1
|
Kumar S, Shah G, Nair R, Rikabi S, Seif M, Ghimire B, Griffin B, Khot UN. Characteristics and Outcomes of New-Onset Cardiomyopathy in Hospitalized COVID-19 Patients. J Clin Med 2025; 14:3258. [PMID: 40364288 PMCID: PMC12072776 DOI: 10.3390/jcm14093258] [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: 03/06/2025] [Revised: 04/24/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Background: The association between Coronavirus Disease-2019 (COVID-19) and new-onset cardiomyopathy (NOC) is unclear. Objectives: We aim to assess the incidence of NOC in hospitalized COVID-19 patients and its impact on short- and long-term survival. Methods: We retrospectively studied 2219 COVID-19 patients hospitalized between March 2020 and February 2022 who underwent an in-hospital echocardiogram. NOC was defined as a left-ventricular ejection fraction (LVEF) reduction of >10%, resulting in an LVEF of <54% for females and <52% for males. The 30-day and 1-year survival outcomes in patients without and with NOC were studied. Results: Among 25,943 hospitalized COVID-19 patients, 2219 met our inclusion criteria, with 209 (9.4%) having NOC. NOC patients were more likely to be male (56.1% vs. 68.4%, p = 0.001) and have chronic kidney disease (51.4% vs. 60.3%, p = 0.018). They had a higher 30-day mortality rate (29.1% vs. 32%, p = 0.033), but the 1-year survival rate was similar between the patients without and with NOC (36.9% vs. 41.6%, p = 0.12). Multivariable regression revealed that advanced age, admission to intensive care unit, mechanical ventilation, treatment with glucocorticoids, and treatment with vasopressors were associated with higher odds of 30-day mortality in NOC patients. Only 74 (35.4%) NOC patients had follow-up echocardiograms after discharge, of which 47 showed persistent cardiomyopathy. Conclusions: NOC can affect around 1 out of 10 hospitalized COVID-19 patients undergoing echocardiography. While NOC was associated with worse short-term survival, it did not impact the long-term mortality of these patients. Persistent LVEF deficits in some patients emphasize the need for improved outpatient follow-up to identify at-risk individuals and optimize treatment.
Collapse
Affiliation(s)
- Sachin Kumar
- Department of Cardiovascular Medicine, Mount Sinai Morningside, New York, NY 10025, USA
| | - Gautam Shah
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Raunak Nair
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Sarah Rikabi
- Department of Internal Medicine, Cleveland Clinic Fairview Hospital, Cleveland, OH 44111, USA
| | - Mohannad Seif
- Department of Internal Medicine, Cleveland Clinic Fairview Hospital, Cleveland, OH 44111, USA
| | - Bindesh Ghimire
- Department of Internal Medicine, Cleveland Clinic Fairview Hospital, Cleveland, OH 44111, USA
| | - Brian Griffin
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Umesh N. Khot
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
2
|
Zhao Q, Li Z, Wang DW, Li F, Zhang L, Jiang J. Lactate Dehydrogenase Indicates Development From Fulminant Myocarditis to Chronic Persistent Myocarditis: A Multicenter Retrospective Cohort Study in China. Int J Gen Med 2025; 18:1333-1344. [PMID: 40078751 PMCID: PMC11897539 DOI: 10.2147/ijgm.s509291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Background Fulminant myocarditis (FM) is a critical manifestation of myocarditis. However, the clinical features and risk factors associated with its adverse outcomes are not fully understood. Given the high mortality and potential for long-term complications, it is crucial to identify factors that could predict the progression of FM to chronic persistent myocarditis. We hypothesize some clinical or laboratory markers may be predictive of this progression. This study aims to identify clinical factors that may help predict the progress of FM to chronic persistent myocarditis. Methods A total of 82 patients with FM treated based on Chinese protocol were included. Kaplan-Meier curve and regression analysis were used to determine the clinical features and prognostic predictors of chronic persistent myocarditis in patients with 24 months of follow-up. Results Chronic persistent myocarditis was observed in 20 patients during the follow-up. ROC curve showed that the critical value for chronic persistent myocarditis caused by lactate dehydrogenase (LDH) was 577.00U/L (sensitivity 75.0%, specificity 74.2%). Time from onset to admission over 6 days and LDH > 577.00U/L were identified as risk factors for chronic persistent myocarditis in patients with FM in both univariate and multivariate cox analysis. The hazard ratio and 95% Confidence intervals were 3.35 (1.32-8.50) (p = 0.011) and 6.11 (2.02-18.48) (p < 0.001), respectively. The per standard deviation of increment in LDH was associated with the 55% (1.55, 1.11-2.18) in HR and 95% CI of the occurrence of chronic persistent myocarditis. Conclusion About 24.4% of the patients with FM treated based on the life support measures proposed in the consensus of Chinese Society of Cardiology have been observed chronic persistent myocarditis. Time from onset to admission over 6 days and LDH levels >577.00 U/L at admission may serve as risk factors for the progression from FM to chronic persistent myocarditis.
Collapse
Affiliation(s)
- Qu Zhao
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, People’s Republic of China
| | - Zeping Li
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, People’s Republic of China
| | - Dao Wen Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, People’s Republic of China
| | - Fan Li
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, People’s Republic of China
| | - Li Zhang
- Division of Cardiology, Minda Hospital of Hubei Minzu University, Enshi, 445099, People’s Republic of China
| | - Jiangang Jiang
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, People’s Republic of China
| |
Collapse
|
3
|
Šerý O, Dziedzinska R. Risk impact of SARS-CoV-2 coronavirus and spike protein on cardiac tissue: a comprehensive review. Physiol Res 2024; 73:S655-S669. [PMID: 39808169 PMCID: PMC11827061 DOI: 10.33549/physiolres.935476] [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: 04/25/2024] [Accepted: 08/19/2024] [Indexed: 01/18/2025] Open
Abstract
The global COVID-19 pandemic, caused by SARS-CoV-2, has led to significant morbidity and mortality, with a profound impact on cardiovascular health. This review investigates the mechanisms of SARS-CoV-2's interaction with cardiac tissue, particularly emphasizing the role of the Spike protein and ACE2 receptor in facilitating viral entry and subsequent cardiac complications. We dissect the structural features of the virus, its interactions with host cell receptors, and the resulting pathophysiological changes in the heart. Highlighting SARS-CoV-2's broad organ tropism, especially its effects on cardiomyocytes via ACE2 and TMPRSS2, the review addresses how these interactions exacerbate cardiovascular issues in patients with pre-existing conditions such as diabetes and hypertension. Additionally, we assess both direct and indirect mechanisms of virus-induced cardiac damage, including myocarditis, arrhythmias, and long-term complications such as 'long COVID'. This review underscores the complexity of SARS-CoV-2's impact on the heart, emphasizing the need for ongoing research to fully understand its long-term effects on cardiovascular health. Key words: COVID-19, Heart, ACE2, Spike protein, Cardiomyocytes, Myocarditis, Long COVID.
Collapse
Affiliation(s)
- O Šerý
- Laboratory of Neurobiology and Molecular Psychiatry, Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic.
| | | |
Collapse
|
4
|
Dmytrenko O, Das S, Kovacs A, Cicka M, Liu M, Scheaffer SM, Bredemeyer A, Mack M, Diamond MS, Lavine KJ. Infiltrating monocytes drive cardiac dysfunction in a cardiomyocyte-restricted mouse model of SARS-CoV-2 infection. J Virol 2024; 98:e0117924. [PMID: 39207134 PMCID: PMC11406924 DOI: 10.1128/jvi.01179-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Cardiovascular manifestations of coronavirus disease 2019 (COVID-19) include myocardial injury, heart failure, and myocarditis and are associated with long-term disability and mortality. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA and antigens are found in the myocardium of COVID-19 patients, and human cardiomyocytes are susceptible to infection in cell or organoid cultures. While these observations raise the possibility that cardiomyocyte infection may contribute to the cardiac sequelae of COVID-19, a causal relationship between cardiomyocyte infection and myocardial dysfunction and pathology has not been established. Here, we generated a mouse model of cardiomyocyte-restricted infection by selectively expressing human angiotensin-converting enzyme 2 (hACE2), the SARS-CoV-2 receptor, in cardiomyocytes. Inoculation of Myh6-Cre Rosa26loxP-STOP-loxP-hACE2 mice with an ancestral, non-mouse-adapted strain of SARS-CoV-2 resulted in viral replication within the heart, accumulation of macrophages, and moderate left ventricular (LV) systolic dysfunction. Cardiac pathology in this model was transient and resolved with viral clearance. Blockade of monocyte trafficking reduced macrophage accumulation, suppressed the development of LV systolic dysfunction, and promoted viral clearance in the heart. These findings establish a mouse model of SARS-CoV-2 cardiomyocyte infection that recapitulates features of cardiac dysfunctions of COVID-19 and suggests that both viral replication and resultant innate immune responses contribute to cardiac pathology.IMPORTANCEHeart involvement after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection occurs in multiple ways and is associated with worse outcomes in coronavirus disease 2019 (COVID-19) patients. It remains unclear if cardiac disease is driven by primary infection of the heart or immune response to the virus. SARS-CoV-2 is capable of entering contractile cells of the heart in a culture dish. However, it remains unclear how such infection affects the function of the heart in the body. Here, we designed a mouse in which only heart muscle cells can be infected with a SARS-CoV-2 strain to study cardiac infection in isolation from other organ systems. In our model, infected mice show viral infection, worse function, and accumulation of immune cells in the heart. A subset of immune cells facilitates such worsening heart function. As this model shows features similar to those observed in patients, it may be useful for understanding the heart disease that occurs as a part of COVID-19.
Collapse
Affiliation(s)
- Oleksandr Dmytrenko
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Shibali Das
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Attila Kovacs
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Markus Cicka
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Meizi Liu
- Department of Medicine, Infectious Disease, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Suzanne M. Scheaffer
- Department of Medicine, Infectious Disease, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Andrea Bredemeyer
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Matthias Mack
- Department of Internal Medicine II, Division of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Michael S. Diamond
- Department of Medicine, Infectious Disease, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Kory J. Lavine
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Developmental Biology, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
5
|
Xu C, Sha Y, Pan J, Pan T, Zhou X, Wang H, Xu Z, Chen B. COVID-19 related acute necrotizing encephalopathy and acute myocarditis in an adult female: a novel case report of brain injury and myocarditis. BMC Neurol 2024; 24:274. [PMID: 39107681 PMCID: PMC11302840 DOI: 10.1186/s12883-024-03786-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Acute necrotizing encephalopathy (ANE) and myocarditis are both acute, life-threatening conditions that can be triggered by COVID-19. We report a case of sequential ANE and myocarditis following a COVID-19 infection. CASE PRESENTATION A 27-year-old female patient was brought to the emergency department due to episodes of fever for two days and a 9-h altered state of consciousness. Her condition rapidly developed into stuporous and hemodynamic instability within serval hours. Veno-arterial extracorporeal membrane oxygenation (ECMO) was rapidly initiated with other supportive treatments. The following-up MRI showed bilateral, symmetrically distributed lesions in the brainstem, bilateral hippocampal regions, and bilateral basal ganglia, consistent with ANE. The diagnosis was confirmed through the detection of SARS-CoV-2 and the exclusion of other potential causes. After weeks of medical treatment, her condition stabilized, and she was transferred for further rehabilitation treatment. CONCLUSIONS This case study indicates that COVID-19 may simultaneously and rapidly affect the central nervous system and cardiovascular system, leading to poor outcomes. Accurate diagnosis and timely invasive bridging therapy, when necessary, can be lifesaving. Further exploration of potential mechanisms underlying COVID-19 central nervous system (CNS) and cardiovascular system manifestations will be important.
