children (< 18 years) with acute viral myocarditis (Population), comparing IVIg (Intervention), versus no IVIg i.e., any other treatment or placebo (Comparator), for efficacy and safety (Outcomes). RESULTS The literature search identified a total of 9,524 records. Two reviewers independently screened these records. A total of 73 citations were deemed potentially eligible, all of which were non-RCTs or review articles on full text examination. There were no RCTs identified to address the review question. CONCLUSION There are no RCTs in children comparing the efficacy and safety of IVIg treatment in acute viral myocarditis. Prospective randomized trials are urgently required.
Collapse
Affiliation(s)
- Lovely Jain
- Indian Council of Medical Research (ICMR) Advanced Centre for Evidence Based Child Health (ACEBCH), India
| | - Davinder Kaur
- Indian Council of Medical Research (ICMR) Advanced Centre for Evidence Based Child Health (ACEBCH), India
| | - Sumaira Khalil
- Department of Pediatrics, University College of Medical Sciences, Delhi, India
| | - Pranita Pradhan
- Indian Council of Medical Research (ICMR) Advanced Centre for Evidence Based Child Health (ACEBCH), India
| | - Meenakshi Malik
- Indian Council of Medical Research (ICMR) Advanced Centre for Evidence Based Child Health (ACEBCH), India
| | - Sarita Dogra
- Indian Council of Medical Research (ICMR) Advanced Centre for Evidence Based Child Health (ACEBCH), India
| | - Kulbir Kaur
- Indian Council of Medical Research (ICMR) Advanced Centre for Evidence Based Child Health (ACEBCH), India
| | - Joseph L Mathew
- Indian Council of Medical Research (ICMR) Advanced Centre for Evidence Based Child Health (ACEBCH) and Department of Pediatrics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| |
Collapse
|
2
|
Xu Y, Tan Y, Peng Z, Liu M, Zhang B, Wei K. Advancing Myocarditis Research: Evaluating Animal Models for Enhanced Pathophysiological Insights. Curr Cardiol Rep 2025; 27:6. [PMID: 39775161 DOI: 10.1007/s11886-024-02182-8] [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] [Accepted: 10/08/2024] [Indexed: 01/11/2025]
Abstract
PURPOSE OF REVIEW This review aims to assess the current landscape of animal models used in myocarditis research, with a focus on understanding their utility in uncovering the pathophysiology of the disease. The goal is to evaluate these models' strengths and weaknesses and propose optimizations to make them more relevant and reliable for both mechanistic studies and therapeutic interventions in myocarditis. RECENT FINDINGS Recent studies have primarily utilized animal models, particularly viral and autoimmune myocarditis models, to study disease mechanisms. Coxsackievirus remains the most common virus used in viral myocarditis models, offering high success rates but limited applicability to human cases due to differences in infection patterns. Autoimmune myocarditis models, often involving humanized mice, have made strides in mimicking human immune responses but still face challenges in accuracy and clinical relevance. COVID-19 has introduced new avenues for research, especially concerning vaccine-induced myocarditis, although findings remain preliminary. Animal models remain crucial for myocarditis research, but each comes with distinct challenges. Viral models excel in success rate but suffer from partial relevance to human conditions. Autoimmune models are useful in immunological studies, though costly and less replicable. Vaccine-associated models are closely related to modern clinical conditions, but lack theoretical support and therefore lack reliability. Optimizing these models could improve our understanding of myocarditis and lead to more effective treatments. Future research should aim to refine these models to better simulate human conditions and enhance their clinical applicability, ultimately advancing the diagnosis and treatment of myocarditis.
Collapse
Affiliation(s)
- Yanzhe Xu
- Medical College, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Yixing Tan
- Medical College, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Zhonghui Peng
- Medical College, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Meiyu Liu
- Medical College, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Bi Zhang
- Medical College, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| | - Ke Wei
- Medical College, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| |
Collapse
|
3
|
Ono K, Fujiwara H. Tribute to Dr. Chuichi Kawai - A Great Cardiologist in Japan. Circ J 2024; 89:2-5. [PMID: 39647899 DOI: 10.1253/circj.cj-66-0232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Affiliation(s)
- Koh Ono
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University
| | - Hisayoshi Fujiwara
- Gifu University
- Hyogo Prefectural Amagasaki General Medical Center
- Ookuma Hospital
| |
Collapse
|
4
|
Omori T, Maruyama K, Ohta-Ogo K, Hatakeyama K, Ishibashi-Ueda H, Onoue K, Nagai T, Kato S, Okumura T, Oikawa M, Amiya E, Yoshizawa S, Suzuki T, Goto H, Nakamura K, Fujino T, Moriwaki K, Nakamori S, Anzai T, Sakata Y, Hiroe M, Imanaka-Yoshida K, Dohi K. Clinical and Histopathological Characteristics of Patients With Myocarditis After mRNA COVID-19 Vaccination. Circ J 2024; 89:120-129. [PMID: 39496392 DOI: 10.1253/circj.cj-24-0506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2024]
Abstract
BACKGROUND The effects of myocarditis after mRNA COVID-19 vaccination (mCV) on myocardial tissue, and the association between cardiomyocyte injury and clinical presentation, are not fully understood. METHODS AND RESULTS We retrospectively registered patients clinically diagnosed with myocarditis after the first or second mCV who underwent endomyocardial biopsy or autopsy from 42 participating centers in Japan. We investigated the histological features and their association with clinical presentation based on cardiomyocyte injury. Forty patients who underwent endomyocardial biopsy were included in the study. Of these, 19 (47.5%) showed mild lymphocytic infiltration and interstitial edema without cardiomyocyte injury. The remaining 21 (52.5%) patients showed cardiomyocyte injury accompanied by infiltrating inflammatory cells: 11 with lymphocytic infiltration, 7 with eosinophilic infiltration, and 3 with myocarditis with both lymphocyte and eosinophil infiltration. Compared with patients without cardiomyocyte injury, those with cardiomyocyte injury were clinically characterized by older age, a balanced sex distribution, less frequent chest pain, and a lower left ventricular ejection fraction. Fifteen of 21 (71.4%) patients with cardiomyocyte injury developed fulminant myocarditis, with 13 (86.7%) requiring mechanical circulatory support; in contrast, none of those without cardiomyocyte injury developed fulminant myocarditis (P<0.001). CONCLUSIONS Our histological examination of patients with myocarditis after mCV revealed varying degrees of cardiomyocyte injury, ranging from pronounced to absent, along with various types of myocarditis. Cardiomyocyte injury was strongly associated with the severity of myocarditis.
Collapse
Affiliation(s)
- Taku Omori
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine
- Department of Histology and Cell Biology, Mie University Graduate School of Medicine
| | - Kazuaki Maruyama
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine
| | - Keiko Ohta-Ogo
- Department of Pathology, National Cerebral and Cardiovascular Center
| | - Kinta Hatakeyama
- Department of Pathology, National Cerebral and Cardiovascular Center
| | - Hatsue Ishibashi-Ueda
- Department of Pathology, National Cerebral and Cardiovascular Center
- Department of Diagnostic Pathology, Hokusetsu General Hospital
| | - Kenji Onoue
- Department of Cardiovascular Medicine, Nara Medical University
| | - Toshiyuki Nagai
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Seiya Kato
- Division of Pathology, Saiseikai Fukuoka General Hospital
| | - Takahiro Okumura
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | - Masayoshi Oikawa
- Department of Cardiovascular Medicine, Fukushima Medical University
| | - Eisuke Amiya
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| | - Saeko Yoshizawa
- Department of Surgical Pathology, Tokyo Women's Medical University
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases
| | - Hidemasa Goto
- Department of Histology and Cell Biology, Mie University Graduate School of Medicine
| | - Kazufumi Nakamura
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences
| | - Takeo Fujino
- Department of Advanced Cardiopulmonary Failure, Faculty of Medical Sciences, Kyushu University
| | - Keishi Moriwaki
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine
| | - Shiro Nakamori
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine
| | - Toshihisa Anzai
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Michiaki Hiroe
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine
- Department of Cardiology, National Center for Global Health and Medicine
| | - Kyoko Imanaka-Yoshida
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine
| | - Kaoru Dohi
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine
| |
Collapse
|
5
|
Van Name J, Wu K, Xi L. Myocarditis - A silent killer in athletes: Comparative analysis on the evidence before and after COVID-19 pandemic. SPORTS MEDICINE AND HEALTH SCIENCE 2024; 6:232-239. [PMID: 39234482 PMCID: PMC11369839 DOI: 10.1016/j.smhs.2024.03.003] [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: 12/11/2023] [Revised: 03/06/2024] [Accepted: 03/12/2024] [Indexed: 09/06/2024] Open
Abstract
Myocarditis is a rare cardiomyocyte inflammatory process, typically caused by viruses, with potentially devastating cardiac sequalae in both competitive athletes and in the general population. Investigation into myocarditis prevalence in the Coronavirus disease 2019 (COVID-19) era suggests that infection with Severe acute respiratory syndrome coronavirus (SARS-CoV-2) is an independent risk factor for myocarditis, which is confirmed mainly through cardiovascular magnetic resonance imaging. Recent studies indicated that athletes have a decreased risk of myocarditis after recent COVID-19 infection compared to the general population. However, given the unique nature of competitive athletics with their frequent participation in high-intensity exercise, athletes possess distinct factors of susceptibility for the development of myocarditis and its subsequent severe cardiac complications (e.g., sudden cardiac death, fulminant heart failure, etc.). Under this context, this review focuses on comparing myocarditis in athletes versus non-athletes, owing special attention to the distinct clinical presentations and outcomes of myocarditis caused by different viral pathogens such as cytomegalovirus, Epstein-Barr virus, human herpesvirus-6, human immunodeficiency virus, and Parvovirus B19, both before and after the COVID-19 pandemic, as compared with SARS-CoV-2. By illustrating distinct clinical presentations and outcomes of myocarditis in athletes versus non-athletes, we also highlight the critical importance of early detection, vigilant monitoring, and effective management of viral and non-viral myocarditis in athletes and the necessity for further optimization of the return-to-play guidelines for athletes in the COVID-19 era, in order to minimize the risks for the rare but devastating cardiac fatality.
Collapse
Affiliation(s)
- Jonathan Van Name
- Virginia Commonwealth University School of Medicine (M.D. Class 2024), Richmond, VA, 23298, USA
| | - Kainuo Wu
- Virginia Commonwealth University School of Medicine (M.D. Class 2024), Richmond, VA, 23298, USA
| | - Lei Xi
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, 23298-0204, USA
| |
Collapse
|
6
|
Thiene G. Storytelling of Myocarditis. Biomedicines 2024; 12:832. [PMID: 38672187 PMCID: PMC11048135 DOI: 10.3390/biomedicines12040832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/20/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
In 1900, Fiedler first reported autopsy cases with peculiar inflammation of the myocardium, which he named interstitial myocarditis. He postulated an isolated cardiac inflammation of the myocardium in the absence of multiorgan involvement and with a poor prognosis due to invisible microorganisms, which years later would have been identified as viruses. The revision of original histologic sections by Schmorl showed cases with lymphocytes and others with giant-cell inflammatory histotypes. The in vivo diagnosis of myocarditis became possible thanks to right cardiac catheterization with endomyocardial biopsy (EMB). The gold standard for diagnosis was achieved with the employment of immunohistochemistry and molecular investigation by Polymerase Chain Reaction (PCR), which allows for the detection of viruses as causal agents. Both RNA and DNA were revealed to be cardiotropic, with a common receptor (CAR). A protease, coded by coxsackie virus, disrupts the cytoskeleton and accounts for cell death. Unfortunately, vaccination, despite having been revealed to be effective in animal experiments, has not yet entered the clinical field for prevention. Cardiac Magnetic Resonance turned out to be a revolutionary tool for in vivo diagnosis through the detection of edema (inflammatory exudate). Myocarditis may be fulminant in terms of clinical presentation but not necessarily fatal. The application of ExtraCorporeal Membrane Oxygenation (ECMO) allows for relieving the overloaded native heart.
Collapse
Affiliation(s)
- Gaetano Thiene
- Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padua, 35121 Padua, Italy
| |
Collapse
|
7
|
Ediger DS, Brady WJ, Koyfman A, Long B. High risk and low prevalence diseases: Myocarditis. Am J Emerg Med 2024; 78:81-88. [PMID: 38241774 DOI: 10.1016/j.ajem.2024.01.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 01/02/2024] [Indexed: 01/21/2024] Open
Abstract
INTRODUCTION Myocarditis is a serious condition that carries with it a high rate of morbidity and mortality. OBJECTIVE This review highlights the pearls and pitfalls of myocarditis, including presentation, diagnosis, and management in the emergency department (ED) based on current evidence. DISCUSSION Myocarditis is an inflammatory syndrome of myocardium, most often resulting from a viral infection, that can cause life-threatening cardiovascular collapse. It has a highly variable presentation and no widely available specific diagnostic test, making it a challenging diagnosis. Emergency clinicians should obtain an electrocardiogram and perform bedside ultrasound to assess cardiac function. Treatment in the ED is largely supportive, focusing on resuscitation, cardiovascular support, cardiology specialist consultation, and appropriate disposition. CONCLUSIONS An understanding of myocarditis can assist emergency clinicians in diagnosing and managing this potentially deadly disease.
Collapse
Affiliation(s)
- David S Ediger
- Department of Emergency Medicine, Brooke Army Medical Center, Fort Sam Houston, TX, USA
| | - William J Brady
- Professor, Department of Emergency Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA.
| | - Alex Koyfman
- Department of Emergency Medicine, UT Southwestern, Dallas, TX, USA
| | - Brit Long
- Department of Emergency Medicine, Brooke Army Medical Center, Fort Sam Houston, TX, USA.
| |
Collapse
|
8
|
Panagiotides NG, Poledniczek M, Andreas M, Hülsmann M, Kocher AA, Kopp CW, Piechota-Polanczyk A, Weidenhammer A, Pavo N, Wadowski PP. Myocardial Oedema as a Consequence of Viral Infection and Persistence-A Narrative Review with Focus on COVID-19 and Post COVID Sequelae. Viruses 2024; 16:121. [PMID: 38257821 PMCID: PMC10818479 DOI: 10.3390/v16010121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/02/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Microvascular integrity is a critical factor in myocardial fluid homeostasis. The subtle equilibrium between capillary filtration and lymphatic fluid removal is disturbed during pathological processes leading to inflammation, but also in hypoxia or due to alterations in vascular perfusion and coagulability. The degradation of the glycocalyx as the main component of the endothelial filtration barrier as well as pericyte disintegration results in the accumulation of interstitial and intracellular water. Moreover, lymphatic dysfunction evokes an increase in metabolic waste products, cytokines and inflammatory cells in the interstitial space contributing to myocardial oedema formation. This leads to myocardial stiffness and impaired contractility, eventually resulting in cardiomyocyte apoptosis, myocardial remodelling and fibrosis. The following article reviews pathophysiological inflammatory processes leading to myocardial oedema including myocarditis, ischaemia-reperfusion injury and viral infections with a special focus on the pathomechanisms evoked by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. In addition, clinical implications including potential long-term effects due to viral persistence (long COVID), as well as treatment options, are discussed.