Collapse
Affiliation(s)
- Chang Xu
- Department of Intensive Care Medicine, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Yuyi Sha
- Department of Intensive Care Medicine, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Jianneng Pan
- Department of Intensive Care Medicine, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Tao Pan
- Department of Intensive Care Medicine, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Xiaoyang Zhou
- Department of Intensive Care Medicine, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Hua Wang
- Department of Intensive Care Medicine, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Zhaojun Xu
- Department of Intensive Care Medicine, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Bixin Chen
- Department of Intensive Care Medicine, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China.
| |
Collapse
|
6
|
Yan A, Zhang R, Feng C, Feng J. Coronavirus disease 2019-related myocarditis genes contribute to ECMO prognosis. BMC Cardiovasc Disord 2024; 24:375. [PMID: 39026189 PMCID: PMC11264513 DOI: 10.1186/s12872-024-04032-5] [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: 02/20/2024] [Accepted: 07/05/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Acute myocardial injury, cytokine storms, hypoxemia and pathogen-mediated damage were the major causes responsible for mortality induced by coronavirus disease 2019 (COVID-19)-related myocarditis. These need ECMO treatment. We investigated differentially expressed genes (DEGs) in patients with COVID-19-related myocarditis and ECMO prognosis. METHODS GSE150392 and GSE93101 were analyzed to identify DEGs. A Venn diagram was used to obtain the same transcripts between myocarditis-related and ECMO-related DEGs. Enrichment pathway analysis was performed and hub genes were identified. Pivotal miRNAs, transcription factors, and chemicals with the screened gene interactions were identified. The GSE167028 dataset and single-cell sequencing data were used to validate the screened genes. RESULTS Using a Venn diagram, 229 overlapping DEGs were identified between myocarditis-related and ECMO-related DEGs, which were mainly involved in T cell activation, contractile actin filament bundle, actomyosin, cyclic nucleotide phosphodiesterase activity, and cytokine-cytokine receptor interaction. 15 hub genes and 15 neighboring DEGs were screened, which were mainly involved in the positive regulation of T cell activation, integrin complex, integrin binding, the PI3K-Akt signaling pathway, and the TNF signaling pathway. Data in GSE167028 and single-cell sequencing data were used to validate the screened genes, and this demonstrated that the screened genes CCL2, APOE, ITGB8, LAMC2, COL6A3 and TNC were mainly expressed in fibroblast cells; IL6, ITGA1, PTK2, ITGB5, IL15, LAMA4, CAV1, SNCA, BDNF, ACTA2, CD70, MYL9, DPP4, ENO2 and VEGFC were expressed in cardiomyocytes; IL6, PTK2, ITGB5, IL15, APOE, JUN, SNCA, CD83, DPP4 and ENO2 were expressed in macrophages; and IL6, ITGA1, PTK2, ITGB5, IL15, VCAM1, LAMA4, CAV1, ACTA2, MYL9, CD83, DPP4, ENO2, VEGFC and IL32 were expressed in vascular endothelial cells. CONCLUSION The screened hub genes, IL6, ITGA1, PTK2, ITGB3, ITGB5, CCL2, IL15, VCAM1, GZMB, APOE, ITGB8, LAMA4, LAMC2, COL6A3 and TNFRSF9, were validated using GEO dataset and single-cell sequencing data, which may be therapeutic targets patients with myocarditis to prevent MI progression and adverse cardiovascular events.
Collapse
Affiliation(s)
- An Yan
- Tianjin Chest Hospital, Taierzhuang North Road 261, Jinnan District, Tianjin, China
- Tianjin Key Laboratory of Cardiovascular Emergency and Critical Care, Taierzhuang North Road 261, Jinnan District, Tianjin, China
| | - Ruiying Zhang
- Tianjin Chest Hospital, Taierzhuang North Road 261, Jinnan District, Tianjin, China
- Tianjin Key Laboratory of Cardiovascular Emergency and Critical Care, Taierzhuang North Road 261, Jinnan District, Tianjin, China
| | - Chao Feng
- Tianjin Chest Hospital, Taierzhuang North Road 261, Jinnan District, Tianjin, China
- Tianjin Key Laboratory of Cardiovascular Emergency and Critical Care, Taierzhuang North Road 261, Jinnan District, Tianjin, China
| | - Jinping Feng
- Tianjin Chest Hospital, Taierzhuang North Road 261, Jinnan District, Tianjin, China.
- Tianjin Key Laboratory of Cardiovascular Emergency and Critical Care, Taierzhuang North Road 261, Jinnan District, Tianjin, China.
| |
Collapse
|
7
|
Xian SP, Li ZY, Li W, Yang PF, Huang SH, Liu Y, Tang L, Lai J, Zeng FM, He JZ, Liu Y. Spatial immune landscapes of SARS-CoV-2 gastrointestinal infection: macrophages contribute to local tissue inflammation and gastrointestinal symptoms. Front Cell Dev Biol 2024; 12:1375354. [PMID: 39100091 PMCID: PMC11295004 DOI: 10.3389/fcell.2024.1375354] [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: 01/23/2024] [Accepted: 07/02/2024] [Indexed: 08/06/2024] Open
Abstract
Background In some patients, persistent gastrointestinal symptoms like abdominal pain, nausea, and diarrhea occur as part of long COVID-19 syndrome following acute respiratory symptoms caused by SARS-CoV-2. However, the characteristics of immune cells in the gastrointestinal tract of COVID-19 patients and their association with these symptoms remain unclear. Methodology Data were collected from 95 COVID-19 patients. Among this cohort, 11 patients who exhibited gastrointestinal symptoms and underwent gastroscopy were selected. Using imaging mass cytometry, the gastrointestinal tissues of these patients were thoroughly analyzed to identify immune cell subgroups and investigate their spatial distribution. Results Significant acute inflammatory responses were found in the gastrointestinal tissues, particularly in the duodenum, of COVID-19 patients. These alterations included an increase in the levels of CD68+ macrophages and CD3+CD4+ T-cells, which was more pronounced in tissues with nucleocapsid protein (NP). The amount of CD68+ macrophages positively correlates with the number of CD3+CD4+ T-cells (R = 0.783, p < 0.001), additionally, spatial neighborhood analysis uncovered decreased interactions between CD68+ macrophages and multiple immune cells were noted in NP-positive tissues. Furthermore, weighted gene coexpression network analysis was employed to extract gene signatures related to clinical features and immune responses from the RNA-seq data derived from gastrointestinal tissues from COVID-19 patients, and we validated that the MEgreen module shown positive correlation with clinical parameter (i.e., Total bilirubin, ALT, AST) and macrophages (R = 0.84, p = 0.001), but negatively correlated with CD4+ T cells (R = -0.62, p = 0.004). By contrast, the MEblue module was inversely associated with macrophages and positively related with CD4+ T cells. Gene function enrichment analyses revealed that the MEgreen module is closely associated with biological processes such as immune response activation, signal transduction, and chemotaxis regulation, indicating its role in the gastrointestinal inflammatory response. Conclusion The findings of this study highlight the role of specific immune cell groups in the gastrointestinal inflammatory response in COVID-19 patients. Gene coexpression network analysis further emphasized the importance of the gene modules in gastrointestinal immune responses, providing potential molecular targets for the treatment of COVID-19-related gastrointestinal symptoms.
Collapse
Affiliation(s)
- Shi-Ping Xian
- Department of Pathology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Zhan-Yu Li
- Department of Pathology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Wei Li
- Department of Pathology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Peng-Fei Yang
- Department of Pathology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Shen-Hao Huang
- Department of Pathology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Ye Liu
- Department of Pathology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Lei Tang
- Department of Pathology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Jun Lai
- Department of Pathology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Fa-Min Zeng
- Department of Pathology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Jian-Zhong He
- Department of Pathology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Yang Liu
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
- Department of Ophthalmology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| |
Collapse
|
8
|
Kubiliute I, Urboniene J, Majauskaite F, Bobkov E, Svetikas L, Jancoriene L. Elevated Cardiac Troponin I as a Mortality Predictor in Hospitalised COVID-19 Patients. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:842. [PMID: 38929459 PMCID: PMC11205688 DOI: 10.3390/medicina60060842] [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: 04/03/2024] [Revised: 05/05/2024] [Accepted: 05/17/2024] [Indexed: 06/28/2024]
Abstract
Background and Objectives: SARS-CoV-2 affects multiple organ systems, including the cardiovascular system, leading to immediate and long-term cardiovascular complications. Acute myocardial injury is one of the earliest and most common cardiac issues in the acute phase of COVID-19. This study aimed to evaluate the prognostic value of cardiac troponin I (cTnI) levels in predicting in-hospital mortality among hospitalised COVID-19 patients. Materials and Methods: A retrospective observational cohort study included 2019 adult patients hospitalised with a confirmed COVID-19 infection stratified by cTnI levels on admission into three groups: <19 ng/L (1416 patients), 19-100 ng/L (431 patients), and >100 ng/L (172 patients). Myocardial injury was defined as blood serum cTnI levels increased above the 99th percentile upper reference limit. Depersonalised datasets were extracted from digital health records. Statistical analysis included multivariable binary logistic and Cox proportional hazards regressions. Results: Overall, 29.87% of patients experienced acute myocardial injury, which development was associated with age, male sex, chronic heart failure, arterial hypertension, obesity, and chronic kidney disease. Among patients with cTnI levels of 19-100 ng/L, the odds ratio for requiring invasive mechanical ventilation was 3.18 (95% CI 2.11-4.79) and, for those with cTnI > 100 ng/L, 5.38 (95% CI 3.26-8.88). The hazard ratio for in-hospital mortality for patients with cTnI levels of 19-100 ng/L was 2.58 (95% CI 1.83-3.62) and, for those with cTnI > 100 ng/L, 2.97 (95% CI 2.01-4.39) compared to patients with normal cTnI levels. Conclusions: Increased cardiac troponin I, indicating myocardial injury, on admission is associated with a more adverse clinical disease course, including a higher likelihood of requiring invasive mechanical ventilation and increased risk of in-hospital mortality. This indicates cardiac troponin I to be a beneficial biomarker for clinicians trying to identify high-risk COVID-19 patients, choosing the optimal monitoring and treatment strategy for these patients.
Collapse
Affiliation(s)
- Ieva Kubiliute
- Clinic of Infectious Diseases and Dermatovenerology, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania; (F.M.); (L.S.); (L.J.)
| | - Jurgita Urboniene
- Centre of Infectious Diseases, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania;
| | - Fausta Majauskaite
- Clinic of Infectious Diseases and Dermatovenerology, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania; (F.M.); (L.S.); (L.J.)
| | - Edgar Bobkov
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania;
| | - Linas Svetikas
- Clinic of Infectious Diseases and Dermatovenerology, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania; (F.M.); (L.S.); (L.J.)
| | - Ligita Jancoriene
- Clinic of Infectious Diseases and Dermatovenerology, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania; (F.M.); (L.S.); (L.J.)
| |
Collapse
|
9
|
Lu RXZ, Zhao Y, Radisic M. The emerging role of heart-on-a-chip systems in delineating mechanisms of SARS-CoV-2-induced cardiac dysfunction. Bioeng Transl Med 2024; 9:e10581. [PMID: 38818123 PMCID: PMC11135153 DOI: 10.1002/btm2.10581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/20/2023] [Accepted: 07/10/2023] [Indexed: 06/01/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) has been a major global health concern since its emergence in 2019, with over 680 million confirmed cases as of April 2023. While COVID-19 has been strongly associated with the development of cardiovascular complications, the specific mechanisms by which viral infection induces myocardial dysfunction remain largely controversial as studies have shown that the severe acute respiratory syndrome coronavirus-2 can lead to heart failure both directly, by causing damage to the heart cells, and indirectly, by triggering an inflammatory response throughout the body. In this review, we summarize the current understanding of potential mechanisms that drive heart failure based on in vitro studies. We also discuss the significance of three-dimensional heart-on-a-chip technology in the context of the current and future pandemics.