Collapse
Affiliation(s)
- Noel G. Panagiotides
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
| | - Michael Poledniczek
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria;
| | - Martin Andreas
- Department of Cardiac Surgery, Medical University of Vienna, 1090 Vienna, Austria; (M.A.); (A.A.K.)
| | - Martin Hülsmann
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
| | - Alfred A. Kocher
- Department of Cardiac Surgery, Medical University of Vienna, 1090 Vienna, Austria; (M.A.); (A.A.K.)
| | - Christoph W. Kopp
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria;
| | | | - Annika Weidenhammer
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
| | - Noemi Pavo
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
| | - Patricia P. Wadowski
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria;
| |
Collapse
|
9
|
Zhou Z, Zhang M, Zhao C, Gao X, Wen Z, Wu J, Chen C, Fleming I, Hu J, Wang DW. Epoxyeicosatrienoic Acids Prevent Cardiac Dysfunction in Viral Myocarditis via Interferon Type I Signaling. Circ Res 2023; 133:772-788. [PMID: 37681352 DOI: 10.1161/circresaha.123.322619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 08/30/2023] [Indexed: 09/09/2023]
Abstract
Myocarditis is a challenging inflammatory disease of the heart, and better understanding of its pathogenesis is needed to develop specific drug therapies. Epoxyeicosatrienoic acids (EETs), active molecules synthesized by CYP (cytochrome P450) enzymes from arachidonic acids and hydrolyzed to less active dihydroxyeicosatrienoic acids by sEH (soluble epoxide hydrolase), have been attributed anti-inflammatory activity. Here, we investigated whether EETs have immunomodulatory activity and exert protective effects on coxsackie B3 virus-induced myocarditis. Viral infection altered eicosanoid epoxide and diol levels in both patients with myocarditis and in the murine heart and correlated with the increased expression and activity of sEH after coxsackie B3 virus infection. Administration of a sEH inhibitor prevented coxsackie B3 virus-induced cardiac dysfunction and inflammatory infiltration. Importantly, EET/sEH inhibitor treatment attenuated viral infection or improved viral resistance by activating type I IFN (interferon) signaling. At the molecular level, EETs enhanced the interaction between GSK3β (glycogen synthase kinase-3 beta) and TBK1 (TANK-binding kinase 1) to promote IFN-β production. Our findings revealed that EETs and sEH inhibitors prevent the progress of coxsackie B3 virus-induced myocarditis, particularly by promoting viral resistance by increasing IFN production.
Collapse
Affiliation(s)
- Zhou Zhou
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.)
| | - Min Zhang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.)
| | - Chengcheng Zhao
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.)
| | - Xu Gao
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.)
| | - Zheng Wen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.)
| | - Junfang Wu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.)
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.)
| | - Ingrid Fleming
- Sino-German Laboratory of CardioPulmonary Science (I.F., J.H., D.W.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute for Vascular Signalling, Goethe University, Frankfurt am Main, Germany (I.F., J.H.)
- German Center of Cardiovascular Research, Partner Site RheinMain, Frankfurt am Main, Germany (I.F., J.H.)
| | - Jiong Hu
- Department of Histology and Embryology, School of Basic Medicine (J.H.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Sino-German Laboratory of CardioPulmonary Science (I.F., J.H., D.W.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute for Vascular Signalling, Goethe University, Frankfurt am Main, Germany (I.F., J.H.)
- German Center of Cardiovascular Research, Partner Site RheinMain, Frankfurt am Main, Germany (I.F., J.H.)
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Sino-German Laboratory of CardioPulmonary Science (I.F., J.H., D.W.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.)
| |
Collapse
|
10
|
Wang SS, Zhu XX, Wu XY, Zhang WW, Ding YD, Jin SW, Zhang PH. Interaction Between Blood Vasculatures and Lymphatic Vasculatures During Inflammation. J Inflamm Res 2023; 16:3271-3281. [PMID: 37560514 PMCID: PMC10408656 DOI: 10.2147/jir.s414891] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 07/21/2023] [Indexed: 08/11/2023] Open
Abstract
Physiological activity cannot be regulated without the blood and lymphatic vasculatures, which play complementary roles in maintaining the body's homeostasis and immune responses. Inflammation is the body's initial response to pathological injury and is responsible for protecting the body, removing damaged tissues, and restoring and maintaining homeostasis in the body. A growing number of researches have shown that blood and lymphatic vessels play an essential role in a variety of inflammatory diseases. In the inflammatory state, the permeability of blood vessels and lymphatic vessels is altered, and angiogenesis and lymphangiogenesis subsequently occur. The blood vascular and lymphatic vascular systems interact to determine the development or resolution of inflammation. In this review, we discuss the changes that occur in the blood vascular and lymphatic vascular systems of several organs during inflammation, describe the different scenarios of angiogenesis and lymphangiogenesis at different sites of inflammation, and demonstrate the prospect of targeting the blood vasculature and lymphatic vasculature systems to limit the development of inflammation and promote the resolution of inflammation in inflammatory diseases.
Collapse
Affiliation(s)
- Shun-Shun Wang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
- Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
| | - Xin-Xu Zhu
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
- Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
| | - Xin-Yi Wu
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
- Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
| | - Wen-Wu Zhang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
- Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
| | - Yang-Dong Ding
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
- Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
| | - Sheng-Wei Jin
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
- Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
| | - Pu-Hong Zhang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
- Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
| |
Collapse
|
11
|
Interdonato L, Impellizzeri D, D’Amico R, Cordaro M, Siracusa R, D’Agostino M, Genovese T, Gugliandolo E, Crupi R, Fusco R, Cuzzocrea S, Di Paola R. Modulation of TLR4/NFκB Pathways in Autoimmune Myocarditis. Antioxidants (Basel) 2023; 12:1507. [PMID: 37627502 PMCID: PMC10451772 DOI: 10.3390/antiox12081507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/14/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
Myocarditis is an inflammatory and oxidative disorder characterized by immune cell recruitment in the damaged tissue and organ dysfunction. In this paper, we evaluated the molecular pathways involved in myocarditis using a natural compound, Coriolus versicolor, in an experimental model of autoimmune myocarditis (EAM). Animals were immunized with an emulsion of pig cardiac myosin and complete Freund's adjuvant supplemented with mycobacterium tuberculosis; thereafter, Coriolus versicolor (200 mg/Kg) was orally administered for 21 days. At the end of the experiment, blood pressure and heart rate measurements were recorded and the body and heart weights as well. From the molecular point of view, the Coriolus versicolor administration reduced the activation of the TLR4/NF-κB pathway and the levels of pro-inflammatory cytokines (INF-γ, TNF-α, IL-6, IL-17, and IL-2) and restored the levels of anti-inflammatory cytokines (IL-10). These anti-inflammatory effects were accompanied with a reduced lipid peroxidation and nitrite levels and restored the antioxidant enzyme activities (SOD and CAT) and GSH levels. Additionally, it reduced the histological injury and the immune cell recruitment (CD4+ and CD68+ cells). Moreover, we observed an antiapoptotic activity in both intrinsic (Fas/FasL/caspase-3) and extrinsic (Bax/Bcl-2) pathways. Overall, our data showed that Coriolus versicolor administration modulates the TLR4/NF-κB signaling in EAM.
Collapse
Affiliation(s)
- Livia Interdonato
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Ramona D’Amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Marika Cordaro
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, Consolare Valeria, 98100 Messina, Italy
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Melissa D’Agostino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Tiziana Genovese
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Enrico Gugliandolo
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Rosalia Crupi
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Rosanna Di Paola
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| |
Collapse
|
12
|
Hou L, Liang H, Zeng S, Li J, Chen Z, Liao X, Liu S, Li M, Li B, Yuan Y. Optimising the ECMO treatment regimen increases the survival rate for adult patients with acute fulminant myocarditis: A single-centre retrospective cohort study. Front Med (Lausanne) 2023; 10:1146570. [PMID: 37007790 PMCID: PMC10060512 DOI: 10.3389/fmed.2023.1146570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
Abstract
BackgroundApplying Extracorporeal membrane oxygenation (ECMO) to patients with acute fulminant myocarditis (AFM) reduces their mortality. The survival rate is 55.6-71.9% for adult AFM patients, which is lower than that for paediatric patients (63-81%). In our centre, the survival rate of ECMO for adult patients with AFM was 66.7% from January 2003 to 2012. In January 2013, the therapeutic regimen was optimised, and then the survival rate increased to 89.1% by January 2022. This article analyses the reasons for the improved survival rate following the optimisation of treatment protocols.MethodsThe data for adult patients with AFM who underwent ECMO for a poor response conventional treatment from January 2003 to January 2022 were reviewed. According to different treatment regimens, the AFM patients were divided into an old and a new regimen group. Univariate and multivariate logistic regression analyses were performed on the data before and after ECMO.ResultsFifty-five patients were enrolled in the age (31.2 ± 11.3), including 24 males. Forty-nine patients were weaned successfully from ECMO [duration: (4.1 ± 1.8) d], all of whom were discharged from the hospital, with a survival rate of 89.1%. Compared with the old regimen group, the new regimen group had a shorter duration of shock to ECMO, a lower proportion of patients receiving extracorporeal cardiopulmonary resuscitation (ECPR), a lower Vasoactive Inotropic Score (VIS), and lower levels of lactic acid, and high-sensitivity troponin T before ECMO (p < 0.05). Compared with the old regimen group, after ECMO, the new regimen group had lower ECMO flow, lower proportion of left ventricular dilation and lower limb ischemia injury, the duration of ECMO was shorter, and significantly improved the survival rate, the difference was statistically significant (P < 0.05). The duration of shock to ECMO and VIS before ECMO were independent risk factors for the survival rate (p < 0.05).ConclusionEarly ECMO initiation in adult AFM patients with a poor response to conventional therapy and low-flow ECMO to meet metabolic needs can reduce serious complications affecting the prognosis, may be associated with better outcomes.
Collapse
Affiliation(s)
- Liusheng Hou
- Department of Critical Care Medicine, Zhongshan City People’s Hospital, Zhongshan, Guangdong, China
| | - Hongkai Liang
- Department of Critical Care Medicine, Zhongshan City People’s Hospital, Zhongshan, Guangdong, China
| | - Shiyong Zeng
- Department of Critical Care Medicine, Zhongshan City People’s Hospital, Zhongshan, Guangdong, China
| | - Jianwei Li
- Department of Critical Care Medicine, Zhongshan City People’s Hospital, Zhongshan, Guangdong, China
| | - Zhou Chen
- Department of Anesthesiology, Zhongshan City People’s Hospital, Zhongshan, Guangdong, China
| | - Xiaozu Liao
- Department of Anesthesiology, Zhongshan City People’s Hospital, Zhongshan, Guangdong, China
| | - Shaozhong Liu
- Department of Ultrasound Medicine, Zhongshan City People’s Hospital, Zhongshan, Guangdong, China
| | - Mingxing Li
- Department of Cardiovascular Center, Zhongshan City People’s Hospital, Zhongshan, Guangdong, China
| | - Binfei Li
- Department of Anesthesiology, Zhongshan City People’s Hospital, Zhongshan, Guangdong, China
- *Correspondence: Binfei Li,
| | - Yong Yuan
- Department of Cardiovascular Center, Zhongshan City People’s Hospital, Zhongshan, Guangdong, China
- Yong Yuan,
| |
Collapse
|
13
|
Tsigkou V, Oikonomou E, Anastasiou A, Lampsas S, Zakynthinos GE, Kalogeras K, Katsioupa M, Kapsali M, Kourampi I, Pesiridis T, Marinos G, Vavuranakis MA, Tousoulis D, Vavuranakis M, Siasos G. Molecular Mechanisms and Therapeutic Implications of Endothelial Dysfunction in Patients with Heart Failure. Int J Mol Sci 2023; 24:ijms24054321. [PMID: 36901752 PMCID: PMC10001590 DOI: 10.3390/ijms24054321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/06/2023] [Accepted: 02/15/2023] [Indexed: 02/25/2023] Open
Abstract
Heart failure is a complex medical syndrome that is attributed to a number of risk factors; nevertheless, its clinical presentation is quite similar among the different etiologies. Heart failure displays a rapidly increasing prevalence due to the aging of the population and the success of medical treatment and devices. The pathophysiology of heart failure comprises several mechanisms, such as activation of neurohormonal systems, oxidative stress, dysfunctional calcium handling, impaired energy utilization, mitochondrial dysfunction, and inflammation, which are also implicated in the development of endothelial dysfunction. Heart failure with reduced ejection fraction is usually the result of myocardial loss, which progressively ends in myocardial remodeling. On the other hand, heart failure with preserved ejection fraction is common in patients with comorbidities such as diabetes mellitus, obesity, and hypertension, which trigger the creation of a micro-environment of chronic, ongoing inflammation. Interestingly, endothelial dysfunction of both peripheral vessels and coronary epicardial vessels and microcirculation is a common characteristic of both categories of heart failure and has been associated with worse cardiovascular outcomes. Indeed, exercise training and several heart failure drug categories display favorable effects against endothelial dysfunction apart from their established direct myocardial benefit.