Collapse
Affiliation(s)
- Rick Xing Ze Lu
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioCanada
| | - Yimu Zhao
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioCanada
- Toronto General Hospital Research InstituteUniversity Health NetworkTorontoOntarioCanada
| | - Milica Radisic
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioCanada
- Toronto General Hospital Research InstituteUniversity Health NetworkTorontoOntarioCanada
- Department of Chemical Engineering and Applied ChemistryUniversity of TorontoTorontoOntarioCanada
- Terence Donnelly Centre for Cellular & Biomolecular ResearchUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
10
|
Casimero FVC, Patalas ED, Stone JR. Sudden unexpected death in a middle-aged woman. J Clin Pathol 2024; 77:297-300. [PMID: 38316542 DOI: 10.1136/jcp-2022-208691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 06/03/2023] [Indexed: 02/07/2024]
Affiliation(s)
| | - Eva D Patalas
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - James R Stone
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| |
Collapse
|
11
|
Lu RXZ, Rafatian N, Zhao Y, Wagner KT, Beroncal EL, Li B, Lee C, Chen J, Churcher E, Vosoughi D, Liu C, Wang Y, Baker A, Trahtemberg U, Li B, Pierro A, Andreazza AC, dos Santos CC, Radisic M. Cardiac tissue model of immune-induced dysfunction reveals the role of free mitochondrial DNA and the therapeutic effects of exosomes. SCIENCE ADVANCES 2024; 10:eadk0164. [PMID: 38536913 PMCID: PMC10971762 DOI: 10.1126/sciadv.adk0164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 02/22/2024] [Indexed: 04/04/2024]
Abstract
Despite tremendous progress in the development of mature heart-on-a-chip models, human cell-based models of myocardial inflammation are lacking. Here, we bioengineered a vascularized heart-on-a-chip with circulating immune cells to model severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced acute myocarditis. We observed hallmarks of coronavirus disease (COVID-19)-induced myocardial inflammation, as the presence of immune cells augmented the secretion of proinflammatory cytokines, triggered progressive impairment of contractile function, and altered intracellular calcium transients. An elevation of circulating cell-free mitochondrial DNA (ccf-mtDNA) was measured first in the heart-on-a-chip and then validated in COVID-19 patients with low left ventricular ejection fraction, demonstrating that mitochondrial damage is an important pathophysiological hallmark of inflammation-induced cardiac dysfunction. Leveraging this platform in the context of SARS-CoV-2-induced myocardial inflammation, we established that administration of endothelial cell-derived exosomes effectively rescued the contractile deficit, normalized calcium handling, elevated the contraction force, and reduced the ccf-mtDNA and cytokine release via Toll-like receptor-nuclear factor κB signaling axis.
Collapse
Affiliation(s)
- Rick Xing Ze Lu
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Mitochondrial Innovation Initiative, MITO2i, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Naimeh Rafatian
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Yimu Zhao
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Karl T. Wagner
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Erika L. Beroncal
- Mitochondrial Innovation Initiative, MITO2i, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Bo Li
- Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Carol Lee
- Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Jingan Chen
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Eryn Churcher
- Interdepartmental Division of Critical Care, Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Daniel Vosoughi
- Latner Thoracic Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2C4, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Chuan Liu
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Ying Wang
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Andrew Baker
- Interdepartmental Division of Critical Care, Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Uriel Trahtemberg
- Interdepartmental Division of Critical Care, Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
- Galilee Medical Center, Nahariya, Israel
| | - Bowen Li
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Agostino Pierro
- Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - Ana C. Andreazza
- Mitochondrial Innovation Initiative, MITO2i, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Claudia C. dos Santos
- Interdepartmental Division of Critical Care, Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Mitochondrial Innovation Initiative, MITO2i, University of Toronto, Toronto, ON M5S 1A8, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2C4, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON M5S 3D5, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON, M5S 3E1
| |
Collapse
|
12
|
Ono R, Iwahana T, Aoki K, Kato H, Okada S, Kobayashi Y. Fulminant Myocarditis with SARS-CoV-2 Infection: A Narrative Review from the Case Studies. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2024; 2024:9000598. [PMID: 38469104 PMCID: PMC10927348 DOI: 10.1155/2024/9000598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 03/13/2024]
Abstract
One of the severe complications of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is myocarditis. However, the characteristics of fulminant myocarditis with SARS-CoV-2 infection are still unclear. We systematically reviewed the previously reported cases of fulminant myocarditis associated with SARS-CoV-2 infection from January 2020 to December 2022, identifying 108 cases. Of those, 67 were male and 41 female. The average age was 34.8 years; 30 patients (27.8%) were ≤20 years old, whereas 10 (9.3%) were ≥60. Major comorbidities included hypertension, obesity, diabetes mellitus, asthma, heart disease, gynecologic disease, hyperlipidemia, and connective tissue disorders. Regarding left ventricular ejection fraction (LVEF) at admission, 93% of the patients with fulminant myocarditis were classified as having heart failure with reduced ejection fraction (LVEF ≤ 40%). Most of the cases were administered catecholamines (97.8%), and mechanical circulatory support (MCS) was required in 67 cases (62.0%). The type of MCS was extracorporeal membrane oxygenation (n = 56, 83.6%), percutaneous ventricular assist device (Impella®) (n = 19, 28.4%), intra-aortic balloon pumping (n = 12, 12.9%), or right ventricular assist device (n = 2, 3.0%); combination of these devices occurred in 20 cases (29.9%). The average duration of MCS was 7.7 ± 3.8 days. Of the 76 surviving patients whose cardiac function was available for follow-up, 65 (85.5%) recovered normally. The overall mortality rate was 22.4%, and the recovery rate was 77.6% (alive: 83 patients, dead: 24 patients; outcome not described: 1 patient).
Collapse
Affiliation(s)
- Ryohei Ono
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Togo Iwahana
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Kaoruko Aoki
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Hirotoshi Kato
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Sho Okada
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Yoshio Kobayashi
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| |
Collapse
|
13
|
Tandon P, Abrams ND, Avula LR, Carrick DM, Chander P, Divi RL, Dwyer JT, Gannot G, Gordiyenko N, Liu Q, Moon K, PrabhuDas M, Singh A, Tilahun ME, Satyamitra MM, Wang C, Warren R, Liu CH. Unraveling Links between Chronic Inflammation and Long COVID: Workshop Report. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:505-512. [PMID: 38315950 DOI: 10.4049/jimmunol.2300804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 12/12/2023] [Indexed: 02/07/2024]
Abstract
As COVID-19 continues, an increasing number of patients develop long COVID symptoms varying in severity that last for weeks, months, or longer. Symptoms commonly include lingering loss of smell and taste, hearing loss, extreme fatigue, and "brain fog." Still, persistent cardiovascular and respiratory problems, muscle weakness, and neurologic issues have also been documented. A major problem is the lack of clear guidelines for diagnosing long COVID. Although some studies suggest that long COVID is due to prolonged inflammation after SARS-CoV-2 infection, the underlying mechanisms remain unclear. The broad range of COVID-19's bodily effects and responses after initial viral infection are also poorly understood. This workshop brought together multidisciplinary experts to showcase and discuss the latest research on long COVID and chronic inflammation that might be associated with the persistent sequelae following COVID-19 infection.
Collapse
Affiliation(s)
- Pushpa Tandon
- National Cancer Institute, National Institutes of Health, Rockville, MD
| | - Natalie D Abrams
- National Cancer Institute, National Institutes of Health, Rockville, MD
| | - Leela Rani Avula
- National Cancer Institute, National Institutes of Health, Rockville, MD
| | | | - Preethi Chander
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD
| | - Rao L Divi
- National Cancer Institute, National Institutes of Health, Rockville, MD
| | - Johanna T Dwyer
- Office of Dietary Supplements, National Institutes of Health, Bethesda, MD
| | - Gallya Gannot
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD
| | | | - Qian Liu
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Kyung Moon
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Mercy PrabhuDas
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Anju Singh
- National Cancer Institute, National Institutes of Health, Rockville, MD
| | - Mulualem E Tilahun
- National Institute on Aging, National Institutes of Health, Bethesda, MD
| | - Merriline M Satyamitra
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Chiayeng Wang
- National Cancer Institute, National Institutes of Health, Rockville, MD
| | - Ronald Warren
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Christina H Liu
- National Institute of General Medical Sciences, National Institutes of Health, Bethesda, MD
| |
Collapse
|
14
|
Troxel AB, Bind MAC, Flotte TJ, Cordon-Cardo C, Decker LA, Finn AV, Padera RF, Reichard RR, Stone JR, Adolphi NL, Casimero FVC, Crary JF, Elifritz J, Faustin A, Ghosh SKB, Krausert A, Martinez-Lage M, Melamed J, Mitchell RA, Sampson BA, Seifert AC, Simsir A, Adams C, Haasnoot S, Hafner S, Siciliano MA, Vallejos BB, Del Boccio P, Lamendola-Essel MF, Young CE, Kewlani D, Akinbo PA, Parent B, Chung A, Cato TC, Mudumbi PC, Esquenazi-Karonika S, Wood MJ, Chan J, Monteiro J, Shinnick DJ, Thaweethai T, Nguyen AN, Fitzgerald ML, Perlowski AA, Stiles LE, Paskett ML, Katz SD, Foulkes AS, on behalf of the RECOVER Initiative Autopsy Group. Researching COVID to enhance recovery (RECOVER) tissue pathology study protocol: Rationale, objectives, and design. PLoS One 2024; 19:e0285645. [PMID: 38198481 PMCID: PMC10781091 DOI: 10.1371/journal.pone.0285645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 11/23/2023] [Indexed: 01/12/2024] Open
Abstract
IMPORTANCE SARS-CoV-2 infection can result in ongoing, relapsing, or new symptoms or organ dysfunction after the acute phase of infection, termed Post-Acute Sequelae of SARS-CoV-2 (PASC), or long COVID. The characteristics, prevalence, trajectory and mechanisms of PASC are poorly understood. The objectives of the Researching COVID to Enhance Recovery (RECOVER) tissue pathology study (RECOVER-Pathology) are to: (1) characterize prevalence and types of organ injury/disease and pathology occurring with PASC; (2) characterize the association of pathologic findings with clinical and other characteristics; (3) define the pathophysiology and mechanisms of PASC, and possible mediation via viral persistence; and (4) establish a post-mortem tissue biobank and post-mortem brain imaging biorepository. METHODS RECOVER-Pathology is a cross-sectional study of decedents dying at least 15 days following initial SARS-CoV-2 infection. Eligible decedents must meet WHO criteria for suspected, probable, or confirmed infection and must be aged 18 years or more at the time of death. Enrollment occurs at 7 sites in four U.S. states and Washington, DC. Comprehensive autopsies are conducted according to a standardized protocol within 24 hours of death; tissue samples are sent to the PASC Biorepository for later analyses. Data on clinical history are collected from the medical records and/or next of kin. The primary study outcomes include an array of pathologic features organized by organ system. Causal inference methods will be employed to investigate associations between risk factors and pathologic outcomes. DISCUSSION RECOVER-Pathology is the largest autopsy study addressing PASC among US adults. Results of this study are intended to elucidate mechanisms of organ injury and disease and enhance our understanding of the pathophysiology of PASC.