Collapse
Affiliation(s)
- Vasiliki Tsigkou
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
- Correspondence: ; Tel.: +30-69-4770-1299
| | - Artemis Anastasiou
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Stamatios Lampsas
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - George E. Zakynthinos
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Konstantinos Kalogeras
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Maria Katsioupa
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Maria Kapsali
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Islam Kourampi
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Theodoros Pesiridis
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Georgios Marinos
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Michael-Andrew Vavuranakis
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Dimitris Tousoulis
- 1st Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Hippokration General Hospital, 11527 Athens, Greece
| | - Manolis Vavuranakis
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Gerasimos Siasos
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
14
|
Ba H, Zhang D, Guan S, Zheng J. Global burden of myocarditis and cardiomyopathy in children and prediction for 2035 based on the global burden of disease study 2019. Front Cardiovasc Med 2023; 10:1173015. [PMID: 37200977 PMCID: PMC10185772 DOI: 10.3389/fcvm.2023.1173015] [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: 02/24/2023] [Accepted: 04/17/2023] [Indexed: 05/20/2023] Open
Abstract
Background Myocarditis and cardiomyopathy are commonly occurring cardiovascular diseases that seriously threaten children's health. It was urgent to update the global incidence and mortality of childhood myocarditis and cardiomyopathy, and to predict the incidence rate of 2035 by the Global Burden of Disease database. Methods The Global Burden of Disease study data from 1990 to 2019 in 204 countries and territories were used to determine: global incidence and mortality rates of childhood myocarditis and cardiomyopathy from 0 to 19 by five age groups; relationship between sociodemographic index (SDI) and incidence and mortality rates by age group; and, based on an age-period-cohort model, the projected incidence of childhood myocarditis and cardiomyopathy for 2035. Results From 1990 to 2019, global age-standardized incidence rate decreased by 0.1% (95% UI 0.0-0.1) to 7.7% (95% UI 5.1-11.1). Boys had higher age-standardized incidence of childhood myocarditis and cardiomyopathy than girls [9.12, (95% UI 6.05-13.07) vs. 6.18, (95% UI 4.06-8.92)]. Childhood myocarditis and cardiomyopathy affected 121,259 (95% UI 80,467-173,790) boys and 77,216 (95% UI 50,684-111,535) girls in 2019. At the regional level, SDI changes in most areas showed no meaningful difference. In East Asia and high-income Asia Pacific, increased SDI was associated with decreased and increased incidence rate, respectively. In 2019, 11,755 (95% UI 9,611-14,509) children died from myocarditis and cardiomyopathy worldwide. Age-standardized mortality rate decreased significantly by 0.4% (95% UI 0.2-0.6)-0.5% (95% UI 0.4-0.6). Number of deaths from childhood myocarditis and cardiomyopathy in 2019 was highest in the <5-year-old group [7,442 (95% UI 5,834-9,699)]. Myocarditis and cardiomyopathy incidence in 10-14- and 15-19-year-olds is projected to increase by 2035. Conclusion Global data on childhood myocarditis and cardiomyopathy from 1990 to 2019 showed a decreasing trend in incidence and mortality, and an increasing trend in older children, especially in high SDI regions.
Collapse
Affiliation(s)
- Hongjun Ba
- Department of Pediatric Cardiology, Heart Centre, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou, China
| | - Daoqi Zhang
- Department of Internal Medicine Teaching and Research Section, Xuancheng Vocational and Technical College, Xuanchen, China
| | - Shiyang Guan
- Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jinxin Zheng
- Department of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, National Institute of Parasitic Diseases, Shanghai, China
- Correspondence: Jinxin Zheng
| |
Collapse
|
15
|
Shi T, Miller EJ. Novel Radiotracers for Molecular Imaging of Myocardial Inflammation: an Update Focused on Clinical Translation of Non-18F-FDG Radiotracers. CURRENT CARDIOVASCULAR IMAGING REPORTS 2023; 16:1-9. [PMID: 36926261 PMCID: PMC9996562 DOI: 10.1007/s12410-023-09574-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2023] [Indexed: 03/11/2023]
Abstract
Purpose of Review The purpose of this paper is to provide a focused update on recent advances in non-18F-FDG radiotracers for myocardial inflammatory diseases, with a focus on cardiac sarcoidosis and myocarditis. Recent Findings Novel radiotracers targeting molecular features of inflammation have the potential to visualize underlying molecular mechanisms key to the pathogenesis of inflammatory cardiomyopathies such as sarcoidosis and myocarditis. These radiotracers may provide unique opportunities for improved mechanistic insight, higher specificity, and better quantification of disease activity, as well as potential for guidance and monitoring of immunomodulatory therapies. Summary Novel radiotracers provide unique possibilities in diagnosis, prognostic performance, and therapy guidance for cardiac sarcoidosis and myocarditis.
Collapse
Affiliation(s)
- Tiantian Shi
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT USA
| | - Edward J Miller
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT USA
| |
Collapse
|
16
|
Qu H, Feldman AM, Hakonarson H. Genetics of BAG3: A Paradigm for Developing Precision Therapies for Dilated Cardiomyopathies. J Am Heart Assoc 2022; 11:e027373. [PMID: 36382946 PMCID: PMC9851466 DOI: 10.1161/jaha.122.027373] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 09/20/2022] [Indexed: 11/18/2022]
Abstract
Nonischemic dilated cardiomyopathy is a common form of heart muscle disease in which genetic factors play a critical etiological role. In this regard, both rare disease-causing mutations and common disease-susceptible variants, in the Bcl-2-associated athanogene 3 (BAG3) gene have been reported, highlighting the critical role of BAG3 in cardiomyocytes and in the development of dilated cardiomyopathy. The phenotypic effects of the BAG3 mutations help investigators understand the structure and function of the BAG3 gene. Indeed, we report herein that all of the known pathogenic/likely pathogenic variants affect at least 1 of 3 protein functional domains, ie, the WW domain, the second IPV (Ile-Pro-Val) domain, or the BAG domain, whereas none of the missense nontruncating pathogenic/likely pathogenic variants affect the proline-rich repeat (PXXP) domain. A common variant, p.Cys151Arg, associated with reduced susceptibility to dilated cardiomyopathy demonstrated a significant difference in allele frequencies among diverse human populations, suggesting evolutionary selective pressure. As BAG3-related therapies for heart failure move from the laboratory to the clinic, the ability to provide precision medicine will depend in large part on having a thorough understanding of the potential effects of both common and uncommon genetic variants on these target proteins. The current review article provides a roadmap that investigators can utilize to determine the potential interactions between a patient's genotype, their phenotype, and their response to therapeutic interventions with both gene delivery and small molecules.
Collapse
Affiliation(s)
- Hui‐Qi Qu
- The Center for Applied Genomics, Children’s Hospital of PhiladelphiaPhiladelphiaPA
| | - Arthur M. Feldman
- Department of Medicine, Division of CardiologyThe Lewis Katz School of Medicine at Temple UniversityPhiladelphiaPA
- The Center for Neurovirology and Gene EditingThe Lewis Katz School of Medicine at Temple UniversityPhiladelphiaPA
| | - Hakon Hakonarson
- The Center for Applied Genomics, Children’s Hospital of PhiladelphiaPhiladelphiaPA
- Department of Pediatrics, The Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA
- Division of Human GeneticsChildren’s Hospital of PhiladelphiaPhiladelphiaPA
- Division of Pulmonary MedicineChildren’s Hospital of PhiladelphiaPhiladelphiaPA
- Faculty of MedicineUniversity of IcelandReykjavikIceland
| |
Collapse
|
17
|
Guo J, Zhao N, Jin P, Yin Y. Effect of Astragalus injection on inflammatory mediators in patients with viral myocarditis: A systematic review and meta-analysis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 107:154436. [PMID: 36115170 DOI: 10.1016/j.phymed.2022.154436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 08/27/2022] [Accepted: 09/04/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND To explore the therapeutic effect and mechanism of Astragalus injection on viral myocarditis, we conducted a systematic review and meta-analysis to identify the influence of Astragalus injection on inflammatory mediators and overall efficiency in patients undergoing viral myocarditis. METHODS EMBASE, Cochrane Library, PubMed, Chinese Biomedical Literature, Chinese National Knowledge Infrastructure (CNKI), and Wanfang databases were searched to screen randomized controlled trials (RCTs) published before July 3, 2022. The quality of participating studies was assessed by the Cochrane Collaboration Risk of Bias tool. The calculation of qualitative data used a risk ratio (RR) with a 95% confidence interval (95% CI), and quantitative data had standardized mean differences (SMDs) with a 95% CI. The heterogeneity among trials was quantified with Cochran's Q test and the I2 statistic. Confounding factors were estimated by sensitivity analysis, meta-regression, and subgroup analysis. The publication bias of participating articles was evaluated by funnel plot and Egger's test. The Grading of Recommendations Assessment, Development and Evaluation (GRADE) system was recommended for assessing the strength of evidence. RESULTS Nineteen available studies were included in our present meta-analysis, all of which were conducted in China. The outcomes expose that Astragalus injection dramatically decreased the levels of pro-inflammatory TNF-α (SMD=-2.271, 95% CI=-2.802 to -1.739, p<0.001, I2=90.6%), IL-6 (SMD=-1.501, 95% CI=-1.872 to -1.130, p<0.001, I2=83.2%), IL-17 (SMD=-3.194, 95% CI=-4.569 to -1.818, p<0.001, I2=88.9%), 1L-8 (SMD=-6.133, 95% CI=-9.938 to -2.328, p = 0.002, I2=97%), 1L-1 (SMD=-1.814, 95% CI=-2.557 to -1.070, p<0.001, I2=92.1%), CRP (SMD=-2.020, 95% CI=-3.107 to -0.932, p<0.001, I2=92.7%), and IFN-γ (SMD=-1.512, 95% CI=-2.771 to -0.253, p = 0.019, I2=92%) and increased the total effective rate of treatment (RR=1.225, 95% CI=1.17 to 1.29, p<0.001, I2=0.0%) in patients with viral myocarditis. CONCLUSION Astragalus injection can play a therapeutic role in patients with viral myocarditis through immunomodulatory effects. Outcomes were treated with caution due to significant heterogeneity among studies. Large-scale RCTs should be performed to support these conclusions.
Collapse
Affiliation(s)
- Jing Guo
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050090, Hebei, China
| | - Nannan Zhao
- Qinhuangdao Traditional Chinese Medicine Hospital, Qinhuangdao 066003, Hebei, China
| | - Peipei Jin
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050090, Hebei, China
| | - Yujie Yin
- Key Laboratory of State Administration of Traditional Chinese Medicine (Cardio-Cerebral Vessel Collateral Disease), Shijiazhuang 050035 Hebei, China; State Key Laboratory of Collateral Disease Research and Innovation Medicine, Shijiazhuang 050035, China
| |
Collapse
|
18
|
Rajak P, Roy S, Podder S, Dutta M, Sarkar S, Ganguly A, Mandi M, Dutta A, Nanda S, Khatun S. Synergistic action of organophosphates and COVID-19 on inflammation, oxidative stress, and renin-angiotensin system can amplify the risk of cardiovascular maladies. Toxicol Appl Pharmacol 2022; 456:116267. [PMID: 36240863 PMCID: PMC9554205 DOI: 10.1016/j.taap.2022.116267] [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: 08/09/2022] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 11/24/2022]
Abstract
Organophosphates (OPs) are ubiquitous environmental contaminants, widely used as pesticides in agricultural fields. In addition, they serve as flame-retardants, plasticizers, antifoaming or antiwear agents in lacquers, hydraulic fluids, and floor polishing agents. Therefore, world-wide and massive application of these compounds have increased the risk of unintentional exposure to non-targets including the human beings. OPs are neurotoxic agents as they inhibit the activity of acetylcholinesterase at synaptic cleft. Moreover, they can fuel cardiovascular issues in the form of myocardities, cardiac oedema, arrhythmia, systolic malfunction, infarction, and altered electrophysiology. Such pathological outcomes might increase the severity of cardiovascular diseases which are the leading cause of mortality in the developing world. Coronavirus disease-19 (COVID-19) is the ongoing global health emergency caused by SARS-CoV-2 infection. Similar to OPs, SARS-CoV-2 disrupts cytokine homeostasis, redox-balance, and angiotensin-II/AT1R axis to promote cardiovascular injuries. Therefore, during the current pandemic milieu, unintentional exposure to OPs through several environmental sources could escalate cardiac maladies in patients with COVID-19.
Collapse
Affiliation(s)
- Prem Rajak
- Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, India,Corresponding author
| | - Sumedha Roy
- Cytogenetics Laboratory, Department of Zoology, The University of Burdwan, West Bengal, India
| | | | - Moumita Dutta
- Departments of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Saurabh Sarkar
- Department of Zoology, Gushkara Mahavidyalaya; Gushkara, Purba Bardhaman, West Bengal, India
| | - Abhratanu Ganguly
- Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, India
| | - Moutushi Mandi
- Toxicology Research Unit, Department of Zoology, The University of Burdwan, Purba Bardhaman, West Bengal, India
| | - Anik Dutta
- Post Graduate Department of Zoology, Darjeeling Govt. College, West Bengal, India
| | - Sayantani Nanda
- Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, India
| | - Salma Khatun
- Department of Zoology, Krishna Chandra College, Hetampur, West Bengal, India
| |
Collapse
|
19
|
Affiliation(s)
- Cristina Basso
- From the Cardiovascular Pathology Unit, Azienda Ospedaliera, Department of Cardiac, Thoracic, and Vascular Sciences and Public Health, University of Padua, Padua, Italy
| |
Collapse
|
20
|
Liu T, Li Y, Chen S, Wang L, Liu X, Yang Q, Wang Y, Qiao X, Tong J, Deng X, Shao S, Wang H, Shen H. CircDDX17 enhances coxsackievirus B3 replication through regulating miR-1248/NOTCH receptor 2 axis. Front Microbiol 2022; 13:1012124. [PMID: 36338034 PMCID: PMC9627658 DOI: 10.3389/fmicb.2022.1012124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/20/2022] [Indexed: 11/21/2022] Open
Abstract
Coxsackievirus B3 (CVB3) was one of the most common pathogens to cause viral myocarditis. Circular RNAs as novel non-coding RNAs with a closed loop molecular structure have been confirmed to be involved in virus infectious diseases, but the function in CVB3 infection was not systematically studied. In this study, we identified that hsa_circ_0063331 (circDDX17) was drastically decreased after CVB3 infection by circRNA microarray. In vivo and in vitro, when cells or mice were infected with CVB3, the expression of circDDX17 was significantly reduced, as demonstrated by quantitative real-time PCR assays. Additionally, circDDX17 enhanced CVB3 replication by downregulating the expression of miR-1248 in HeLa and HL-1 cells, and miR-1248 regulated CVB3 replication through interacting with the gene coding for NOTCH Receptor 2 (NOTCH2), and NOTCH2 could upregulate methyltransferase-like protein 3 (METTL3). Taken together, this study suggested that circDDX17 promoted CVB3 replication and regulated NOTCH2 by targeting miR-1248 as a miRNAs sponge.