Collapse
Affiliation(s)
- Andrea B. Troxel
- Department of Population Health, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Marie-Abele C. Bind
- Department of Biostatistics, Massachusetts General Hospital, Boston, MA, United States of America
| | - Thomas J. Flotte
- Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, Rochester, MN, United States of America
| | - Carlos Cordon-Cardo
- Department of Pathology, Molecular and Cell-Based Medicine, Mount Sinai Health System, New York, NY, United States of America
| | - Lauren A. Decker
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
| | - Aloke V. Finn
- Department of Pathology, CVPath Institute, Gaithersburg, MD, United States of America
| | - Robert F. Padera
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, United States of America
| | - R. Ross Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, Rochester, MN, United States of America
| | - James R. Stone
- Department of Pathology, Massachusetts General Hospital, Boston, MA, United States of America
| | - Natalie L. Adolphi
- Office of the Medical Investigator, University of New Mexico School of Medicine, Albuquerque, NM, United States of America
| | | | - John F. Crary
- Department of Pathology, Icahn School of Medicine at Mount Sinai Hospital, New York, NY, United States of America
| | - Jamie Elifritz
- Departments of Radiology and Pathology, University of New Mexico, Albuquerque, NM, United States of America
| | - Arline Faustin
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Saikat Kumar B. Ghosh
- Department of Molecular Biology and Genomics, CVPath Institute, Gaithersburg, MD, United States of America
| | - Amanda Krausert
- Department of Pathology, Molecular and Cell-Based Medicine, Mount Sinai Health System, New York, NY, United States of America
| | - Maria Martinez-Lage
- Department of Pathology, Massachusetts General Hospital, Boston, MA, United States of America
| | - Jonathan Melamed
- Department of Anatomical Pathology, NYU Langone Hospital—Long Island, Mineola, NY, United States of America
| | - Roger A. Mitchell
- Department of Pathology, Howard University College of Medicine, Washington DC, United States of America
| | - Barbara A. Sampson
- Department of Pathology, Molecular and Cell-Based Medicine, Mount Sinai Health System, New York, NY, United States of America
| | - Alan C. Seifert
- Biomedical Engineering and Imaging Institute, Department of Radiology, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Aylin Simsir
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Cheryle Adams
- Department of Pathology, Howard University College of Medicine, Washington DC, United States of America
| | - Stephanie Haasnoot
- Department of Pathology, Icahn School of Medicine at Mount Sinai Hospital, New York, NY, United States of America
| | - Stephanie Hafner
- Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, Rochester, MN, United States of America
| | - Michelle A. Siciliano
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, United States of America
| | - Brittany B. Vallejos
- Office of the Medical Investigators, Department of Research, University of New Mexico, Albuquerque, NM, United States of America
| | - Phoebe Del Boccio
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Michelle F. Lamendola-Essel
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Chloe E. Young
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Deepshikha Kewlani
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Precious A. Akinbo
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Brendan Parent
- Department of Population Health, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Alicia Chung
- Department of Population Health, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Teresa C. Cato
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Praveen C. Mudumbi
- Department of Population Health, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Shari Esquenazi-Karonika
- Department of Population Health, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Marion J. Wood
- Department of Population Health, New York University Grossman School of Medicine, New York, NY, United States of America
| | - James Chan
- Department of Biostatistics, Massachusetts General Hospital, Boston, MA, United States of America
| | - Jonathan Monteiro
- Department of Biostatistics, Massachusetts General Hospital, Boston, MA, United States of America
| | - Daniel J. Shinnick
- Department of Biostatistics, Massachusetts General Hospital, Boston, MA, United States of America
| | - Tanayott Thaweethai
- Department of Biostatistics, Massachusetts General Hospital, Boston, MA, United States of America
| | - Amber N. Nguyen
- Department of Biostatistics, Massachusetts General Hospital, Boston, MA, United States of America
| | - Megan L. Fitzgerald
- Patient-Led Research Collaborative on COVID-19, Washington DC, United States of America
| | | | - Lauren E. Stiles
- Department of Neurology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, United States of America
| | - Moira L. Paskett
- Department of Anatomical Pathology, NYU Langone Hospital—Long Island, Mineola, NY, United States of America
| | - Stuart D. Katz
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Andrea S. Foulkes
- Department of Biostatistics, Massachusetts General Hospital, Boston, MA, United States of America
| | | |
Collapse
|
15
|
Ashok D, Liu T, Criscione J, Prakash M, Kim B, Chow J, Craney M, Papanicolaou KN, Sidor A, Brian Foster D, Pekosz A, Villano J, Kim DH, O'Rourke B. Innate Immune Activation and Mitochondrial ROS Invoke Persistent Cardiac Conduction System Dysfunction after COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.05.574280. [PMID: 38260287 PMCID: PMC10802485 DOI: 10.1101/2024.01.05.574280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Background Cardiac risk rises during acute SARS-CoV-2 infection and in long COVID syndrome in humans, but the mechanisms behind COVID-19-linked arrhythmias are unknown. This study explores the acute and long term effects of SARS-CoV-2 on the cardiac conduction system (CCS) in a hamster model of COVID-19. Methods Radiotelemetry in conscious animals was used to non-invasively record electrocardiograms and subpleural pressures after intranasal SARS-CoV-2 infection. Cardiac cytokines, interferon-stimulated gene expression, and macrophage infiltration of the CCS, were assessed at 4 days and 4 weeks post-infection. A double-stranded RNA mimetic, polyinosinic:polycytidylic acid (PIC), was used in vivo and in vitro to activate viral pattern recognition receptors in the absence of SARS-CoV-2 infection. Results COVID-19 induced pronounced tachypnea and severe cardiac conduction system (CCS) dysfunction, spanning from bradycardia to persistent atrioventricular block, although no viral protein expression was detected in the heart. Arrhythmias developed rapidly, partially reversed, and then redeveloped after the pulmonary infection was resolved, indicating persistent CCS injury. Increased cardiac cytokines, interferon-stimulated gene expression, and macrophage remodeling in the CCS accompanied the electrophysiological abnormalities. Interestingly, the arrhythmia phenotype was reproduced by cardiac injection of PIC in the absence of virus, indicating that innate immune activation was sufficient to drive the response. PIC also strongly induced cytokine secretion and robust interferon signaling in hearts, human iPSC-derived cardiomyocytes (hiPSC-CMs), and engineered heart tissues, accompanied by alterations in electrical and Ca 2+ handling properties. Importantly, the pulmonary and cardiac effects of COVID-19 were blunted by in vivo inhibition of JAK/STAT signaling or by a mitochondrially-targeted antioxidant. Conclusions The findings indicate that long term dysfunction and immune cell remodeling of the CCS is induced by COVID-19, arising indirectly from oxidative stress and excessive activation of cardiac innate immune responses during infection, with implications for long COVID Syndrome.
Collapse
|
16
|
Shu H, Wen Z, Li N, Zhang Z, Ceesay BM, Peng Y, Zhou N, Wang DW. COVID-19 and Cardiovascular Diseases: From Cellular Mechanisms to Clinical Manifestations. Aging Dis 2023; 14:2071-2088. [PMID: 37199573 PMCID: PMC10676802 DOI: 10.14336/ad.2023.0314] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 03/14/2023] [Indexed: 05/19/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), quickly spread worldwide and led to over 581 million confirmed cases and over 6 million deaths as 1 August 2022. The binding of the viral surface spike protein to the human angiotensin-converting enzyme 2 (ACE2) receptor is the primary mechanism of SARS-CoV-2 infection. Not only highly expressed in the lung, ACE2 is also widely distributed in the heart, mainly in cardiomyocytes and pericytes. The strong association between COVID-19 and cardiovascular disease (CVD) has been demonstrated by increased clinical evidence. Preexisting CVD risk factors, including obesity, hypertension, and diabetes etc., increase susceptibility to COVID-19. In turn, COVID-19 exacerbates the progression of CVD, including myocardial damage, arrhythmia, acute myocarditis, heart failure, and thromboembolism. Moreover, cardiovascular risks post recovery and the vaccination-associated cardiovascular problems have become increasingly evident. To demonstrate the association between COVID-19 and CVD, this review detailly illustrated the impact of COVID-19 on different cells (cardiomyocytes, pericytes, endothelial cells, and fibroblasts) in myocardial tissue and provides an overview of the clinical manifestations of cardiovascular involvements in the pandemic. Finally, the issues related to myocardial injury post recovery, as well as vaccination-induced CVD, has also been emphasized.
Collapse
Affiliation(s)
- Hongyang Shu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan 430000, China.
| | - Zheng Wen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan 430000, China.
| | - Na Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan 430000, China.
| | - Zixuan Zhang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan 430000, China.
| | - Bala Musa Ceesay
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan 430000, China.
| | - Yizhong Peng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Ning Zhou
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan 430000, China.
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan 430000, China.
| |
Collapse
|
17
|
Wang W, Yang J, Kang P, Bai J, Feng X, Huang L, Zhang Y, Wu Y, Tang B, Wang H, Jiang J, Li M, Zhao B, Yang X. Direct infection of SARS-CoV-2 in human iPSC-derived 3D cardiac organoids recapitulates COVID-19 myocarditis. Virol Sin 2023; 38:971-974. [PMID: 37783340 PMCID: PMC10786654 DOI: 10.1016/j.virs.2023.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/27/2023] [Indexed: 10/04/2023] Open
Abstract
•Establishment of 3D cardiac organoids composed of cardiomyocyte and endothelial layers. •SARS-CoV-2 infection causes multi-lineage cardiac injuries. •Cardiovascular health should be greatly concerned in COVID-19 patients.
Collapse
Affiliation(s)
- Weijie Wang
- Department of Cardiovascular Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China; State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Jinxuan Yang
- Yunnan Key Laboratory of Biodiversity Information, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - Pinfang Kang
- Department of Cardiovascular Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China.
| | - Jinsong Bai
- Kunming Third People's Hospital, Kunming, 650041, China
| | - Xiaoli Feng
- Kunming National High-level Biosafety Research Center for Non-human Primates, Kunming Institute of Zoology, Chinese Academic of Sciences, Kunming, 650107, China
| | - Liuqi Huang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Yiming Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Yuechun Wu
- Yunnan Key Laboratory of Biodiversity Information, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - Bi Tang
- Department of Cardiovascular Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China
| | - Hongju Wang
- Department of Cardiovascular Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China
| | - JianJie Jiang
- Kunming Third People's Hospital, Kunming, 650041, China
| | - Minghua Li
- Kunming National High-level Biosafety Research Center for Non-human Primates, Kunming Institute of Zoology, Chinese Academic of Sciences, Kunming, 650107, China
| | - Bing Zhao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China; Institute of Organoid Technology, Kunming Medical University, Kunming, 650500, China.
| | - Xinglou Yang
- Yunnan Key Laboratory of Biodiversity Information, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China.
| |
Collapse
|
18
|
Ntchana A, Shrestha S, Pippin M. Cardiovascular Complications of COVID-19: A Scoping Review of Evidence. Cureus 2023; 15:e48275. [PMID: 38054135 PMCID: PMC10695704 DOI: 10.7759/cureus.48275] [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: 11/04/2023] [Indexed: 12/07/2023] Open
Abstract
This scoping review sought to identify the nature and extent of clinical evidence regarding the acute and long-term cardiovascular complications associated with COVID-19. Forty-nine studies published between 2020 and 2023 were selected for review. The studies were divided into two groups. The referential group included 22 studies. The second group of 27 studies was used for a detailed review to assess the strength of the evidence. The aggregate evidence indicates that the most common cardiac complications associated with COVID-19 include but are not limited to acute pericarditis, acute myocardial injury, acute myocarditis, various arrhythmias, microvascular angiopathy, left ventricular dysfunction, heart failure, acute cardiac injury, and acute coronary syndrome. Clinical and epidemiological implications of the findings are investigated, and future research recommendations are proposed.
Collapse
Affiliation(s)
- Armand Ntchana
- Family Medicine-Alexandria, Louisiana State University Health Sciences Center, Alexandria, USA
| | - Sanjay Shrestha
- Family Medicine-Alexandria, Louisiana State University Health Sciences Center, Alexandria, USA
| | - Micah Pippin
- Family Medicine-Alexandria, Louisiana State University Health Sciences Center, Alexandria, USA
| |
Collapse
|
19
|
Mitrofanova L, Makarov I, Goncharova E, Makarova T, Starshinova A, Kudlay D, Shlaykhto E. High Risk of Heart Tumors after COVID-19. Life (Basel) 2023; 13:2087. [PMID: 37895467 PMCID: PMC10608002 DOI: 10.3390/life13102087] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 10/12/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
An emergence of evidence suggests that severe COVID-19 is associated with an increased risk of developing breast and gastrointestinal cancers. The aim of this research was to assess the risk of heart tumors development in patients who have had COVID-19. METHODS A comparative analysis of 173 heart tumors was conducted between 2016 and 2023. Immunohistochemical examination with antibodies against spike SARS-CoV-2 was performed on 21 heart tumors: 10 myxomas operated before 2020 (the control group), four cardiac myxomas, one proliferating myxoma, three papillary fibroelastomas, two myxofibrosarcomas, one chondrosarcoma resected in 2022-2023. Immunohistochemical analysis with antibodies against CD34 and CD68 was also conducted on the same 11 Post-COVID period heart tumors. Immunofluorescent examination with a cocktail of antibodies against spike SARS-CoV-2/CD34 and spike SARS-CoV-2/CD68 was performed in 2 cases out of 11 (proliferating myxoma and classic myxoma). RESULTS A 1.5-fold increase in the number of heart tumors by 2023 was observed, with a statistically significant increase in the number of myxomas. There was no correlation with vaccination, and no significant differences were found between patients from 2016-2019 and 2021-2023 in terms of gender, age, and cardiac rhythm dis-orders. Morphological examination revealed the expression of spike SARS-CoV-2 in tumor cells, endothelial cells, and macrophages in 10 out of 11 heart tumors. CONCLUSION The detection of SARS-CoV-2 persistence in endothelium and macrophages as well as in tumor cells of benign and malignant cardiac neoplasms, the increase in the number of these tumors, especially cardiac myxomas, after the pandemic by 2023 may indicate a trend toward an increased risk of cardiac neoplasms in COVID-19 patients, which re-quires further research on this issue and a search for new evidence.
Collapse
Affiliation(s)
- Lubov Mitrofanova
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (L.M.); (I.M.); (E.G.); (T.M.); (E.S.)
| | - Igor Makarov
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (L.M.); (I.M.); (E.G.); (T.M.); (E.S.)
| | - Ekaterina Goncharova
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (L.M.); (I.M.); (E.G.); (T.M.); (E.S.)
| | - Taiana Makarova
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (L.M.); (I.M.); (E.G.); (T.M.); (E.S.)
| | - Anna Starshinova
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (L.M.); (I.M.); (E.G.); (T.M.); (E.S.)
| | - Dmitry Kudlay
- Department of Pharmacology, I.M. Sechenov First Moscow State Medical University, 119992 Moscow, Russia;
- Institute of Immunology, 115478 Moscow, Russia
| | - Evgeny Shlaykhto
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (L.M.); (I.M.); (E.G.); (T.M.); (E.S.)
| |
Collapse
|
20
|
Krauson AJ, Casimero FVC, Siddiquee Z, Stone JR. Duration of SARS-CoV-2 mRNA vaccine persistence and factors associated with cardiac involvement in recently vaccinated patients. NPJ Vaccines 2023; 8:141. [PMID: 37758751 PMCID: PMC10533894 DOI: 10.1038/s41541-023-00742-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
At the start of the COVID-19 pandemic, the BNT162b2 (BioNTech-Pfizer) and mRNA-1273 (Moderna) mRNA vaccines were expediently designed and mass produced. Both vaccines produce the full-length SARS-CoV-2 spike protein for gain of immunity and have greatly reduced mortality and morbidity from SARS-CoV-2 infection. The distribution and duration of SARS-CoV-2 mRNA vaccine persistence in human tissues is unclear. Here, we developed specific RT-qPCR-based assays to detect each mRNA vaccine and screened lymph nodes, liver, spleen, and myocardium from recently vaccinated deceased patients. Vaccine was detected in the axillary lymph nodes in the majority of patients dying within 30 days of vaccination, but not in patients dying more than 30 days from vaccination. Vaccine was not detected in the mediastinal lymph nodes, spleen, or liver. Vaccine was detected in the myocardium in a subset of patients vaccinated within 30 days of death. Cardiac ventricles in which vaccine was detected had healing myocardial injury at the time of vaccination and had more myocardial macrophages than the cardiac ventricles in which vaccine was not detected. These results suggest that SARS-CoV-2 mRNA vaccines routinely persist up to 30 days from vaccination and can be detected in the heart.
Collapse
Affiliation(s)
- Aram J Krauson
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Faye Victoria C Casimero
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Zakir Siddiquee
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - James R Stone
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA.
- Department of Pathology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
21
|
Tucci AR, da Rosa RM, Rosa AS, Augusto Chaves O, Ferreira VNS, Oliveira TKF, Coutinho Souza DD, Borba NRR, Dornelles L, Rocha NS, Mayer JCP, da Rocha JBT, Rodrigues OED, Miranda MD. Antiviral Effect of 5'-Arylchalcogeno-3-aminothymidine Derivatives in SARS-CoV-2 Infection. Molecules 2023; 28:6696. [PMID: 37764472 PMCID: PMC10537738 DOI: 10.3390/molecules28186696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/04/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
The understanding that zidovudine (ZDV or azidothymidine, AZT) inhibits the RNA-dependent RNA polymerase (RdRp) of SARS-CoV-2 and that chalcogen atoms can increase the bioactivity and reduce the toxicity of AZT has directed our search for the discovery of novel potential anti-coronavirus compounds. Here, the antiviral activity of selenium and tellurium containing AZT derivatives in human type II pneumocytes cell model (Calu-3) and monkey kidney cells (Vero E6) infected with SARS-CoV-2, and their toxic effects on these cells, was evaluated. Cell viability analysis revealed that organoselenium (R3a-R3e) showed lower cytotoxicity than organotellurium (R3f, R3n-R3q), with CC50 ≥ 100 µM. The R3b and R3e were particularly noteworthy for inhibiting viral replication in both cell models and showed better selectivity index. In Vero E6, the EC50 values for R3b and R3e were 2.97 ± 0.62 µM and 1.99 ± 0.42 µM, respectively, while in Calu-3, concentrations of 3.82 ± 1.42 µM and 1.92 ± 0.43 µM (24 h treatment) and 1.33 ± 0.35 µM and 2.31 ± 0.54 µM (48 h) were observed, respectively. The molecular docking calculations were carried out to main protease (Mpro), papain-like protease (PLpro), and RdRp following non-competitive, competitive, and allosteric inhibitory approaches. The in silico results suggested that the organoselenium is a potential non-competitive inhibitor of RdRp, interacting in the allosteric cavity located in the palm region. Overall, the cell-based results indicated that the chalcogen-zidovudine derivatives were more potent than AZT in inhibiting SARS-CoV-2 replication and that the compounds R3b and R3e play an important inhibitory role, expanding the knowledge about the promising therapeutic capacity of organoselenium against COVID-19.
Collapse
Affiliation(s)
- Amanda Resende Tucci
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| | - Raquel Mello da Rosa
- LabSelen-NanoBio—Departamento de Química, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (R.M.d.R.); (L.D.); (N.S.R.); (J.C.P.M.)
| | - Alice Santos Rosa
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| | - Otávio Augusto Chaves
- CQC-IMS, Departamento de Química, Universidade de Coimbra, Rua Larga, 3004-535 Coimbra, Portugal
- Laboratório de Imunofarmacologia, Centro de Pesquisa, Inovação e Vigilância em COVID-19 e Emergências Sanitárias (CPIV), Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro 21040-900, RJ, Brazil
| | - Vivian Neuza Santos Ferreira
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
| | - Thamara Kelcya Fonseca Oliveira
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| | - Daniel Dias Coutinho Souza
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| | - Nathalia Roberto Resende Borba
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
| | - Luciano Dornelles
- LabSelen-NanoBio—Departamento de Química, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (R.M.d.R.); (L.D.); (N.S.R.); (J.C.P.M.)
| | - Nayra Salazar Rocha
- LabSelen-NanoBio—Departamento de Química, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (R.M.d.R.); (L.D.); (N.S.R.); (J.C.P.M.)
| | - João Candido Pilar Mayer
- LabSelen-NanoBio—Departamento de Química, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (R.M.d.R.); (L.D.); (N.S.R.); (J.C.P.M.)
| | - João B. Teixeira da Rocha
- Programa de Pós-Graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil;
| | - Oscar Endrigo D. Rodrigues
- LabSelen-NanoBio—Departamento de Química, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (R.M.d.R.); (L.D.); (N.S.R.); (J.C.P.M.)
| | - Milene Dias Miranda
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| |
Collapse
|
22
|
He W, Zhou L, Xu K, Li H, Wang JJ, Chen C, Wang D. Immunopathogenesis and immunomodulatory therapy for myocarditis. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2112-2137. [PMID: 37002488 PMCID: PMC10066028 DOI: 10.1007/s11427-022-2273-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 01/16/2023] [Indexed: 04/03/2023]
Abstract
Myocarditis is an inflammatory cardiac disease characterized by the destruction of myocardial cells, infiltration of interstitial inflammatory cells, and fibrosis, and is becoming a major public health concern. The aetiology of myocarditis continues to broaden as new pathogens and drugs emerge. The relationship between immune checkpoint inhibitors, severe acute respiratory syndrome coronavirus 2, vaccines against coronavirus disease-2019, and myocarditis has attracted increased attention. Immunopathological processes play an important role in the different phases of myocarditis, affecting disease occurrence, development, and prognosis. Excessive immune activation can induce severe myocardial injury and lead to fulminant myocarditis, whereas chronic inflammation can lead to cardiac remodelling and inflammatory dilated cardiomyopathy. The use of immunosuppressive treatments, particularly cytotoxic agents, for myocarditis, remains controversial. While reasonable and effective immunomodulatory therapy is the general trend. This review focuses on the current understanding of the aetiology and immunopathogenesis of myocarditis and offers new perspectives on immunomodulatory therapies.
Collapse
Affiliation(s)
- Wu He
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Ling Zhou
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Ke Xu
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Huihui Li
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - James Jiqi Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Chen Chen
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China.
| | - DaoWen Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China.
| |
Collapse
|
23
|
Li D, Zhao B, Zhuang P, Mei X. Development of nanozymes for promising alleviation of COVID-19-associated arthritis. Biomater Sci 2023; 11:5781-5796. [PMID: 37475700 DOI: 10.1039/d3bm00095h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has been identified as a culprit in the development of a variety of disorders, including arthritis. Although the emergence of arthritis following SARS-CoV-2 infection may not be immediately discernible, its underlying pathogenesis is likely to involve a complex interplay of infections, oxidative stress, immune responses, abnormal production of inflammatory factors, cellular destruction, etc. Fortunately, recent advancements in nanozymes with enzyme-like activities have shown potent antiviral effects and the ability to inhibit oxidative stress and cytokines and provide immunotherapeutic effects while also safeguarding diverse cell populations. These adaptable nanozymes have already exhibited efficacy in treating common types of arthritis, and their distinctive synergistic therapeutic effects offer great potential in the fight against arthritis associated with COVID-19. In this comprehensive review, we explore the potential of nanozymes in alleviating arthritis following SARS-CoV-2 infection by neutralizing the underlying factors associated with the disease. We also provide a detailed analysis of the common therapeutic pathways employed by these nanozymes and offer insights into how they can be further optimized to effectively address COVID-19-associated arthritis.