Collapse
Affiliation(s)
- Tingjun Liu
- Cardiothoracic Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yuhan Li
- Cardiothoracic Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Shengjie Chen
- Cardiothoracic Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lulu Wang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xiaolan Liu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Qingru Yang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yan Wang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xiaorong Qiao
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jing Tong
- Xuzhou Center for Disease Control and Prevention, Xuzhou, China
| | - Xintao Deng
- People’s Hospital of Xinghua, Jiangsu University Teaching Hospital, Xinghua, China
| | - Shihe Shao
- Cardiothoracic Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Hua Wang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
- Hua Wang,
| | - Hongxing Shen
- Cardiothoracic Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
- *Correspondence: Hongxing Shen,
| |
Collapse
|
21
|
Yang Y, Li W, You B, Zhou C. Advances in cell death mechanisms involved in viral myocarditis. Front Cardiovasc Med 2022; 9:968752. [PMID: 36017100 PMCID: PMC9395613 DOI: 10.3389/fcvm.2022.968752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 07/19/2022] [Indexed: 11/29/2022] Open
Abstract
Viral myocarditis is an acute inflammatory disease of the myocardium. Although many etiopathogenic factors exist, coxsackievirus B3 is a the leading cause of viral myocarditis. Abnormal cardiomyocyte death is the underlying problem for most cardiovascular diseases and fatalities. Various types of cell death occur and are regulated to varying degrees. In this review, we discuss the different cell death mechanisms in viral myocarditis and the potential interactions between them. We also explore the role and mechanism of cardiomyocyte death with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Exploring the mechanisms may help in the early identification and the development of effective treatments, thus improving the quality of life of patients with viral myocarditis. We believe that the inhibition of cardiomyocyte death has immense therapeutic potential in increasing the longevity and health of the heart.
Collapse
Affiliation(s)
- Yang Yang
- Taizhou People’s Hospital Affiliated to Nanjing Medical University, Taizhou, China
- Clinical Laboratory Center, Jiangsu Taizhou People’s Hospital, Taizhou, China
- *Correspondence: Yang Yang,
| | - Wang Li
- Taizhou People’s Hospital Affiliated to Nanjing Medical University, Taizhou, China
- Clinical Laboratory Center, Jiangsu Taizhou People’s Hospital, Taizhou, China
| | - Benshuai You
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Chenglin Zhou
- Taizhou People’s Hospital Affiliated to Nanjing Medical University, Taizhou, China
- Clinical Laboratory Center, Jiangsu Taizhou People’s Hospital, Taizhou, China
- Chenglin Zhou,
| |
Collapse
|
22
|
Montera MW, Marcondes-Braga FG, Simões MV, Moura LAZ, Fernandes F, Mangine S, Oliveira Júnior ACD, Souza ALADAGD, Ianni BM, Rochitte CE, Mesquita CT, de Azevedo Filho CF, Freitas DCDA, Melo DTPD, Bocchi EA, Horowitz ESK, Mesquita ET, Oliveira GH, Villacorta H, Rossi Neto JM, Barbosa JMB, Figueiredo Neto JAD, Luiz LF, Hajjar LA, Beck-da-Silva L, Campos LADA, Danzmann LC, Bittencourt MI, Garcia MI, Avila MS, Clausell NO, Oliveira NAD, Silvestre OM, Souza OFD, Mourilhe-Rocha R, Kalil Filho R, Al-Kindi SG, Rassi S, Alves SMM, Ferreira SMA, Rizk SI, Mattos TAC, Barzilai V, Martins WDA, Schultheiss HP. Brazilian Society of Cardiology Guideline on Myocarditis - 2022. Arq Bras Cardiol 2022; 119:143-211. [PMID: 35830116 PMCID: PMC9352123 DOI: 10.36660/abc.20220412] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
| | - Fabiana G Marcondes-Braga
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | - Marcus Vinícius Simões
- Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo, São Paulo, SP - Brasil
| | | | - Fabio Fernandes
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | - Sandrigo Mangine
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | | | | | - Bárbara Maria Ianni
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | - Carlos Eduardo Rochitte
- Instituto do Coração (InCor) - Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil
- Hospital do Coração (HCOR), São Paulo, SP - Brasil
| | - Claudio Tinoco Mesquita
- Hospital Pró-Cardíaco, Rio de Janeiro, RJ - Brasil
- Universidade Federal Fluminense,Rio de Janeiro, RJ - Brasil
- Hospital Vitória, Rio de Janeiro, RJ - Brasil
| | | | | | | | - Edimar Alcides Bocchi
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | | | - Evandro Tinoco Mesquita
- Universidade Federal Fluminense,Rio de Janeiro, RJ - Brasil
- Centro de Ensino e Treinamento Edson de Godoy Bueno / UHG, Rio de Janeiro, RJ - Brasil
| | | | | | | | | | | | | | - Ludhmila Abrahão Hajjar
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
- Instituto do Câncer do Estado de São Paulo da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil
| | - Luis Beck-da-Silva
- Hospital de Clínicas de Porto Alegre, Porto Alegre, RS - Brasil
- Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS - Brasil
| | | | | | - Marcelo Imbroise Bittencourt
- Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ - Brasil
- Hospital Universitário Pedro Ernesto, Rio de Janeiro, RJ - Brasil
| | - Marcelo Iorio Garcia
- Hospital Universitário Clementino Fraga Filho (HUCFF) da Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ - Brasil
| | - Monica Samuel Avila
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | | | | | | | | | | | | | - Sadeer G Al-Kindi
- Harrington Heart and Vascular Institute, University Hospitals and Case Western Reserve University,Cleveland, Ohio - EUA
| | | | - Silvia Marinho Martins Alves
- Pronto Socorro Cardiológico de Pernambuco (PROCAPE), Recife, PE - Brasil
- Universidade de Pernambuco (UPE), Recife, PE - Brasil
| | - Silvia Moreira Ayub Ferreira
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | - Stéphanie Itala Rizk
- Instituto do Câncer do Estado de São Paulo da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil
- Hospital Sírio Libanês, São Paulo, SP - Brasil
| | | | - Vitor Barzilai
- Instituto de Cardiologia do Distrito Federal, Brasília, DF - Brasil
| | - Wolney de Andrade Martins
- Universidade Federal Fluminense,Rio de Janeiro, RJ - Brasil
- DASA Complexo Hospitalar de Niterói, Niterói, RJ - Brasil
| | | |
Collapse
|
23
|
Ohta-Ogo K, Sugano Y, Ogata S, Nakayama T, Komori T, Eguchi K, Dohi K, Yokokawa T, Kanamori H, Nishimura S, Nakamura K, Ikeda Y, Nishimura K, Takemura G, Anzai T, Hiroe M, Hatakeyama K, Ishibashi-Ueda H, Imanaka-Yoshida K. Myocardial T-Lymphocytes as a Prognostic Risk-Stratifying Marker of Dilated Cardiomyopathy - Results of the Multicenter Registry to Investigate Inflammatory Cell Infiltration in Dilated Cardiomyopathy in Tissues of Endomyocardial Biopsy (INDICATE Study). Circ J 2022; 86:1092-1101. [PMID: 35264513 DOI: 10.1253/circj.cj-21-0529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) associated with inflammation is diagnosed by endomyocardial biopsy; patients with this have a poorer prognosis than patients without inflammation. To date, standard diagnostic criteria have not been established. METHODS AND RESULTS This study analyzed clinical records and endomyocardial biopsy samples of 261 patients with DCM (201 males, median left ventricular ejection fraction; 28%) from 8 institutions in a multicenter retrospective study. Based on the European Society of Cardiology criteria and CD3 (T-lymphocytes) and CD68 (macrophages) immunohistochemistry, 48% of patients were categorized as having inflammatory DCM. For risk-stratification, we divided patients into 3 groups using Akaike Information Criterion/log-rank tests, which can determine multiple cut-off points: CD3+-Low, <13/mm2(n=178, 68%); CD3+-Moderate, 13-24/mm2(n=58, 22%); and CD3+-High, ≥24/mm2(n=25, 10%). The survival curves for cardiac death or left ventricular assist device implantation differed significantly among the 3 groups (10-year survival rates: CD3+-Low: 83.4%; CD3+-Moderate: 68.4%; CD3+-High: 21.1%; Log-rank P<0.001). Multivariate Cox analysis revealed CD3+count as a potent independent predictive factor for survival (fully adjusted hazard ratio: CD3+-High: 5.70, P<0.001; CD3+-Moderate: 2.64, P<0.01). CD3+-High was also associated with poor left ventricular functional and morphological recovery at short-term follow up. CONCLUSIONS Myocardial CD3+T-lymphocyte infiltration has a significant prognostic impact in DCM and a 3-tiered risk-stratification model could be helpful to refine patient categorization.
Collapse
Affiliation(s)
- Keiko Ohta-Ogo
- Department of Pathology, National Cerebral and Cardiovascular Center
| | | | - Soshiro Ogata
- Department of Preventive Medicine and Epidemiology, National Cerebral and Cardiovascular Center
| | - Takafumi Nakayama
- Department of Cardiology, Nagoya City University Graduate School of Medical Sciences
| | - Takahiro Komori
- Department of Cardiovascular Medicine, Jichi Medical University School of Medicine
| | - Kazuo Eguchi
- Department of General Internal Medicine, Saitama Red Cross Hospital
| | - Kaoru Dohi
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine
| | - Tetsuro Yokokawa
- Department of Cardiovascular Medicine, Fukushima Medical University
| | | | - Shigeyuki Nishimura
- Department of Cardiology, Saitama Medical University International Medical Center
| | - Kazufumi Nakamura
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceuticals
| | - Yoshihiko Ikeda
- Department of Pathology, National Cerebral and Cardiovascular Center
| | - Kunihiro Nishimura
- Department of Preventive Medicine and Epidemiology, National Cerebral and Cardiovascular Center
| | - Genzou Takemura
- Department of Internal Medicine, Asahi University School of Dentistry
| | - Toshihisa Anzai
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine
| | - Michiaki Hiroe
- Department of Cardiology, National Center for Global Health and Medicine
| | - Kinta Hatakeyama
- Department of Pathology, National Cerebral and Cardiovascular Center
| | | | - Kyoko Imanaka-Yoshida
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine
| |
Collapse
|
24
|
Zhu T, Wang M, Quan J, Du Z, Li Q, Xie Y, Lin M, Xu C, Xie Y. Identification and Verification of Feature Biomarkers Associated With Immune Cells in Dilated Cardiomyopathy by Bioinformatics Analysis. Front Genet 2022; 13:874544. [PMID: 35646094 PMCID: PMC9133742 DOI: 10.3389/fgene.2022.874544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 03/30/2022] [Indexed: 11/18/2022] Open
Abstract
Objective: To explore immune-related feature genes in patients with dilated cardiomyopathy (DCM). Methods: Expression profiles from three datasets (GSE1145, GSE21610 and GSE21819) of human cardiac tissues of DCM and healthy controls were downloaded from the GEO database. After data preprocessing, differentially expressed genes (DEGs) were identified by the ‘limma’ package in R software. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were then performed to identify biological functions of the DEGs. The compositional patterns of stromal and immune cells were estimated using xCell. Hub genes and functional modules were identified based on protein-protein interaction (PPI) network analysis by STRING webtool and Cytoscape application. Correlation analysis was performed between immune cell subtypes and hub genes. Hub genes with |correlation coefficient| > 0.5 and p value <0.05 were selected as feature biomarkers. A logistic regression model was constructed based on the selected biomarkers and validated in datasets GSE5406 and GSE57338. Results: A total of 1,005 DEGs were identified. Functional enrichment analyses indicated that extracellular matrix remodeling and immune and inflammation disorder played important roles in the pathogenesis of DCM. Immune cells, including CD8+ T-cells, macrophages M1 and Th1 cells, were proved to be significantly changed in DCM patients by immune cell infiltration analysis. In the PPI network analysis, STAT3, IL6, CCL2, PIK3R1, ESR1, CCL5, IL17A, TLR2, BUB1B and MYC were identified as hub genes, among which CCL2, CCL5 and TLR2 were further screened as feature biomarkers by using hub genes and immune cells correlation analysis. A diagnosis model was successfully constructed by using the three biomarkers with area under the curve (AUC) scores 0.981, 0.867 and 0.946 in merged dataset, GSE5406 and GSE57338, respectively. Conclusion: The present study identified three immune-related genes as diagnostic biomarkers for DCM, providing a novel perspective of immune and inflammatory response for the exploration of DCM molecular mechanisms.
Collapse
Affiliation(s)
- Tingfang Zhu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingjie Wang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinwei Quan
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zunhui Du
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiheng Li
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Xie
- Johns Hopkins University, Baltimore, MD, United States
| | - Menglu Lin
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cathy Xu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yucai Xie
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yucai Xie,
| |
Collapse
|
25
|
Bablis P, Pollard H, Rosner AL. Stress reduction via neuro-emotional technique to achieve the simultaneous resolution of chronic low back pain with multiple inflammatory and biobehavioural indicators: A randomized, double-blinded, placebo-controlled trial. JOURNAL OF INTEGRATIVE MEDICINE 2022; 20:135-144. [PMID: 34924332 DOI: 10.1016/j.joim.2021.12.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 10/27/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Beginning with the concepts of stress developed by Selye, an approach to stress and pain management, known as neuro-emotional technique (NET), has been developed. It is a treatment approach based on the principle that the stressor effects of dormant and/or current unresolved issues or trauma are what determine one's bodily responses. These responses are relatively personalized to the conditioned, experiential and emotional reality of the individual. OBJECTIVE To determine the effect of NET on patients with chronic low back pain (CLBP) over time. DESIGN, SETTING, PARTICIPANTS, AND INTERVENTIONS In a randomized, double-blinded, placebo-controlled study conducted in a single clinic, NET or control treatments were given twice weekly for 4 weeks in a population of 112 patients. MAIN OUTCOME MEASURES Outcome measures, including Oswestry Disability Index, Quadruple Visual Analogue Scale, the psychoneuroimmunology markers of blood serum levels of C-reactive protein, tumour necrosis factor-α, interleukin-1 (IL-1), IL-6, and IL-10, and 10 dimensions of the Short Form Health Survey scale, were assessed at baseline and at 1, 3 and 6 months following the intervention period. RESULTS Compared to placebo, NET produced clinical and statistical significance (P < 0.001) via declines of virtually all physiological, pain and disability markers, accompanied by gains in quality-of-life indicators at 0 (baseline), 1, 3 and 6 months. Reductions of the percentages of patients whose 5 biomarkers lay outside the normative range were achieved at 1, 3 and 6 months by NET but not control interventions. CONCLUSION A randomized, controlled trial of CLBP patients indicated that 8 NET interventions, compared to placebo, produced clinically and statistically significant reductions in pain, disability and inflammatory biomarkers, and improvements in quality-of-life measures. TRIAL REGISTRATION The trial was registered with the Australian and New Zealand Clinical Trials Registry (No. ACTRN12608000002381).