Collapse
Affiliation(s)
- Dan Li
- Department of Pharmacy, Jinzhou Medical University, Jinzhou, 121000, China.
| | - Baofeng Zhao
- Liaoning Provincial Key Laboratory of Medical Testing, Jinzhou Medical University, Jinzhou, 121001, China.
| | - Pengfei Zhuang
- Department of Pharmacy, Jinzhou Medical University, Jinzhou, 121000, China.
| | - Xifan Mei
- Liaoning Provincial Key Laboratory of Medical Testing, Jinzhou Medical University, Jinzhou, 121001, China.
| |
Collapse
|
24
|
Lu RXZ, Rafatian N, Zhao Y, Wagner KT, Beroncal EL, Li B, Lee C, Chen J, Churcher E, Vosoughi D, Wang Y, Baker A, Trahtemberg U, Li B, Pierro A, Andreazza AC, Dos Santos CC, Radisic M. Heart-on-a-chip model of immune-induced cardiac dysfunction reveals the role of free mitochondrial DNA and therapeutic effects of endothelial exosomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.09.552495. [PMID: 37609237 PMCID: PMC10441383 DOI: 10.1101/2023.08.09.552495] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Cardiovascular disease continues to take more human lives than all cancer combined, prompting the need for improved research models and treatment options. Despite a significant progress in development of mature heart-on-a-chip models of fibrosis and cardiomyopathies starting from induced pluripotent stem cells (iPSCs), human cell-based models of myocardial inflammation are lacking. Here, we bioengineered a vascularized heart-on-a-chip system with circulating immune cells to model SARS-CoV-2-induced acute myocarditis. Briefly, we observed hallmarks of COVID-19-induced myocardial inflammation in the heart-on-a-chip model, as the presence of immune cells augmented the expression levels of proinflammatory cytokines, triggered progressive impairment of contractile function and altered intracellular calcium transient activities. An elevation of circulating cell-free mitochondrial DNA (ccf-mtDNA) was measured first in the in vitro heart-on-a-chip model and then validated in COVID-19 patients with low left ventricular ejection fraction (LVEF), demonstrating that mitochondrial damage is an important pathophysiological hallmark of inflammation induced cardiac dysfunction. Leveraging this platform in the context of SARS-CoV-2 induced myocardial inflammation, we established that administration of human umbilical vein-derived EVs effectively rescued the contractile deficit, normalized intracellular calcium handling, elevated the contraction force and reduced the ccf- mtDNA and chemokine release via TLR-NF-kB signaling axis.
Collapse
|
25
|
Wang L, Western D, Timsina J, Repaci C, Song WM, Norton J, Kohlfeld P, Budde J, Climer S, Butt OH, Jacobson D, Garvin M, Templeton AR, Campagna S, O’Halloran J, Presti R, Goss CW, Mudd PA, Ances BM, Zhang B, Sung YJ, Cruchaga C. Plasma proteomics of SARS-CoV-2 infection and severity reveals impact on Alzheimer's and coronary disease pathways. iScience 2023; 26:106408. [PMID: 36974157 PMCID: PMC10010831 DOI: 10.1016/j.isci.2023.106408] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 01/21/2023] [Accepted: 03/10/2023] [Indexed: 03/14/2023] Open
Abstract
Identification of proteins dysregulated by COVID-19 infection is critically important for better understanding of its pathophysiology, building prognostic models, and identifying new targets. Plasma proteomic profiling of 4,301 proteins was performed in two independent datasets and tested for the association for three COVID-19 outcomes (infection, ventilation, and death). We identified 1,449 proteins consistently associated in both datasets with any of these three outcomes. We subsequently created highly accurate models that distinctively predict infection, ventilation, and death. These proteins were enriched in specific biological processes including cytokine signaling, Alzheimer's disease, and coronary artery disease. Mendelian randomization and gene network analyses identified eight causal proteins and 141 highly connected hub proteins including 35 with known drug targets. Our findings provide distinctive prognostic biomarkers for two severe COVID-19 outcomes, reveal their relationship to Alzheimer's disease and coronary artery disease, and identify potential therapeutic targets for COVID-19 outcomes.
Collapse
Affiliation(s)
- Lihua Wang
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, MO, USA
| | - Daniel Western
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, MO, USA
| | - Jigyasha Timsina
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, MO, USA
| | - Charlie Repaci
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, MO, USA
| | - Won-Min Song
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joanne Norton
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, MO, USA
| | - Pat Kohlfeld
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, MO, USA
| | - John Budde
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, MO, USA
| | - Sharlee Climer
- Department of Computer Science, University of Missouri-St. Louis, St. Louis, MO, USA
| | - Omar H. Butt
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Daniel Jacobson
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Michael Garvin
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Alan R. Templeton
- Department of Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Shawn Campagna
- Department of Chemistry, University of Tennessee, Knoxville, TN, USA
| | - Jane O’Halloran
- Division of Infectious Diseases, Washington University School of Medicine, St Louis, MO, USA
| | - Rachel Presti
- Division of Infectious Diseases, Washington University School of Medicine, St Louis, MO, USA
| | - Charles W. Goss
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Philip A. Mudd
- Department of Emergency Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Beau M. Ances
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yun Ju Sung
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, MO, USA
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, MO, USA
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
26
|
Shu H, Zhao C, Wang DW. Understanding COVID-19-related myocarditis: pathophysiology, diagnosis, and treatment strategies. CARDIOLOGY PLUS 2023; 8:72-81. [PMID: 37539019 PMCID: PMC10364646 DOI: 10.1097/cp9.0000000000000046] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/27/2023] [Indexed: 08/05/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) disease has infected nearly 600 million people, resulting in > 6 million deaths, with many of them dying from cardiovascular diseases. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is caused by a combination of the virus surface spike protein and the human angiotensin-converting enzyme 2 (ACE2) receptor. In addition to being highly expressed in the lungs, ACE2 is widely distributed in the heart, mainly in myocardial cells and pericytes. Like other types of viruses, SARS-CoV-2 can cause myocarditis after infecting the myocardial tissue, which is attributed to the direct damage of the virus and uncontrolled inflammatory reactions. Patients with chest tightness, palpitation, abnormal electrocardiogram, and cardiac troponin elevation, should be suspected of myocarditis within 1-3 weeks of COVID-19 infection. When the hemodynamics change rapidly, fulminant myocarditis should be suspected. Cardiac ultrasound, myocardial biopsy, cytokine detection, cardiac magnetic resonance imaging, 18F-fluorodeoxyglucose positron emission tomography, and other examination methods can assist in the diagnosis. Although scientists and clinicians have made concerted efforts to seek treatment and prevention measures, there are no clear recommendations for the treatment of COVID-19-related myocarditis. For most cases of common myocarditis, general symptomatic and supportive treatments are used. For COVID-19-related fulminant myocarditis, it is emphasized to achieve "early identification, early diagnosis, early prediction, and early treatment" based on the "life support-based comprehensive treatment regimen." Mechanical circulatory support therapy can rest the heart, which is a cure for symptoms, and immune regulation therapy can control the inflammatory storms which is a cure for the disease. Furthermore, complications of COVID-19-related myocarditis, such as arrhythmia, thrombosis, and infection, should be actively treated. Herein, we summarized the incidence rate, manifestations, and diagnosis of COVID-19-related myocarditis and discussed in detail the treatment of COVID-19-related myocarditis, especially the treatment strategy of fulminant myocarditis.
Collapse
Affiliation(s)
- Hongyang Shu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Chunxia Zhao
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| |
Collapse
|
27
|
|
28
|
Abstract
PURPOSE OF REVIEW Cardiac consequences occur in both acute COVID-19 and post-acute sequelae of COVID-19 (PASC). Here, we highlight the current understanding about COVID-19 cardiac effects, based upon clinical, imaging, autopsy, and molecular studies. RECENT FINDINGS COVID-19 cardiac effects are heterogeneous. Multiple, concurrent cardiac histopathologic findings have been detected on autopsies of COVID-19 non-survivors. Microthrombi and cardiomyocyte necrosis are commonly detected. Macrophages often infiltrate the heart at high density but without fulfilling histologic criteria for myocarditis. The high prevalences of microthrombi and inflammatory infiltrates in fatal COVID-19 raise the concern that recovered COVID-19 patients may have similar but subclinical cardiac pathology. Molecular studies suggest that SARS-CoV-2 infection of cardiac pericytes, dysregulated immunothrombosis, and pro-inflammatory and anti-fibrinolytic responses underlie COVID-19 cardiac pathology. The extent and nature by which mild COVID-19 affects the heart is unknown. Imaging and epidemiologic studies of recovered COVID-19 patients suggest that even mild illness confers increased risks of cardiac inflammation, cardiovascular disorders, and cardiovascular death. The mechanistic details of COVID-19 cardiac pathophysiology remain under active investigation. The ongoing evolution of SARS-CoV-2 variants and vast numbers of recovered COVID-19 patients portend a burgeoning global cardiovascular disease burden. Our ability to prevent and treat cardiovascular disease in the future will likely depend on comprehensive understanding of COVID-19 cardiac pathophysiologic phenotypes.
Collapse
Affiliation(s)
- Lorenzo R. Sewanan
- Department of Medicine, Columbia University Irving Medical Center, New York, NY USA
| | - Kevin J. Clerkin
- Center for Advanced Cardiac Care, Division of Cardiology, Columbia University Irving Medical Center, New York, NY USA
| | | | - Emily J. Tsai
- Center for Advanced Cardiac Care, Division of Cardiology, Columbia University Irving Medical Center, New York, NY USA
| |
Collapse
|
29
|
Kulasinghe A, Liu N, Tan CW, Monkman J, Sinclair JE, Bhuva DD, Godbolt D, Pan L, Nam A, Sadeghirad H, Sato K, Bassi GL, O'Byrne K, Hartmann C, Dos Santos Miggiolaro AFR, Marques GL, Moura LZ, Richard D, Adams M, de Noronha L, Baena CP, Suen JY, Arora R, Belz GT, Short KR, Davis MJ, Guimaraes FSF, Fraser JF. Transcriptomic profiling of cardiac tissues from SARS-CoV-2 patients identifies DNA damage. Immunology 2023; 168:403-419. [PMID: 36107637 PMCID: PMC9537957 DOI: 10.1111/imm.13577] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 09/12/2022] [Indexed: 12/15/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is known to present with pulmonary and extra-pulmonary organ complications. In comparison with the 2009 pandemic (pH1N1), SARS-CoV-2 infection is likely to lead to more severe disease, with multi-organ effects, including cardiovascular disease. SARS-CoV-2 has been associated with acute and long-term cardiovascular disease, but the molecular changes that govern this remain unknown. In this study, we investigated the host transcriptome landscape of cardiac tissues collected at rapid autopsy from seven SARS-CoV-2, two pH1N1, and six control patients using targeted spatial transcriptomics approaches. Although SARS-CoV-2 was not detected in cardiac tissue, host transcriptomics showed upregulation of genes associated with DNA damage and repair, heat shock, and M1-like macrophage infiltration in the cardiac tissues of COVID-19 patients. The DNA damage present in the SARS-CoV-2 patient samples, were further confirmed by γ-H2Ax immunohistochemistry. In comparison, pH1N1 showed upregulation of interferon-stimulated genes, in particular interferon and complement pathways, when compared with COVID-19 patients. These data demonstrate the emergence of distinct transcriptomic profiles in cardiac tissues of SARS-CoV-2 and pH1N1 influenza infection supporting the need for a greater understanding of the effects on extra-pulmonary organs, including the cardiovascular system of COVID-19 patients, to delineate the immunopathobiology of SARS-CoV-2 infection, and long term impact on health.