Collapse
Affiliation(s)
- Peter Bablis
- Universal Health, New South Wales 2028, Australia
| | - Henry Pollard
- School of Medical and Applied Sciences, University of Central Queensland, Brisbane, New South Wales 4000, Australia
| | | |
Collapse
|
26
|
Altered Phenotype of Circulating Dendritic Cells and Regulatory T Cells from Patients with Acute Myocarditis. J Immunol Res 2022; 2022:8873146. [PMID: 35265721 PMCID: PMC8901353 DOI: 10.1155/2022/8873146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/19/2022] [Accepted: 02/05/2022] [Indexed: 11/17/2022] Open
Abstract
Dendritic cells (DCs) and regulatory T cells (Tregs) play an essential role in myocarditis. However, a particular DC phenotype in this disease has not been assessed. Herein, we aim to evaluate myeloid (mDCs) and plasmacytoid DC (pDC) phenotype, as well as Treg levels from myocarditis patients and healthy controls. Using multiparametric flow cytometry, we evaluated the levels of myeloid DCs (mDCs), plasmacytoid DCs (pDCs), and Tregs in peripheral blood from myocarditis patients (n = 16) and healthy volunteers (n = 16) and performed correlation analysis with clinical parameters through Sperman test. DCs from myocarditis patients showed a higher expression of costimulatory molecules while a diminished expression of the inhibitory receptors, ILT2 and ILT4. Even more, Treg cells from myocarditis patients displayed higher levels of FOXP3 compared to controls. Clinically, the increased levels of mDCs and their higher expression of costimulatory molecules correlate with a worse myocardial function, higher levels of acute phase reactants, and higher cardiac enzymes. This study shows an activating phenotype of circulating DCs from myocarditis patients. This proinflammatory status may contribute to the pathogenesis and immune deregulation in acute myocarditis.
Collapse
|
27
|
Tymińska A, Ozierański K, Skwarek A, Kapłon-Cieślicka A, Baritussio A, Grabowski M, Marcolongo R, Caforio ALP. Personalized Management of Myocarditis and Inflammatory Cardiomyopathy in Clinical Practice. J Pers Med 2022; 12:jpm12020183. [PMID: 35207671 PMCID: PMC8874629 DOI: 10.3390/jpm12020183] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/20/2022] [Accepted: 01/24/2022] [Indexed: 02/01/2023] Open
Abstract
Myocarditis is an inflammatory heart disease induced by infectious and non-infectious causes frequently triggering immune-mediated pathologic mechanisms leading to myocardial damage and dysfunction. In approximately half of the patients, acute myocarditis resolves spontaneously while in the remaining cases, it may evolve into serious complications including inflammatory cardiomyopathy, arrhythmias, death, or heart transplantation. Due to the large variability in clinical presentation, unpredictable course of the disease, and lack of established causative treatment, myocarditis represents a challenging diagnosis in modern cardiology. Moreover, an increase in the incidence of myocarditis and inflammatory cardiomyopathy has been observed in recent years. However, there is a growing potential of available non-invasive diagnostic methods (biomarkers, serum anti-heart autoantibodies (AHA), microRNAs, speckle tracking echocardiography, cardiac magnetic resonance T1 and T2 tissue mapping, positron emission tomography), which may refine the diagnostic workup and/or noninvasive follow-up. Personalized management should include the use of endomyocardial biopsy and AHA, which may allow the etiopathogenetic subsets of myocarditis (infectious, non-infectious, and/or immune-mediated) to be distinguished and implementation of disease-specific therapies. In this review, we summarize current knowledge on myocarditis and inflammatory cardiomyopathy, and outline some practical diagnostic, therapeutic, and follow-up algorithms to facilitate comprehensive individualized management of these patients.
Collapse
Affiliation(s)
- Agata Tymińska
- First Department of Cardiology, Medical University of Warsaw, 1a Banacha St., 02-097 Warsaw, Poland; (A.T.); (A.S.); (A.K.-C.); (M.G.)
| | - Krzysztof Ozierański
- First Department of Cardiology, Medical University of Warsaw, 1a Banacha St., 02-097 Warsaw, Poland; (A.T.); (A.S.); (A.K.-C.); (M.G.)
- Correspondence: ; Tel.: +48-22-599-29-58; Fax: +48-22-599-19-57
| | - Aleksandra Skwarek
- First Department of Cardiology, Medical University of Warsaw, 1a Banacha St., 02-097 Warsaw, Poland; (A.T.); (A.S.); (A.K.-C.); (M.G.)
| | - Agnieszka Kapłon-Cieślicka
- First Department of Cardiology, Medical University of Warsaw, 1a Banacha St., 02-097 Warsaw, Poland; (A.T.); (A.S.); (A.K.-C.); (M.G.)
| | - Anna Baritussio
- Division of Cardiology, Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padova, 2-35128 Padova, Italy; (A.B.); (R.M.); (A.L.C.)
| | - Marcin Grabowski
- First Department of Cardiology, Medical University of Warsaw, 1a Banacha St., 02-097 Warsaw, Poland; (A.T.); (A.S.); (A.K.-C.); (M.G.)
| | - Renzo Marcolongo
- Division of Cardiology, Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padova, 2-35128 Padova, Italy; (A.B.); (R.M.); (A.L.C.)
| | - Alida LP Caforio
- Division of Cardiology, Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padova, 2-35128 Padova, Italy; (A.B.); (R.M.); (A.L.C.)
| |
Collapse
|
28
|
Reichman-Warmusz E, Warmusz O, Wojnicz R. The rationale for using low-molecular weight heparin in the therapy of symptomatic COVID-19 patients. Open Med (Wars) 2022; 17:216-220. [PMID: 35178474 PMCID: PMC8812716 DOI: 10.1515/med-2021-0374] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/01/2021] [Accepted: 09/29/2021] [Indexed: 11/30/2022] Open
Abstract
Accumulated evidence suggest that the adverse outcome of severe coronavirus disease 2019 (COVID-19) is closely related to prothrombotic microvascular pathology with a high risk of venous thromboembolism. Furthermore, the first observational studies indicated that adjunct therapy with low-molecular weight heparin (LMWH) was associated with lower mortality in this cohort of patients. However, the timing of starting LMWH and the dose remain controversial in COVID-19 patients. Considering the above, the aim of this study was to reveal the rationale for using LMWH in the therapy of symptomatic COVID-19 patients based on experimental and clinical studies on LMWH in inflammatory settings with special consideration given to randomized trials.
Collapse
Affiliation(s)
- Edyta Reichman-Warmusz
- Department of Histology and Cell Pathology in Zabrze, School of Medicine with the Division of Dentistry, Medical University of Silesia in Katowice , Jordana 19 , 41-808 Zabrze , Poland
| | - Oliwia Warmusz
- Department of Histology and Cell Pathology in Zabrze, School of Medicine with the Division of Dentistry, Medical University of Silesia in Katowice , Zabrze , Poland
| | - Romuald Wojnicz
- Department of Histology and Cell Pathology in Zabrze, School of Medicine with the Division of Dentistry, Medical University of Silesia in Katowice , Jordana 19 , 41-808 Zabrze , Poland
- Silesian Centre for Heart Disease in Zabrze , Zabrze , Poland
| |
Collapse
|
29
|
Molecular Mechanisms behind Persistent Presence of Parvovirus B19 in Human Dilated Myocardium. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1376:181-202. [DOI: 10.1007/5584_2021_702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
30
|
Mitochondrial calpain-1 activates NLRP3 inflammasome by cleaving ATP5A1 and inducing mitochondrial ROS in CVB3-induced myocarditis. Basic Res Cardiol 2022; 117:40. [PMID: 35997820 PMCID: PMC9399059 DOI: 10.1007/s00395-022-00948-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/04/2022] [Accepted: 08/04/2022] [Indexed: 01/31/2023]
Abstract
Treatment options for myocarditis are currently limited. Inhibition of calpains has been shown to prevent Coxsackievirus B3 (CVB3)-induced cardiac injuries, but the underlying mechanism of action of calpains has not been elucidated. We investigated whether NOD-, LRR-, and pyrin domain-containing 3 (NLRP3) inflammasome participated in CVB3-induced myocarditis, and investigated the effects of calpain-1 on CVB3-induced cardiac injury. NLRP3 inflammasome was activated in CVB3-infected hearts, evidenced by elevated protein levels of NLRP3, N-terminal domain of Gasdermin D, and cleaved caspase-1, and the increased co-localization of NLRP3 and apoptosis-associated speck-like protein. The intraperitoneal administration of MCC950, a selective inhibitor of the NLRP3 inflammasome, led to decreased levels of serum creatine kinase-MB, cardiac troponin I, lactate dehydrogenase, interleukin-18, interleukin-1β, prevention of the infiltration of inflammatory cells, and improvement of cardiac function under CVB3 infection. Transgenic mice overexpressing the endogenous calpain inhibitor calpastatin (Tg-CAST mice) exhibited not only decreased apoptosis, inflammation, fibrosis, and enhanced cardiac function but also inhibition of NLRP3 inflammasome and pyroptosis. The selective inhibition of calpain-1 using PD151746 protected cardiomyocytes in vitro from CVB3 infection by downregulating NLRP3 inflammasome and, thus, preserved cell viability. Mechanistically, we showed that mitochondrial dysfunction preceded inflammatory response after CVB3 treatment and elimination of mitochondrial reactive oxygen species (ROS) using mitochondria-targeted antioxidants (mito-TEMPO) recapitalized the phenotype observed in Tg-CAST mice. Furthermore, the promotion or inhibition of calpain-1 activation in vitro regulated the mitochondrial respiration chain. Mito-TEMPO reversed calpain-1-mediated NLRP3 inflammation activation and cell death. We also found that mitochondrial calpain-1, which was increased after CVB3 stimulation, activated the NLRP3 inflammasome and resulted in cell death. Furthermore, ATP synthase-α (ATP5A1) was revealed to be the cleaving target of calpain-1 after CVB3 treatment. Downregulating ATP5A1 using ATP5A1-small interfering RNA impaired mitochondrial function, decreased cell viability, and induced NLRP3 inflammasome activation. In conclusion, CVB3 infection induced calpain-1 accumulation in mitochondria, and led to subsequent ATP5A1 cleavage, mitochondrial ROS overproduction, and impaired mitochondrial function, eventually causing NLRP3 inflammasome activation and inducing pyroptosis. Therefore, our findings established the role of calpain in viral myocarditis and unveiled its underlying mechanism of its action. Calpain appears as a promising target for the treatment of viral myocarditis.
Collapse
|
31
|
Schultheiss HP, Baumeier C, Aleshcheva G, Bock CT, Escher F. Viral Myocarditis-From Pathophysiology to Treatment. J Clin Med 2021; 10:jcm10225240. [PMID: 34830522 PMCID: PMC8623269 DOI: 10.3390/jcm10225240] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/06/2021] [Accepted: 11/08/2021] [Indexed: 12/17/2022] Open
Abstract
The diagnosis of acute and chronic myocarditis remains a challenge for clinicians. Characterization of this disease has been hampered by its diverse etiologies and heterogeneous clinical presentations. Most cases of myocarditis are caused by infectious agents. Despite successful research in the last few years, the pathophysiology of viral myocarditis and its sequelae leading to severe heart failure with a poor prognosis is not fully understood and represents a significant public health issue globally. Most likely, at a certain point, besides viral persistence, several etiological types merge into a common pathogenic autoimmune process leading to chronic inflammation and tissue remodeling, ultimately resulting in the clinical phenotype of dilated cardiomyopathy. Understanding the underlying molecular mechanisms is necessary to assess the prognosis of patients and is fundamental to appropriate specific and personalized therapeutic strategies. To reach this clinical prerequisite, there is the need for advanced diagnostic tools, including an endomyocardial biopsy and guidelines to optimize the management of this disease. The severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) has currently led to the worst pandemic in a century and has awakened a special sensitivity throughout the world to viral infections. This work aims to summarize the pathophysiology of viral myocarditis, advanced diagnostic methods and the current state of treatment options.
Collapse
Affiliation(s)
| | - Christian Baumeier
- Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, 12203 Berlin, Germany
| | - Ganna Aleshcheva
- Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, 12203 Berlin, Germany
| | - C-Thomas Bock
- Division of Viral Gastroenteritis and Hepatitis Pathogens and Enteroviruses, Department of Infectious Diseases, Robert Koch Institute, 13353 Berlin, Germany
- Institute of Tropical Medicine, University of Tuebingen, 72074 Tuebingen, Germany
| | - Felicitas Escher
- Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, 12203 Berlin, Germany
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universitaet Berlin and Humboldt-Universitaet zu Berlin, 13353 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| |
Collapse
|
32
|
An L, Wuri J, Zheng Z, Li W, Yan T. Microbiota modulate Doxorubicin induced cardiotoxicity. Eur J Pharm Sci 2021; 166:105977. [PMID: 34416387 DOI: 10.1016/j.ejps.2021.105977] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/31/2021] [Accepted: 08/15/2021] [Indexed: 01/14/2023]
Abstract
Chemotherapy has several adverse effects to patients, some of which are life-threatening. We hypothesized that Doxorubicin induced microbiome imbalance and intestinal damage may contribute to Doxorubicin induced cardiac dysfunction. Male adult (2-3 months) C57BL/6 mice were administered 3 mg/kg, 5 mg/kg, 7.5 mg/kg,15 mg/kg, 20 mg/kg doses of Doxorubicin. Echocardiography was performed at 7 and 14 days after Doxorubicin administration. 16S rRNA amplicon sequencing was used to characterize microbiome changes. Fecal microbiota transplantation (FMT) was performed to evaluate the role of the microbiota on Doxorubicin induced cardiac dysfunction. Doxorubicin dose dependently increases mortality rate and induces cardiac dysfunction. 5 mg/kg-Doxorubicin significantly induces decreased left ventricular ejection fraction (LVEF) and fraction shortening (FS) as well as increased cardiac fibrosis, inflammation and oxidative stress respond without increasing mortality. 5 mg/kg-Doxorubicin induces significant decreased colorectum length, increased loss of goblet cells, numbers of ulcers and infiltration of lymphocyte clusters and decreased tight junction protein ZO-1, as well as increased plasma endotoxin level measured by ELISA assay. 16S rRNA microbiota analysis shows that Doxorubicin-induced microbiota dysbiosis with decreased community richness compared with normal control mice. FMT to Doxorubicin-5 mg treated mice significantly improved cardiac function by increasing LVEF and FS as well as decreased perivascular and interstitial fibrosis; increased colorectum length, decreased the loss of goblet cells,infiltration of lymphocyte clusters,the number of ulcers and plasma endotoxin level; improved microbiota composition, function and diversity with increased abundance of Alloprevotella, Prevotellaceae_UCG-001 and Rikenellaceae_RC9_gut_group. We find that normal fecal transplantation improves cardiac function, decreases gut damage and alter microbiota composition induced by Doxorubicin. The microbiota appears to contribute to heart-gut interaction.
Collapse
Affiliation(s)
- Lulu An
- Neurology, Tianjin Medical University General Hospital Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin-300052, China.
| | - Jimusi Wuri
- Neurology, Tianjin Medical University General Hospital Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin-300052, China.
| | - Zhitong Zheng
- Neurology, Tianjin Medical University General Hospital Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin-300052, China.
| | - Wenqui Li
- Neurology, Tianjin Medical University General Hospital Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin-300052, China.
| | - Tao Yan
- Neurology, Tianjin Medical University General Hospital Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin-300052, China.
| |
Collapse
|
33
|
Hirono K, Takarada S, Okabe M, Miyao N, Nakaoka H, Ibuki K, Ozawa S, Origasa H, Ichida F, Imanaka-Yoshida K. Clinical significance of chronic myocarditis: systematic review and meta-analysis. Heart Vessels 2021; 37:300-314. [PMID: 34365565 DOI: 10.1007/s00380-021-01914-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/28/2021] [Indexed: 11/30/2022]
Abstract
Chronic myocarditis is a prolonged inflammatory condition in the myocardium and its histological manifestation is defined by the presence of an inflammatory infiltrate. Chronic myocarditis has not been well known and its treatment of chronic myocarditis has not been established. Primary outcome of this study was to assess the efficacy of immunomodulatory treatment in addition to conventional treatment, and secondary outcomes were to clarity the prognosis of natural history of chronic myocarditis and incidence of chronic myocarditis in patients with dilated cardiomyopathy (DCM). We searched for studies in Medline, Embase, Cochrane Central Register of Controlled Trials, and Igaku Chuo Zasshi published between January 1946 and June 2020. Sixteen studies met the inclusion criteria. A meta-analysis revealed that patients receiving immunomodulatory treatment showed an improvement in left ventricular ejection fraction after immunomodulatory treatment compared to the control group (hazard ratio, 16.65; confidence interval, 4.55-28.74; p = 0.007). Five-year survival rate of the patients with inflammatory DCM (iDCM) and DCM was 52.7-70.3% and 51.9-91.1%, respectively. Moreover, 51.5%-62.7% of patients with DCM met the criteria of iDCM. Our systematic review revealed that patients with chronic myocarditis had poor prognosis and immunomodulatory treatment was significantly effective in addition to conventional treatment.
Collapse
Affiliation(s)
- Keiichi Hirono
- Department of Pediatrics, Graduate School of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama, 930-0194, Japan.
| | - Shinya Takarada
- Department of Pediatrics, Graduate School of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama, 930-0194, Japan
| | - Mako Okabe
- Department of Pediatrics, Graduate School of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama, 930-0194, Japan
| | - Nariaki Miyao
- Department of Pediatrics, Graduate School of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama, 930-0194, Japan
| | - Hideyuki Nakaoka
- Department of Pediatrics, Graduate School of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama, 930-0194, Japan
| | - Keijiro Ibuki
- Department of Pediatrics, Graduate School of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama, 930-0194, Japan
| | - Sayaka Ozawa
- Department of Pediatrics, Graduate School of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama, 930-0194, Japan
| | - Hideki Origasa
- Biostatistics and Clinical Epidemiology, Graduate School of Medicine, University of Toyama, Toyama, Japan
| | - Fukiko Ichida
- Department of Pediatrics, International University of Health&Welfare, Tokyo, Japan
| | - Kyoko Imanaka-Yoshida
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine, Mie, Japan
| |
Collapse
|
34
|
Diagnosis and Management of Myocarditis: An Evidence-Based Review for the Emergency Medicine Clinician. J Emerg Med 2021; 61:222-233. [PMID: 34108120 DOI: 10.1016/j.jemermed.2021.03.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/17/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Myocarditis is a potentially fatal condition that can be misdiagnosed in the emergency department (ED) setting. OBJECTIVE The purpose of this narrative review article is to provide a summary of the background, pathophysiology, diagnosis, and management of myocarditis, with a focus on emergency clinicians. DISCUSSION Myocarditis occurs when inflammation of the heart musculature causes cardiac dysfunction. Symptoms may range from mild to severe and are often preceded by a viral prodrome. Laboratory assessment and an electrocardiogram can be helpful for the diagnosis, but echocardiography is the ideal test in the ED setting. Some patients may also require advanced imaging, though this will often occur during hospitalization or follow-up. Treatment is primarily focused on respiratory and hemodynamic support. Initial hemodynamic management includes vasopressors and inotropes, whereas more severe cases may require an intra-aortic balloon pump, extracorporeal membrane oxygenation, or a ventricular assist device. Nonsteroidal anti-inflammatory drugs should be avoided while intravenous immunoglobulin is controversial. CONCLUSION Myocarditis is a serious condition with the potential for significant morbidity and mortality. It is important for clinicians to be aware of the current evidence regarding the diagnosis, management, and disposition of these patients.
Collapse
|
35
|
Imanaka-Yoshida K. Tenascin-C in Heart Diseases-The Role of Inflammation. Int J Mol Sci 2021; 22:ijms22115828. [PMID: 34072423 PMCID: PMC8198581 DOI: 10.3390/ijms22115828] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/25/2021] [Accepted: 05/27/2021] [Indexed: 12/20/2022] Open
Abstract
Tenascin-C (TNC) is a large extracellular matrix (ECM) glycoprotein and an original member of the matricellular protein family. TNC is transiently expressed in the heart during embryonic development, but is rarely detected in normal adults; however, its expression is strongly up-regulated with inflammation. Although neither TNC-knockout nor -overexpressing mice show a distinct phenotype, disease models using genetically engineered mice combined with in vitro experiments have revealed multiple significant roles for TNC in responses to injury and myocardial repair, particularly in the regulation of inflammation. In most cases, TNC appears to deteriorate adverse ventricular remodeling by aggravating inflammation/fibrosis. Furthermore, accumulating clinical evidence has shown that high TNC levels predict adverse ventricular remodeling and a poor prognosis in patients with various heart diseases. Since the importance of inflammation has attracted attention in the pathophysiology of heart diseases, this review will focus on the roles of TNC in various types of inflammatory reactions, such as myocardial infarction, hypertensive fibrosis, myocarditis caused by viral infection or autoimmunity, and dilated cardiomyopathy. The utility of TNC as a biomarker for the stratification of myocardial disease conditions and the selection of appropriate therapies will also be discussed from a clinical viewpoint.
Collapse
Affiliation(s)
- Kyoko Imanaka-Yoshida
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan;
- Mie University Research Center for Matrix Biology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| |
Collapse
|
36
|
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
|
37
|
Müller I, Janson L, Sauter M, Pappritz K, Linthout SV, Tschöpe C, Klingel K. Myeloid-Derived Suppressor Cells Restrain Natural Killer Cell Activity in Acute Coxsackievirus B3-Induced Myocarditis. Viruses 2021; 13:v13050889. [PMID: 34065891 PMCID: PMC8151145 DOI: 10.3390/v13050889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/03/2021] [Accepted: 05/10/2021] [Indexed: 12/13/2022] Open
Abstract
Murine models of coxsackievirus B3 (CVB3)-induced myocarditis well represent the different outcomes of this inflammatory heart disease. Previously, we found that CVB3-infected A.BY/SnJ mice, susceptible for severe acute and chronic myocarditis, have lower natural killer (NK) cell levels than C57BL/6 mice, with mild acute myocarditis. There is evidence that myeloid-derived suppressor cells (MDSC) may inhibit NK cells, influencing the course of myocarditis. To investigate the MDSC/NK interrelationship in acute myocarditis, we used CVB3-infected A.BY/SnJ mice. Compared to non-infected mice, we found increased cell numbers of MDSC in the spleen and heart of CVB3-infected A.BY/SnJ mice. In parallel, S100A8 and S100A9 were increased in the heart, spleen, and especially in splenic MDSC cells compared to non-infected mice. In vitro experiments provided evidence that MDSC disrupt cytotoxic NK cell function upon co-culturing with MDSC. MDSC-specific depletion by an anti-Ly6G antibody led to a significant reduction in the virus load and injury in hearts of infected animals. The decreased cardiac damage in MDSC-depleted mice was associated with fewer Mac3+ macrophages and CD3+ T lymphocytes and a reduced cardiac expression of S100A8, S100A9, IL-1β, IL-6, and TNF-α. In conclusion, impairment of functional NK cells by MDSC promotes the development of chronic CVB3 myocarditis in A.BY/SnJ mice.
Collapse
Affiliation(s)
- Irene Müller
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 10017 Berlin, Germany; (I.M.); (K.P.); (S.V.L.); (C.T.)
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, 10017 Berlin, Germany
| | - Lisa Janson
- Cardiopathology, Institute for Pathology and Neuropathology, University Hospital Tübingen, 72076 Tübingen, Germany; (L.J.); (M.S.)
| | - Martina Sauter
- Cardiopathology, Institute for Pathology and Neuropathology, University Hospital Tübingen, 72076 Tübingen, Germany; (L.J.); (M.S.)
| | - Kathleen Pappritz
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 10017 Berlin, Germany; (I.M.); (K.P.); (S.V.L.); (C.T.)
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, 10017 Berlin, Germany
| | - Sophie Van Linthout
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 10017 Berlin, Germany; (I.M.); (K.P.); (S.V.L.); (C.T.)
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, 10017 Berlin, Germany
| | - Carsten Tschöpe
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 10017 Berlin, Germany; (I.M.); (K.P.); (S.V.L.); (C.T.)
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, 10017 Berlin, Germany
- Department of Cardiology, Campus Virchow Clinic, Charité-Universitätsmedizin Berlin, 10017 Berlin, Germany
| | - Karin Klingel
- Cardiopathology, Institute for Pathology and Neuropathology, University Hospital Tübingen, 72076 Tübingen, Germany; (L.J.); (M.S.)
- Correspondence: ; Tel.: +49-7071-2980205
| |
Collapse
|
38
|
Raukar NP, Cooper LT. Implications of SARS-CoV-2-Associated Myocarditis in the Medical Evaluation of Athletes. Sports Health 2021; 13:145-148. [PMID: 33201768 PMCID: PMC8167355 DOI: 10.1177/1941738120974747] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
CONTEXT Myocarditis is a known cause of death in athletes. As we consider clearance of athletes to participate in sports during the COVID-19 pandemic, we offer a brief review of the myocardial effects of SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) through the lens of what is known about myocarditis and exercise. All athletes should be queried about any recent illness suspicious for COVID-19 prior to sports participation. EVIDENCE ACQUISITION The PubMed database was evaluated through 2020, with the following keywords: myocarditis, COVID-19, SARS-CoV-2, cardiac, and athletes. Selected articles identified through the primary search, along with position statements from around the world, and the relevant references from those articles, were reviewed for pertinent clinical information regarding the identification, evaluation, risk stratification, and management of myocarditis in patients, including athletes, with and without SARS-CoV-2. STUDY DESIGN Systematic review. LEVEL OF EVIDENCE Level 3. RESULTS Since myocarditis can present with a variety of symptoms, and can be asymptomatic, the sports medicine physician needs to have a heightened awareness of athletes who may have had COVID-19 and be at risk for myocarditis and should have a low threshold to obtain further cardiovascular testing. Symptomatic athletes with SARS-CoV-2 may require cardiac evaluation including an electrocardiogram and possibly an echocardiogram. Athletes with cardiomyopathy may benefit from cardiac magnetic resonance imaging in the recovery phase and, rarely, endocardial biopsy. CONCLUSION Myocarditis is a known cause of sudden cardiac death in athletes. The currently reported rates of cardiac involvement of COVID-19 makes myocarditis a risk, and physicians who clear athletes for participation in sport as well as sideline personnel should be versed with the diagnosis, management, and clearance of athletes with suspected myocarditis. Given the potentially increased risk of arrhythmias, sideline personnel should practice their emergency action plans and be comfortable using an automated external defibrillator.
Collapse
Affiliation(s)
- Neha P. Raukar
- Department of Emergency Medicine,
Mayo Clinic, Rochester, Minnesota
| | - Leslie T. Cooper
- Department of Cardiovascular
Medicine, Mayo Clinic, Jacksonville, Florida
| |
Collapse
|
39
|
Adventitial Progenitor Cells of Human Great Saphenous Vein Enhance the Resolution of Venous Thrombosis via Neovascularization. Stem Cells Int 2021; 2021:8816763. [PMID: 33679991 PMCID: PMC7926266 DOI: 10.1155/2021/8816763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 01/20/2021] [Accepted: 02/06/2021] [Indexed: 11/24/2022] Open
Abstract
Background Vascular adventitia contains progenitor cells and is shown to participate in vascular remolding. Progenitor cells are recruited into the venous thrombi in mice to promote neovascularization. We hypothesized that the adventitial progenitor cells of human great saphenous vein (HGSV-AdPC) enhance the resolution of venous thrombosis via neovascularization. Methods Human great saphenous vein (HGSV) was harvested from the patients with great saphenous vein varicose and sectioned for immunohistochemistry, or minced for progenitor cell primary culture, or placed in sodium dodecyl sulfate solution for decellularization. Human venous thrombi were collected from patients with great saphenous vein varicose and superficial thrombophlebitis. Infrarenal abdominal aorta of New Zealand white rabbits was replaced with interposing decellularized vessel, and the patency of the grafts was confirmed by ultrasonic examination. Animal venous thrombi in the left infrarenal vena cava of mice were produced with Prolene suture ligation and ophthalmic force clipping of this portion. After HGSVs were digested by collagenase, the CD34+CD117+ HGSV-AdPC were isolated on FACS system, labelled with CM-Dil, and transplanted into the adventitia of infrarenal vena cava of nude mice. The percentage of thrombus organization area to the thrombus area was calculated as the organization rate. The thrombus cell, endothelial cells, and macrophages in the thrombi were counted in sections. Cell smears and frozen sections of human saphenous veins and venous thrombi were labeled with Sca1, CD34, CD117, Flk1, CD31, and F4/80 antibodies. The CD34+CD117+ HGSV-AdPC were cultured in endothelial growth medium with vascular endothelial growth factor (VEGF) to induce endothelial cell differentiation and analyzed with real time-PCR, Western blotting, and tube formation assays. Results Immunohistochemical staining showed that the CD34+CD117+ cells were located within the adventitia of HGSVs, and many CD34+ and CD117+ cells have emerged in the human venous thrombi. The number of progenitor cells within the marginal area of 7 days mice thrombi was shown to be Sca1+ ≈21%, CD34+ ≈12%, CD117+ ≈9%, and Flk1+ ≈5%. Many CD34+adventitial progenitor cells have migrated into the decellularized vessels. FACS showed that the number of CD34+CD117+ HGSV-AdPC in primary cultured cells as 1.2 ± 0.07%. After CD34+CD117+HGSV-AdPC were transplanted into the adventitia of nude mice vena cava with venous thrombi, the organization rate, nucleate cell count, endothelial cells, and macrophage cells of thrombi were shown to be significantly increased. The transplanted CD34+CD117+ HGSV-AdPC at the adventitia have crossed the vein wall, entered the venous thrombi, and differentiated into endothelial cells. The CD34+CD117+ HGSV-AdPC in the culture medium in the presence of VEGF-promoted gene and protein expression of endothelial cell markers in vitro and induced tube formation. Conclusions HGSV-AdPC could cross the vein wall and migrate from the adventitia into the venous thrombi. Increased HGSV-AdPC in the adventitia has enhanced the resolution of venous thrombi via differentiating into endothelial cells of neovascularization.