Collapse
Affiliation(s)
- Arutha Kulasinghe
- Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Ning Liu
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Chin Wee Tan
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - James Monkman
- Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Jane E Sinclair
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Dharmesh D Bhuva
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - David Godbolt
- Pathology Queensland, The Prince Charles Hospital, Chermside, Queensland, Australia
| | - Liuliu Pan
- Nanostring Technologies, Inc, Seattle, Washington, USA
| | - Andy Nam
- Nanostring Technologies, Inc, Seattle, Washington, USA
| | - Habib Sadeghirad
- Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Kei Sato
- Critical Care Research Group, Faculty of Medicine, University of Queensland and The Prince Charles Hospital, Brisbane, Queensland, Australia
| | - Gianluigi Li Bassi
- Critical Care Research Group, Faculty of Medicine, University of Queensland and The Prince Charles Hospital, Brisbane, Queensland, Australia
| | - Ken O'Byrne
- The Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Camila Hartmann
- Pontifical Catholic University of Parana, Curitiba, Brazil
- Marcelino Champagnat Hospital, Curitiba, Brazil
| | | | - Gustavo Lenci Marques
- Pontifical Catholic University of Parana, Curitiba, Brazil
- Marcelino Champagnat Hospital, Curitiba, Brazil
| | - Lidia Zytynski Moura
- Pontifical Catholic University of Parana, Curitiba, Brazil
- Marcelino Champagnat Hospital, Curitiba, Brazil
| | - Derek Richard
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Mark Adams
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | | | - Cristina Pellegrino Baena
- Pontifical Catholic University of Parana, Curitiba, Brazil
- Marcelino Champagnat Hospital, Curitiba, Brazil
| | - Jacky Y Suen
- Critical Care Research Group, Faculty of Medicine, University of Queensland and The Prince Charles Hospital, Brisbane, Queensland, Australia
| | - Rakesh Arora
- Department of Surgery, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Gabrielle T Belz
- Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Kirsty R Short
- Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Melissa J Davis
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Clinical Pathology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
| | | | - John F Fraser
- Pathology Queensland, The Prince Charles Hospital, Chermside, Queensland, Australia
| |
Collapse
|
30
|
Colombo D, Del Nonno F, Marchioni L, Lalle E, Gallì P, Vaia F, Falasca L. Autopsies Revealed Pathological Features of COVID-19 in Unvaccinated vs. Vaccinated Patients. Biomedicines 2023; 11:biomedicines11020551. [PMID: 36831087 PMCID: PMC9953314 DOI: 10.3390/biomedicines11020551] [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: 01/28/2023] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 02/16/2023] Open
Abstract
Background: In Italy, by the end of 2021, a new pandemic wave led to increased hospitalizations and death, even in some vaccinated people. We aimed to investigate the death of COVID-19-vaccinated patients who acquired infection and developed severe disease, and to assess differences with fatal COVID-19 in unvaccinated subjects by studying the pathological events triggered by SARS-CoV-2. Methods: Detailed autoptic examination was performed on five fully vaccinated compared to five unvaccinated patients. Histopathological analysis focused on the lung and heart, the two major affected organs. Results: COVID-19 caused, or contributed to death, in all the unvaccinated cases. By contrast, in vaccinated group, pre-existing pathologies played a major role, and death was not COVID-19-related in four out of five patients. These patients did not show the histological features of SARS-CoV-2 lung damage. Diffuse inflammatory macrophages infiltration recently emerged as the main feature of COVID-19 cardiac injury. Interestingly, the most striking difference between the two groups was the absence of increased macrophage infiltration in the heart of vaccinated patients. Conclusions: Results of this study confirm the efficacy of anti-SARS-CoV-2 vaccination in protecting organs from injury and support the need to maintain an adequate immune response by booster dose administration.
Collapse
Affiliation(s)
- Daniele Colombo
- Pathology Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani”-IRCCS, 00149 Rome, Italy
| | - Franca Del Nonno
- Pathology Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani”-IRCCS, 00149 Rome, Italy
| | - Luisa Marchioni
- Clinical Department, National Institute for Infectious Diseases “Lazzaro Spallanzani”-IRCCS, 00149 Rome, Italy
| | - Eleonora Lalle
- Laboratory of Virology, National Institute for Infectious Diseases “Lazzaro Spallanzani”-IRCCS, 00149 Rome, Italy
| | - Paola Gallì
- Health Direction, National Institute for Infectious Diseases “Lazzaro Spallanzani”-IRCCS, 00149 Rome, Italy
| | - Francesco Vaia
- Health Direction, National Institute for Infectious Diseases “Lazzaro Spallanzani”-IRCCS, 00149 Rome, Italy
| | - Laura Falasca
- Laboratory of Electron Microscopy, National Institute for Infectious Diseases “Lazzaro Spallanzani”-IRCCS, 00149 Rome, Italy
- Correspondence:
| |
Collapse
|
31
|
Altman NL, Berning AA, Saxon CE, Adamek KE, Wagner JA, Slavov D, Quaife RA, Gill EA, Minobe WA, Jonas ER, Carroll IA, Huebler SP, Raines J, Messenger JC, Ambardekar AV, Mestroni L, Rosenberg RM, Rove J, Campbell TB, Bristow MR. Myocardial Injury and Altered Gene Expression Associated With SARS-CoV-2 Infection or mRNA Vaccination. JACC Basic Transl Sci 2023; 8:124-137. [PMID: 36281440 PMCID: PMC9581498 DOI: 10.1016/j.jacbts.2022.08.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/17/2022] [Accepted: 08/17/2022] [Indexed: 11/07/2022]
Abstract
SARS CoV-2 enters host cells via its Spike protein moiety binding to the essential cardiac enzyme angiotensin-converting enzyme (ACE) 2, followed by internalization. COVID-19 mRNA vaccines are RNA sequences that are translated into Spike protein, which follows the same ACE2-binding route as the intact virion. In model systems, isolated Spike protein can produce cell damage and altered gene expression, and myocardial injury or myocarditis can occur during COVID-19 or after mRNA vaccination. We investigated 7 COVID-19 and 6 post-mRNA vaccination patients with myocardial injury and found nearly identical alterations in gene expression that would predispose to inflammation, coagulopathy, and myocardial dysfunction.
Collapse
Key Words
- ACE, angiotensin I–converting enzyme gene
- ACE2, angiotensin-converting enzyme 2 gene
- AGT, angiotensinogen gene
- AGTR1, angiotensin II receptor type 1 gene
- ANG II, angiotensin II
- BNP, B-type natriuretic peptide
- CMR, cardiac magnetic resonance
- COVID-19
- EM, electron microscopy
- F3, coagulation factor III (tissue factor) gene
- ITGA5, integrin subunit alpha 5 gene
- IVS, interventricular septum
- LGE, late gadolinium enhancement
- LM, light microscopy
- LV, left ventricular
- LVEF, left ventricular ejection fraction
- NDC, nonischemic dilated cardiomyopathy
- NPPB, natriuretic peptide B gene
- RV, right ventricular
- S, SARS-CoV-2 Spike
- TnI, troponin I
- gene expression
- mRNA vaccines
- myocardial injury
- myocarditis
Collapse
Affiliation(s)
- Natasha L. Altman
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Amber A. Berning
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Cara E. Saxon
- Department of Internal Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kylie E. Adamek
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jessica A. Wagner
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Dobromir Slavov
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Robert A. Quaife
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Edward A. Gill
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Wayne A. Minobe
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Eric R. Jonas
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | | | | | - Joshua Raines
- Department of Internal Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - John C. Messenger
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Amrut V. Ambardekar
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Luisa Mestroni
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Rachel M. Rosenberg
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jessica Rove
- Division of Cardiothoracic Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Thomas B. Campbell
- Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Michael R. Bristow
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- ARCA Biopharma, Westminster, Colorado, USA
| |
Collapse
|
32
|
Brumback BD, Dmytrenko O, Robinson AN, Bailey AL, Ma P, Liu J, Hicks SC, Ng S, Li G, Zhang DM, Lipovsky CE, Lin CY, Diamond MS, Lavine KJ, Rentschler SL. Human Cardiac Pericytes Are Susceptible to SARS-CoV-2 Infection. JACC Basic Transl Sci 2023; 8:109-120. [PMID: 36124009 PMCID: PMC9473702 DOI: 10.1016/j.jacbts.2022.09.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/06/2022] [Accepted: 09/06/2022] [Indexed: 11/10/2022]
Abstract
COVID-19 is associated with serious cardiovascular complications, with incompletely understood mechanism(s). Pericytes have key functions in supporting endothelial cells and maintaining vascular integrity. We demonstrate that human cardiac pericytes are permissive to SARS-CoV-2 infection in organotypic slice and primary cell cultures. Viral entry into pericytes is mediated by endosomal proteases, and infection leads to up-regulation of inflammatory markers, vasoactive mediators, and nuclear factor kappa-B-dependent cell death. Furthermore, we present evidence of cardiac pericyte infection in COVID-19 myocarditis patients. These data demonstrate that human cardiac pericytes are susceptible to SARS-CoV-2 infection and suggest a role for pericyte infection in COVID-19.
Collapse
Affiliation(s)
- Brittany D Brumback
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Oleksandr Dmytrenko
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Ashley N Robinson
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Adam L Bailey
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Pan Ma
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Jing Liu
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Stephanie C Hicks
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Sherwin Ng
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Gang Li
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - David M Zhang
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Catherine E Lipovsky
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Developmental Biology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Chieh-Yu Lin
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michael S Diamond
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Medicine, Infectious Disease, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Kory J Lavine
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Developmental Biology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Stacey L Rentschler
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Developmental Biology, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
33
|
Mitochondria Dysfunction at the Heart of Viral Myocarditis: Mechanistic Insights and Therapeutic Implications. Viruses 2023; 15:v15020351. [PMID: 36851568 PMCID: PMC9963085 DOI: 10.3390/v15020351] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
The myocardium/heart is the most mitochondria-rich tissue in the human body with mitochondria comprising approximately 30% of total cardiomyocyte volume. As the resident "powerhouse" of cells, mitochondria help to fuel the high energy demands of a continuously beating myocardium. It is no surprise that mitochondrial dysfunction underscores the pathogenesis of many cardiovascular ailments, including those of viral origin such as virus-induced myocarditis. Enteroviruses have been especially linked to injuries of the myocardium and its sequelae dilated cardiomyopathy for which no effective therapies currently exist. Intriguingly, recent mechanistic insights have demonstrated viral infections to directly damage mitochondria, impair the mitochondrial quality control processes of the cell, such as disrupting mitochondrial antiviral innate immune signaling, and promoting mitochondrial-dependent pathological inflammation of the infected myocardium. In this review, we briefly highlight recent insights on the virus-mitochondria crosstalk and discuss the therapeutic implications of targeting mitochondria to preserve heart function and ultimately combat viral myocarditis.
Collapse
|
34
|
Nishiyama K, Morikawa K, Shinozaki Y, Ueno J, Tanaka S, Tsuruoka H, Azagami S, Ishida A, Yanagisawa N, Akashi YJ, Mineshita M. Case report: Electrocardiographic changes in pembrolizumab-induced fatal myocarditis. Front Immunol 2023; 14:1078838. [PMID: 36875060 PMCID: PMC9980342 DOI: 10.3389/fimmu.2023.1078838] [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: 10/24/2022] [Accepted: 02/06/2023] [Indexed: 02/18/2023] Open
Abstract
Immune checkpoint inhibitor (ICI)-induced myocarditis is rare but fatal. Because of the rapid course of ICI-induced myocarditis, understanding of clinical course is only possible through information from case reports. We report a case of pembrolizumab-induced myocarditis in which we were able to document the course of electrocardiographic changes from onset to death. A 58-year-old woman with stage IV lung adenocarcinoma, who had completed her first cycle of pembrolizumab, carboplatin, and pemetrexed, was admitted with pericardial effusion. She underwent pericardiocentesis after admission. A second cycle of chemotherapy was administered 3 weeks after the first cycle. Twenty-two days after admission, she developed a mild sore throat and tested positive for SARS-CoV-2 antigen. She was diagnosed with mild coronavirus disease 2019 (COVID-19), isolated, and treated with sotrovimab. Thirty-two days after admission, an electrocardiogram showed monomorphic ventricular tachycardia (VT). Suspecting myocarditis caused by pembrolizumab, the patient was started on daily methylprednisolone after coronary angiography and endocardial biopsy. Eight days after the start of methylprednisolone administration, she was considered to have passed the acute stage. However, four days later, R-on-T phenomenon triggered polymorphic VT and she died. The impact of viral infections such as COVID-19 on patients be treated with immune checkpoint inhibitors is still unknown and we need to be careful with systemic management after viral infections.