Collapse
|
40
|
Li H, Zhu H, Yang Z, Tang D, Huang L, Xia L. Application of Multiparametric Quantitative Cardiac Magnetic Resonance for Detection and Monitoring of Myocardial Injury in Patients with Fulminant Myocarditis. Acad Radiol 2021; 28:e35-e43. [PMID: 32199722 DOI: 10.1016/j.acra.2020.01.034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/29/2020] [Accepted: 01/29/2020] [Indexed: 11/25/2022]
Abstract
RATIONALE AND OBJECTIVES To investigate whether multiparametric cardiac magnetic resonance (CMR) could detect and monitor inflammatory myocardial alterations in fulminant myocarditis. MATERIALS AND METHODS Nineteen patients (35 ± 14 years, 37% male) with clinical diagnosis of fulminant myocarditis underwent CMR examinations at 3.0T in the acute phase and at 3-months follow up. The control group consisted of 19 healthy volunteers. The CMR protocol included cine, black blood T2-weighted imaging, T1 mapping, T2 mapping and late gadolinium enhancement (LGE). Cardiac parameters, such as edema ratio, LGE mass, native T1, T2 and extracellular volume were measured. RESULTS The left ventricular mass index (67 ± 15 versus 55 ± 12 g/m2, p < 0.05) and interventricular septum thickness (10.4 ± 1.5 versus 8.3 ± 1.8 mm, p < 0.001) in acute stage was significantly higher compared to controls, and normalized at the chronic stage. All quantitative inflammation metrics, including edema ratio, LGE mass, native T1, T2 and extracellular volume were significantly (all p < 0.001) decreased in the follow-up scan, but still higher compared to controls. Compared to the controls, all global strain indices including circumferential, longitudinal and radial strain values were significantly impaired in acute stage (all p < 0.001). Native T1 and T2 values led to excellent diagnostic accuracy for discriminating fulminant myocarditis from healed myocarditis, with AUC of 0.947 and 0.931. CONCLUSION Multiparametric CMR could detect and monitor inflammation myocardial injuries in patients with fulminant myocarditis. Native T1 and T2 values achieved excellent diagnostic performance in distinguishing acute from healed myocarditis.
Collapse
|
41
|
Fiegle DJ, Schöber M, Dittrich S, Cesnjevar R, Klingel K, Volk T, Alkassar M, Seidel T. Severe T-System Remodeling in Pediatric Viral Myocarditis. Front Cardiovasc Med 2021; 7:624776. [PMID: 33537349 PMCID: PMC7848076 DOI: 10.3389/fcvm.2020.624776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 12/22/2020] [Indexed: 12/26/2022] Open
Abstract
Chronic heart failure (HF) in adults causes remodeling of the cardiomyocyte transverse tubular system (t-system), which contributes to disease progression by impairing excitation-contraction (EC) coupling. However, it is unknown if t-system remodeling occurs in pediatric heart failure. This study investigated the t-system in pediatric viral myocarditis. The t-system and integrity of EC coupling junctions (co-localization of L-type Ca2+ channels with ryanodine receptors and junctophilin-2) were analyzed by 3D confocal microscopy in left-ventricular (LV) samples from 5 children with myocarditis (age 14 ± 3 months), undergoing ventricular assist device (VAD) implantation, and 5 children with atrioventricular septum defect (AVSD, age 17 ± 3 months), undergoing corrective surgery. LV ejection fraction (EF) was 58.4 ± 2.3% in AVSD and 12.2 ± 2.4% in acute myocarditis. Cardiomyocytes from myocarditis samples showed increased t-tubule distance (1.27 ± 0.05 μm, n = 34 cells) and dilation of t-tubules (volume-length ratio: 0.64 ± 0.02 μm2) when compared with AVSD (0.90 ± 0.02 μm, p < 0.001; 0.52 ± 0.02 μm2, n = 61, p < 0.01). Intriguingly, 4 out of 5 myocarditis samples exhibited sheet-like t-tubules (t-sheets), a characteristic feature of adult chronic heart failure. The fraction of extracellular matrix was slightly higher in myocarditis (26.6 ± 1.4%) than in AVSD samples (24.4 ± 0.8%, p < 0.05). In one case of myocarditis, a second biopsy was taken and analyzed at VAD explantation after extensive cardiac recovery (EF from 7 to 56%) and clinical remission. When compared with pre-VAD, t-tubule distance and density were unchanged, as well as volume-length ratio (0.67 ± 0.04 μm2 vs. 0.72 ± 0.05 μm2, p = 0.5), reflecting extant t-sheets. However, junctophilin-2 cluster density was considerably higher (0.12 ± 0.02 μm−3 vs. 0.05 ± 0.01 μm−3, n = 9/10, p < 0.001), approaching values of AVSD (0.13 ± 0.05 μm−3, n = 56), and the measure of intact EC coupling junctions showed a distinct increase (20.2 ± 5.0% vs. 6.8 ± 2.2%, p < 0.001). Severe t-system loss and remodeling to t-sheets can occur in acute HF in young children, resembling the structural changes of chronically failing adult hearts. T-system remodeling might contribute to cardiac dysfunction in viral myocarditis. Although t-system recovery remains elusive, recovery of EC coupling junctions may be possible and deserves further investigation.
Collapse
Affiliation(s)
- Dominik J Fiegle
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Schöber
- Department of Pediatric Cardiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sven Dittrich
- Department of Pediatric Cardiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Robert Cesnjevar
- Department of Pediatric Cardiac Surgery, University Hospital Erlangen, Erlangen, Germany
| | - Karin Klingel
- Cardiopathology, University Hospital Tuebingen, Tübingen, Germany
| | - Tilmann Volk
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Muscle Research Center Erlangen (MURCE), Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Muhannad Alkassar
- Department of Pediatric Cardiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Seidel
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Muscle Research Center Erlangen (MURCE), Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
42
|
Tajiri K, Imanaka-Yoshida K, Tsujimura Y, Matsuo K, Hiroe M, Aonuma K, Ieda M, Yasutomi Y. A New Mouse Model of Chronic Myocarditis Induced by Recombinant Bacille Calmette-Guèrin Expressing a T-Cell Epitope of Cardiac Myosin Heavy Chain-α. Int J Mol Sci 2021; 22:E794. [PMID: 33466825 PMCID: PMC7829923 DOI: 10.3390/ijms22020794] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is a potentially lethal disorder characterized by progressive impairment of cardiac function. Chronic myocarditis has long been hypothesized to be one of the causes of DCM. However, owing to the lack of suitable animal models of chronic myocarditis, its pathophysiology remains unclear. Here, we report a novel mouse model of chronic myocarditis induced by recombinant bacille Calmette-Guérin (rBCG) expressing a CD4+ T-cell epitope of cardiac myosin heavy chain-α (rBCG-MyHCα). Mice immunized with rBCG-MyHCα developed chronic myocarditis, and echocardiography revealed dilation and impaired contraction of ventricles, similar to those observed in human DCM. In the heart, CD62L-CD4+ T cells were increased and produced significant amounts of IFN-γ and IL-17 in response to cardiac myosin. Adoptive transfer of CD62L-CD4+ T cells induced myocarditis in the recipient mice, which indicated that CD62L-CD4+ T cells were the effector cells in this model. rBCG-MyHCα-infected dendritic cells produced proinflammatory cytokines and induced MyHCα-specific T-cell proliferation and Th1 and Th17 polarization. This novel chronic myocarditis mouse model may allow the identification of the central pathophysiological and immunological processes involved in the progression to DCM.
Collapse
Affiliation(s)
- Kazuko Tajiri
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Tsukuba 305-0843, Japan;
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan; (K.A.); (M.I.)
| | - Kyoko Imanaka-Yoshida
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine, Tsu 514-8507, Japan;
- Mie University Matrix Biology Research Center, Mie University Graduate School of Medicine, Tsu 514-8507, Japan
| | - Yusuke Tsujimura
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Tsukuba 305-0843, Japan;
- Leprosy Research Center, National Institute of Infectious Diseases, Higashimurayama 189-0002, Japan
| | - Kazuhiro Matsuo
- Department of Research and Development, Japan BCG Laboratory, Kiyose 204-0022, Japan;
| | - Michiaki Hiroe
- Department of Cardiology, National Center for Global Health and Medicine, Tokyo 162-8655, Japan;
| | - Kazutaka Aonuma
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan; (K.A.); (M.I.)
| | - Masaki Ieda
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan; (K.A.); (M.I.)
| | - Yasuhiro Yasutomi
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Tsukuba 305-0843, Japan;
| |
Collapse
|
43
|
Turgeon PY, Massot M, Beaupré F, Belzile D, Beaudoin J, Bernier M, Bourgault C, Germain V, Laliberté C, Morin J, Gervais P, Trahan S, Charbonneau É, Dagenais F, Sénéchal M. Effect of Acute Immunosuppression on Left Ventricular Recovery and Mortality in Fulminant Viral Myocarditis: A Case Series and Review of Literature. CJC Open 2020; 3:292-302. [PMID: 33778446 PMCID: PMC7985012 DOI: 10.1016/j.cjco.2020.10.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 10/30/2020] [Indexed: 12/26/2022] Open
Abstract
Background Fulminant viral myocarditis (FVM) is a rare cause of cardiogenic shock associated with high morbidity and mortality rates. An inappropriately activated immune system results in severe myocardial inflammation. Acute immunosuppressive therapy for FVM therefore gained in popularity and was described in numerous retrospective studies. Methods We conducted an extensive review of the literature and compared it with our single-centre retrospective review of all cases of FVM from 2009-2019 to evaluate the possible effect of acute immunosuppression with intravenous immunoglobulins and/or high dose corticosteroids in patients with FVM. Results We report on 17 patients with a mean age of 46 ± 15 years with a mean left ventricular ejection fraction (LVEF) of 15 ± 9% at admission. Fourteen (82%) of our patients had acute LVEF recovery to ≥ 45% after a mean time from immunosuppression of 74 ± 49 hours (3.1 days). Extracorporeal membrane oxygenation (ECMO) was required in 35% (6/17) of our patients for an average support of 126 ± 37 hours. Overall mortality was 12% (2/17). No patient needed a long-term left ventricular assist device or heart transplant. All surviving patients achieved complete long-term LVEF recovery. Conclusions Our cohort of 17 severely ill patients received acute immunosuppressive therapy and showed a rapid LVEF recovery, short duration of ECMO support, and low mortality rate. Our suggested scheme of investigation and treatment is presented. These results bring more cases of successfully treated FVM with immunosuppression and ECMO to the literature, which might stimulate further prospective trials or a registry.
Collapse
Affiliation(s)
- Pierre Yves Turgeon
- Department of Cardiology, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Québec, Canada
| | - Montse Massot
- Department of Cardiology, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Québec, Canada
| | - Frédéric Beaupré
- Department of Cardiology, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Québec, Canada
| | - David Belzile
- Department of Cardiology, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Québec, Canada
| | - Jonathan Beaudoin
- Department of Cardiology, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Québec, Canada
| | - Mathieu Bernier
- Department of Cardiology, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Québec, Canada
| | - Christine Bourgault
- Department of Cardiology, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Québec, Canada
| | - Valérie Germain
- Department of Cardiology, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Québec, Canada
| | - Claudine Laliberté
- Department of Cardiology, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Québec, Canada
| | - Joëlle Morin
- Department of Cardiology, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Québec, Canada
| | - Philippe Gervais
- Department of Infectious Disease, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Québec, Canada
| | - Sylvain Trahan
- Department of Pathology, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Canada
| | - Éric Charbonneau
- Department of Cardiac Surgery, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Québec, Canada
| | - François Dagenais
- Department of Cardiac Surgery, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Québec, Canada
| | - Mario Sénéchal
- Department of Cardiology, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Québec, Canada
| |
Collapse
|
44
|
Dolladille C, Ederhy S, Allouche S, Dupas Q, Gervais R, Madelaine J, Sassier M, Plane AF, Comoz F, Cohen AA, Thuny FR, Cautela J, Alexandre J. Late cardiac adverse events in patients with cancer treated with immune checkpoint inhibitors. J Immunother Cancer 2020; 8:jitc-2019-000261. [PMID: 31988143 PMCID: PMC7057417 DOI: 10.1136/jitc-2019-000261] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2019] [Indexed: 12/17/2022] Open
Abstract
Background Immune checkpoint inhibitor (ICI)-associated early cardiac adverse events (CAEs), mostly acute and fulminant myocarditis, have been well characterized and mainly occur during the first 90 days after ICI therapy initiation. ICI-associated late CAEs (occurring after the first 90 days of treatment) have not yet been described. Methods First, we compared characteristics of a cohort involving early (defined as a CAE time to onset (TTO) of <90 days after ICI therapy initiation) and late (defined as a CAE TTO of ≥90 days after ICI therapy initiation) ICI-associated CAE consecutive cases who were referred to three French cardio-oncology units. Second, ICI-associated CAE cases were searched in VigiBase, the WHO global individual case safety report database, and early and late ICI-associated CAEs were compared. Results In the cohort study, compared with early CAE cases (n=19, median TTO of 14 days), late ICI-associated CAE cases (n=19, median TTO of 304 days) exhibited significantly more left ventricular systolic dysfunction (LVSD) and heart failure (HF) and less frequent supraventricular arrhythmias. In VigiBase, compared with early cases (n=437, 73.3%, median TTO 21 days), the late ICI-associated CAE reports (n=159, 26.7%, median TTO 178 days) had significantly more frequent HF (21.1% vs 31.4%, respectively, p=0.01). Early and late ICI-associated CAE cases had similarly high mortality rates (40.0% vs 44.4% in the cohort and 30.0% vs 27.0% in VigiBase, respectively). Conclusions Late CAEs could occur with ICI therapy and were mainly revealed to be HF with LVSD. Trial registration numbers NCT03678337, NCT03882580, and NCT03492528.
Collapse
Affiliation(s)
- Charles Dolladille
- CHU de Caen, PICARO Cardio-oncology Program, Department of Pharmacology, CHU de Caen, Caen, France .,CHU de Caen, Department of Cardiology, CHU de Caen, Caen F-14000, France
| | - Stephane Ederhy
- Department of Cardiology, Cardio-oncology Unit, Cardio-oncology Research G, INSERM U 856, Thrombose, Athérothrombose et Pharmacologie Appliquée, Assistance Publique - Hopitaux de Paris, Paris, France
| | - Stéphane Allouche
- EA4650, Signalisation, Électrophysiologie et Imagerie des Lésions d'Ischémie-reperfusion Myocardique, Université de Caen Normandie, Caen, Normandie, France
| | - Querntin Dupas
- EA4650, Signalisation, Électrophysiologie et Imagerie des Lésions d'Ischémie-reperfusion Myocardique, Université de Caen Normandie, Caen, Normandie, France
| | - Radj Gervais
- Onco-pneumology, Centre François Baclesse Centre de Lutte Contre le Cancer, Caen, France
| | | | - Marion Sassier
- CHU de Caen, PICARO Cardio-oncology Program, Department of Pharmacology, CHU de Caen, Caen, France
| | - Anne-Flore Plane
- CHU de Caen, Department of Cardiology, CHU de Caen, Caen F-14000, France
| | | | - Ariel Aron Cohen
- Department of Cardiology, Cardio-oncology Unit, Cardio-oncology Research G, INSERM U 856, Thrombose, Athérothrombose et Pharmacologie Appliquée, Assistance Publique - Hopitaux de Paris, Paris, France
| | - Franck Roland Thuny
- Mediterranean University Cardio-oncology Center (MEDI-CO Center), Unit of Heart Failure and Valvular Heart Diseases, Department of Cardiology, Hôpital Nord, Marseille, APHM, Marseille, Provence-Alpes-Côte d'Azu, France
| | - Jennifer Cautela
- Mediterranean University Cardio-oncology Center (MEDI-CO Center), Unit of Heart Failure and Valvular Heart Diseases, Department of Cardiology, Hôpital Nord, Marseille, APHM, Marseille, Provence-Alpes-Côte d'Azu, France
| | - Joachim Alexandre
- CHU de Caen, PICARO Cardio-oncology Program, Department of Pharmacology, CHU de Caen, Caen, France.,EA4650, Signalisation, Électrophysiologie et Imagerie des Lésions d'Ischémie-reperfusion Myocardique, Université de Caen Normandie, Caen, Normandie, France
| |
Collapse
|
45
|
Abstract
Myocarditis in the pediatric population can be a challenging diagnosis to make and often requires utilization of multiple diagnostic modalities. The cause is often due to a viral infection with activation of the innate and acquired immune response with either recovery or disease progression. Laboratory testing often includes inflammatory markers, cardiac troponin levels, and natriuretic peptides. Noninvasive testing should include electrocardiogram, echocardiogram, and possibly an MRI. Treatment of myocarditis remains controversial with most providers using immune modulators with intravenous immunoglobulin and corticosteroids.
Collapse
|
46
|
Emrich T, Hahn F, Fleischmann D, Halfmann MC, Düber C, Varga-Szemes A, Escher F, Pefani E, Münzel T, Schultheiss HP, Kreitner KF, Wenzel P. T1 and T2 mapping to detect chronic inflammation in cardiac magnetic resonance imaging in heart failure with reduced ejection fraction. ESC Heart Fail 2020; 7:2544-2552. [PMID: 32790159 PMCID: PMC7524213 DOI: 10.1002/ehf2.12830] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/20/2020] [Accepted: 05/23/2020] [Indexed: 12/19/2022] Open
Abstract
Aims The purpose of this retrospective single‐centre study was to evaluate the non‐invasive detection of endomyocardial biopsy (EMB)‐established chronic myocardial inflammation in patients with heart failure with reduced ejection fraction (HFrEF) using T1 and T2 mapping. Methods and results The study population consisted of 52 retrospectively identified HFrEF patients who underwent EMB and cardiac magnetic resonance imaging at 3 Tesla. EMB was defined according to the position statement of the European Society of Cardiology and served as reference to identify inflammation in all patients. A control group of healthy volunteers with prior cardiac magnetic resonance imaging studies (n = 58) was also identified. Global and segmental T1 and T2 values as well as septal measurements and tissue heterogeneity parameters were calculated. Out of the 52 patients with HFrEF, 33 patients had myocardial inflammation detected by EMB, while 19 patients were EMB negative for inflammation. Mean left ventricular ejection fraction was 31% in both groups (P = 0.97). Global T1 and T2 values in HFrEF patients were significantly higher compared with healthy controls (T1 1275 ± 69 ms vs. 1,175 ± 44 ms, P < 0.001; T2 40.0 ± 3.4 ms vs. 37.9 ± 1.6 ms, P < 0.001). The distribution of T1 and T2 values between patients with and without EMB‐proven chronic myocardial inflammation was not statistically different when regarding global (T1 1292 ± 71 ms vs. 1266 ± 67 ms, P = 0.26; T2 40.0 ± 2.6 ms vs. 40.0 ± 3.9 ms, P = 1.0), septal (T1 1299 ± 63 ms vs. 1289 ± 76 ms, P = 0.76; T2 40.1 ± 3.5 ms vs 40.0 ± 6.4 ms, P = 0.49) or maximum segmental values (T1 1414 ± 111 ms vs. 1363 ± 88 ms, P = 0.15; T2 47.3 ± 5.2 ms vs. 48.8 ± 11.8 ms, P = 0.53). Mean absolute deviation of segmental T1 and T2 values and log‐transformed pixel‐wise standard deviation as parameters of tissue heterogeneity did not reveal statistical significant differences between inflammation‐positive and inflammation‐negative HFrEF patients (all P > 0.4). Conclusions Conventionally performed quantitative T1 and T2 mapping values significantly correlated with prevalence of HFrEF but did not discriminate HFrEF patients with or without chronic myocardial inflammation in our cohort. This suggests that EMB is the preferred method to detect chronic myocardial inflammation in HFrEF.
Collapse
Affiliation(s)
- Tilman Emrich
- Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), partner site Rhine-Main, Mainz, Germany
| | - Felix Hahn
- Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany
| | - David Fleischmann
- Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany
| | - Moritz C Halfmann
- Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany
| | - Christoph Düber
- Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany
| | - Akos Varga-Szemes
- Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC, USA
| | - Felicitas Escher
- IKDT Institut Kardiale Diagnostik und Therapie GmbH, Berlin, Germany.,Department of Cardiology, Campus Virchow-Klinikum, Charité-University Medicine Berlin, Berlin, Germany
| | - Evgenia Pefani
- Center for Cardiology, Cardiology 1, University Medical Center Mainz, Mainz, Germany
| | - Thomas Münzel
- German Center for Cardiovascular Research (DZHK), partner site Rhine-Main, Mainz, Germany.,Center for Cardiology, Cardiology 1, University Medical Center Mainz, Mainz, Germany
| | | | - Karl-Friedrich Kreitner
- Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany
| | - Philip Wenzel
- German Center for Cardiovascular Research (DZHK), partner site Rhine-Main, Mainz, Germany.,Center for Cardiology, Cardiology 1, University Medical Center Mainz, Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
47
|
Wang L, Zhang Y, Zhu G, Ma Y, Zuo H, Tian X. miR-16 exhibits protective function in LPS-treated cardiomyocytes by targeting DOCK2 to repress cell apoptosis and exert anti-inflammatory effect. Cell Biol Int 2020; 44:1760-1768. [PMID: 32369253 DOI: 10.1002/cbin.11371] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 04/23/2020] [Accepted: 05/03/2020] [Indexed: 12/12/2022]
Abstract
This study aims to investigate the effects of microRNA (miR)-16/dedicator of cytokinesis 2 (DOCK2) on myocarditis. The differences in the expression of genes in acute myocarditis were filtered out across Gene Expression Omnibus (GEO) database. Myocarditis cell model was established by lipopolysaccharide (LPS) stimulation in cardiomyocytes. The association between miR-16 and DOCK2 was predicted by bioinformatics software and confirmed by dual-luciferase assay. Polymerase chain reaction and western blot analysis were employed to assess the expression levels of miR-16 and DOCK2 under different conditions. Cells viability, apoptosis, and inflammatory reaction were evaluated by Cell Counting Kit-8, flow cytometry, and enzyme-linked immunosorbent assays. miR-16, as an upstream regulator of DOCK2, exhibited lower expression in LPS-induced myocarditis model. More importantly, we revealed that a marked augmentation of miR-16 promoted the growth of LPS-stimulated cardiomyocytes, and attenuated cell apoptosis and inflammatory response. However, an increasing expression of DOCK2 inhibited the remission of LPS-induced myocardial injury caused by miR-16 mimic. Herein, our results highlighted that upregulation of miR-16 resulted in the protective effects on LPS-induced myocardial injury by reducing DOCK2 expression, affording a pair of novel target molecules for ameliorating the symptoms of myocarditis.
Collapse
Affiliation(s)
- Lei Wang
- Department of Cardiology, Tengzhou Central People's Hospital Affiliated to Jining Medical College, Tengzhou, Shandong, China
| | - Yangyang Zhang
- Department of Cardiology, Tengzhou Central People's Hospital Affiliated to Jining Medical College, Tengzhou, Shandong, China
| | - Guangfu Zhu
- Intervention Room, Department of Cardiology, Tengzhou Central People's Hospital Affiliated to Jining Medical College, Tengzhou, Shandong, China
| | - Yuncong Ma
- Department of Cardiology, Tengzhou Central People's Hospital Affiliated to Jining Medical College, Tengzhou, Shandong, China
| | - Huan Zuo
- Department of Neurology, Tengzhou Traditional Chinese Medicine Hospital, Tengzhou, Shandong, China
| | - Xia Tian
- Intervention Room, Department of Cardiology, Tengzhou Central People's Hospital Affiliated to Jining Medical College, Tengzhou, Shandong, China
| |
Collapse
|
48
|
Abstract
Dilated cardiomyopathy (DCM) is the most common form of cardiomyopathy and one of the most common causes of heart failure. It is characterized by left or biventricular dilation and a reduced systolic function. The causes are manifold and range from myocarditis to alcohol and other toxins, to rheumatological, endocrinological, and metabolic diseases. Peripartum cardiomyopathy is a special form that occurs at the end of or shortly after pregnancy. Genetic mutations can be detected in approximately 30-50% of DCM patients. Owing to the growing possibilities of genetic diagnostics, increasingly more triggering variants and hereditary mechanisms emerge. This is particularly important with regard to risk stratification for patients with variants with an increased risk of arrhythmias. Patient prognosis is determined by the occurrence of heart failure and arrhythmias. In addition to the treatment of the underlying disease or the elimination of triggering harmful toxins, therapy consists in guideline-directed heart failure treatment including drug and device therapy.
Collapse
Affiliation(s)
- A Hänselmann
- Dept. of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - C Veltmann
- Dept. of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - J Bauersachs
- Dept. of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - D Berliner
- Dept. of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
49
|
Wang L, Zhang Y, Zhang S. Cardiovascular Impairment in COVID-19: Learning From Current Options for Cardiovascular Anti-Inflammatory Therapy. Front Cardiovasc Med 2020; 7:78. [PMID: 32426374 PMCID: PMC7203508 DOI: 10.3389/fcvm.2020.00078] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 04/15/2020] [Indexed: 12/15/2022] Open
Abstract
In December 2019, Coronavirus Disease 2019 (COVID-19) caused by SARS-CoV-2, occurred in China and has currently led to a global pandemic. In addition to respiratory involvement, COVID-19 was also associated with significant multiple organ dysfunction syndrome (MODS). Cardiovascular impairment has been observed and is now drawing growing attention. Cardiovascular protective strategies are urgent and of great significance to the overall prognosis of COVID-19 patients. Direct viral infection, cytokine storm, and aggravation of existing cardiovascular diseases were recognized as possible mechanisms of cardiovascular impairment in COVID-19. Hyperactivated inflammation plays an important role in all three mechanisms and is considered to be fundamental in the development of cardiovascular impairment and MODS in COVID-19. Therefore, in addition to conventional cardiovascular treatment, anti-inflammatory therapy is a reasonable strategy for severe cases to further enhance cardiovascular protection and potentially mitigate MODS. We reviewed the inflammatory features and current promising treatments of COVID-19 as well as cardiovascular anti-inflammatory therapies that have been verified in previous clinical trials with positive outcomes. We believe that targeting the central pathway (IL-1β, TNF-α, IL-6), balancing the Th1 and Th2 response, and administering long-term anti-inflammatory therapy might be promising prospects to reduce cardiovascular impairment and even MODS during the acute and recovery phases of COVID-19. The cardiovascular anti-inflammatory therapies might be of great application value to the management of COVID-19 patients and we further propose an algorithm for the selection of anti-inflammatory therapy for COVID-19 patients with or at high risk of cardiovascular impairment. We recommend to take the experiences in cardiovascular anti-inflammatory therapy as references in the management of COVID-19 and conduct related clinical trials, while the clinical translation of novel treatments from preclinical studies or in vitro drug screening should proceed with caution due to unguaranteed efficacy and safety profiles.
Collapse
Affiliation(s)
| | | | - Shuyang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| |
Collapse
|
50
|
Che Y, Su Z, Xia L. Effects of IL-22 on cardiovascular diseases. Int Immunopharmacol 2020; 81:106277. [PMID: 32062077 DOI: 10.1016/j.intimp.2020.106277] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/29/2020] [Accepted: 02/01/2020] [Indexed: 12/11/2022]
Abstract
Interleukin-22 (IL-22), which belongs to the IL-10 family, is an alpha helix cytokine specifically produced by many lymphocytes, such as Th1, Th17, Th22, ILCs, CD4+ and CD8+ T cells. In recent years, more and more studies have demonstrated that IL-22 has an interesting relationship with various cardiovascular diseases, including myocarditis, myocardial infarction, atherosclerosis, and other cardiovascular diseases, and IL-22 signal may play a dual role in cardiovascular diseases. Here, we summarize the recent progress on the source, function, regulation of IL-22 and the effects of IL-22 signal in cardiovascular diseases. The study of IL-22 will suggest more specific strategies to maneuver these functions for the effective treatment of cardiovascular diseases and future clinical treatment.
Collapse
Affiliation(s)
- Yang Che
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; International Genome Center, Jiangsu University, Zhenjiang 212013, China
| | - Zhaoliang Su
- International Genome Center, Jiangsu University, Zhenjiang 212013, China; Department of Immunology, Jiangsu University, Zhenjiang 212013, China
| | - Lin Xia
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; International Genome Center, Jiangsu University, Zhenjiang 212013, China.
| |
Collapse
|