Collapse
Affiliation(s)
- Kazuhiro Nishiyama
- Division of Respiratory Medicine, Department of Internal Medicine, St Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| | - Kei Morikawa
- Division of Respiratory Medicine, Department of Internal Medicine, St Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| | - Yusuke Shinozaki
- Division of Respiratory Medicine, Department of Internal Medicine, St Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| | - Junko Ueno
- Division of Respiratory Medicine, Department of Internal Medicine, St Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| | - Satoshi Tanaka
- Division of Respiratory Medicine, Department of Internal Medicine, St Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| | - Hajime Tsuruoka
- Division of Respiratory Medicine, Department of Internal Medicine, St Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| | - Shinya Azagami
- Division of Respiratory Medicine, Department of Internal Medicine, St Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| | - Atsuko Ishida
- Division of Respiratory Medicine, Department of Internal Medicine, St Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| | - Nobuyuki Yanagisawa
- Department of Pathology, St. Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| | - Yoshihiro J Akashi
- Division of Cardiology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| | - Masamichi Mineshita
- Division of Respiratory Medicine, Department of Internal Medicine, St Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| |
Collapse
|
35
|
Clemens DJ, Ye D, Zhou W, Kim CSJ, Pease DR, Navaratnarajah CK, Barkhymer A, Tester DJ, Nelson TJ, Cattaneo R, Schneider JW, Ackerman MJ. SARS-CoV-2 spike protein-mediated cardiomyocyte fusion may contribute to increased arrhythmic risk in COVID-19. PLoS One 2023; 18:e0282151. [PMID: 36888581 PMCID: PMC9994677 DOI: 10.1371/journal.pone.0282151] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 02/07/2023] [Indexed: 03/09/2023] Open
Abstract
BACKGROUND SARS-CoV-2-mediated COVID-19 may cause sudden cardiac death (SCD). Factors contributing to this increased risk of potentially fatal arrhythmias include thrombosis, exaggerated immune response, and treatment with QT-prolonging drugs. However, the intrinsic arrhythmic potential of direct SARS-CoV-2 infection of the heart remains unknown. OBJECTIVE To assess the cellular and electrophysiological effects of direct SARS-CoV-2 infection of the heart using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). METHODS hiPSC-CMs were transfected with recombinant SARS-CoV-2 spike protein (CoV-2 S) or CoV-2 S fused to a modified Emerald fluorescence protein (CoV-2 S-mEm). Cell morphology was visualized using immunofluorescence microscopy. Action potential duration (APD) and cellular arrhythmias were measured by whole cell patch-clamp. Calcium handling was assessed using the Fluo-4 Ca2+ indicator. RESULTS Transfection of hiPSC-CMs with CoV-2 S-mEm produced multinucleated giant cells (syncytia) displaying increased cellular capacitance (75±7 pF, n = 10 vs. 26±3 pF, n = 10; P<0.0001) consistent with increased cell size. The APD90 was prolonged significantly from 419±26 ms (n = 10) in untransfected hiPSC-CMs to 590±67 ms (n = 10; P<0.05) in CoV-2 S-mEm-transfected hiPSC-CMs. CoV-2 S-induced syncytia displayed delayed afterdepolarizations, erratic beating frequency, and calcium handling abnormalities including calcium sparks, large "tsunami"-like waves, and increased calcium transient amplitude. After furin protease inhibitor treatment or mutating the CoV-2 S furin cleavage site, cell-cell fusion was no longer evident and Ca2+ handling returned to normal. CONCLUSION The SARS-CoV-2 spike protein can directly perturb both the cardiomyocyte's repolarization reserve and intracellular calcium handling that may confer the intrinsic, mechanistic substrate for the increased risk of SCD observed during this COVID-19 pandemic.
Collapse
Affiliation(s)
- Daniel J. Clemens
- Department of Molecular Pharmacology & Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN, United States of America
| | - Dan Ye
- Department of Molecular Pharmacology & Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN, United States of America
- Division of Heart Rhythm Services, Department of Cardiovascular Medicine, Windland Smith Rice Genetic Heart Rhythm Clinic, Mayo Clinic, Rochester, MN, United States of America
| | - Wei Zhou
- Department of Molecular Pharmacology & Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN, United States of America
- Division of Heart Rhythm Services, Department of Cardiovascular Medicine, Windland Smith Rice Genetic Heart Rhythm Clinic, Mayo Clinic, Rochester, MN, United States of America
| | - C. S. John Kim
- Department of Molecular Pharmacology & Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN, United States of America
- Division of Heart Rhythm Services, Department of Cardiovascular Medicine, Windland Smith Rice Genetic Heart Rhythm Clinic, Mayo Clinic, Rochester, MN, United States of America
| | - David R. Pease
- Discovery Engine/Program for Hypoplastic Left Heart Syndrome, Mayo Clinic, Rochester, MN, United States of America
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, United States of America
| | | | - Alison Barkhymer
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - David J. Tester
- Department of Molecular Pharmacology & Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN, United States of America
- Division of Heart Rhythm Services, Department of Cardiovascular Medicine, Windland Smith Rice Genetic Heart Rhythm Clinic, Mayo Clinic, Rochester, MN, United States of America
| | - Timothy J. Nelson
- Discovery Engine/Program for Hypoplastic Left Heart Syndrome, Mayo Clinic, Rochester, MN, United States of America
- Wanek Family Program for HLHS-Stem Cell Pipeline, Mayo Clinic, Rochester, MN, United States of America
- Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Roberto Cattaneo
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Jay W. Schneider
- Discovery Engine/Program for Hypoplastic Left Heart Syndrome, Mayo Clinic, Rochester, MN, United States of America
| | - Michael J. Ackerman
- Department of Molecular Pharmacology & Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN, United States of America
- Division of Heart Rhythm Services, Department of Cardiovascular Medicine, Windland Smith Rice Genetic Heart Rhythm Clinic, Mayo Clinic, Rochester, MN, United States of America
- Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, United States of America
- * E-mail:
| |
Collapse
|
36
|
Razaghi A, Szakos A, Al-Shakarji R, Björnstedt M, Szekely L. Morphological changes without histological myocarditis in hearts of COVID-19 deceased patients. Scand Cardiovasc J Suppl 2022; 56:166-173. [PMID: 35678649 DOI: 10.1080/14017431.2022.2085320] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Objective. Patients with underlying heart diseases have a higher risk of dying from Covid-19. It has also been suggested that Covid-19 affects the heart through myocarditis. Despite the rapidly growing research on the management of Covid-19 associated complications, most of the ongoing research is focused on the respiratory complications of Covid-19, and little is known about the prevalence of myocarditis. Design. This study aimed to characterize myocardial involvement by using a panel of antibodies to detect hypoxic and inflammatory changes and the presence of SARS-CoV-2 proteins in heart tissues obtained during the autopsy procedure of Covid-19 deceased patients. Thirty-seven fatal COVID-19 cases and 21 controls were included in this study. Results. Overall, the Covid-19 hearts had several histopathological changes like the waviness of myocytes, fibrosis, contract band necrosis, infiltration of polymorphonuclear neutrophils, vacuolization, and necrosis of myocytes. In addition, endothelial damage and activation were detected in heart tissue. However, viral replication was not detected using RNA in situ hybridization. Also, lymphocyte infiltration, as a hallmark of myocarditis, was not seen in this study. Conclusion. No histological sign of myocarditis was detected in any of our cases; our findings are thus most congruent with the hypothesis of the presence of a circulating endothelium activating factor such as VEGF, originating outside of the heart, probably from the hypoxic part of the Covid-19 lungs.
Collapse
Affiliation(s)
- Ali Razaghi
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Attila Szakos
- Laboratory of Clinical Pathology and Cytology, Karolinska University Hospital, Stockholm, Sweden
| | - Riham Al-Shakarji
- Laboratory of Clinical Pathology and Cytology, Karolinska University Hospital, Stockholm, Sweden
| | - Mikael Björnstedt
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.,Laboratory of Clinical Pathology and Cytology, Karolinska University Hospital, Stockholm, Sweden
| | - Laszlo Szekely
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.,Laboratory of Clinical Pathology and Cytology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
37
|
Zhang Q, Zhang H, Yan X, Ma S, Yao X, Shi Y, Ping Y, Cao M, Peng C, Wang S, Luo M, Yan C, Zhang S, Han Y, Bian X. Neutrophil infiltration and myocarditis in patients with severe COVID-19: A post-mortem study. Front Cardiovasc Med 2022; 9:1026866. [PMID: 36312241 PMCID: PMC9614157 DOI: 10.3389/fcvm.2022.1026866] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 09/26/2022] [Indexed: 12/15/2022] Open
Abstract
Aims To investigate cardiac pathology in critically ill patients with coronavirus disease 2019 (COVID-19) and identify associations between pathological changes and clinical characteristics. Methods The present autopsy cohort study included hearts from 26 deceased patients hospitalized in intensive care units due to COVID-19, and was conducted at four sites in Wuhan, China. Cases were divided into a neutrophil infiltration group and a no-neutrophil group based on the presence or absence of histopathologically identified neutrophilic infiltrates. Results Among the 26 patients, histopathological examination identified active myocarditis in four patients. All patients with myocarditis exhibited extensive accompanying neutrophil infiltration, and all patients without myocarditis did not. The neutrophil infiltration group exhibited significantly higher rates of detection of interleukin-6 (100 vs. 4.6%) and tumor necrosis factor-alpha (100 vs. 31.8%) than the no-neutrophil group (both p < 0.05). On admission, four patients with neutrophil infiltration in myocardium had significantly higher baseline levels of aspartate aminotransferase, D dimer, and high-sensitivity C reactive protein than the other 22 patients (all p < 0.05). During hospitalization, patients with neutrophil infiltration had significantly higher maximum creatine kinase-MB (median 280.0 IU/L vs. 38.7 IU/L, p = 0.04) and higher troponin I (median 1.112 ng/ml vs. 0.220 ng/ml, p = 0.56) than patients without neutrophil infiltration. Conclusion Active myocarditis was frequently associated with neutrophil infiltration in the hearts of deceased patients with severe COVID-19. Patients with neutrophil-infiltrated myocarditis had a series of severely abnormal laboratory test results on admission, and high maximum creatine kinase-MB during hospitalization. The role of neutrophils in severe heart injury and systemic conditions in patients with COVID-19 should be emphasized.
Collapse
Affiliation(s)
- Quanyu Zhang
- Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Huarong Zhang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Beijing, China
| | - Xiaowei Yan
- Division of Cardiology, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Sicong Ma
- Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Xiaohong Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Beijing, China
| | - Yu Shi
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Beijing, China
| | - Yifang Ping
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Beijing, China
| | - Mianfu Cao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Beijing, China
| | - Chengfei Peng
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Beijing, China
| | - Shuai Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Beijing, China
| | - Min Luo
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Beijing, China
| | - Chenghui Yan
- Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Shuyang Zhang
- Division of Cardiology, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Yaling Han
- Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Xiuwu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Beijing, China
| |
Collapse
|
38
|
Meeting the Challenges of Myocarditis: New Opportunities for Prevention, Detection, and Intervention—A Report from the 2021 National Heart, Lung, and Blood Institute Workshop. J Clin Med 2022; 11:jcm11195721. [PMID: 36233593 PMCID: PMC9571285 DOI: 10.3390/jcm11195721] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/21/2022] [Accepted: 09/23/2022] [Indexed: 12/05/2022] Open
Abstract
The National Heart, Lung, and Blood Institute (NHLBI) convened a workshop of international experts to discuss new research opportunities for the prevention, detection, and intervention of myocarditis in May 2021. These experts reviewed the current state of science and identified key gaps and opportunities in basic, diagnostic, translational, and therapeutic frontiers to guide future research in myocarditis. In addition to addressing community-acquired myocarditis, the workshop also focused on emerging causes of myocarditis including immune checkpoint inhibitors and SARS-CoV-2 related myocardial injuries and considered the use of systems biology and artificial intelligence methodologies to define workflows to identify novel mechanisms of disease and new therapeutic targets. A new priority is the investigation of the relationship between social determinants of health (SDoH), including race and economic status, and inflammatory response and outcomes in myocarditis. The result is a proposal for the reclassification of myocarditis that integrates the latest knowledge of immunological pathogenesis to refine estimates of prognosis and target pathway-specific treatments.
Collapse
|
39
|
Alam SR, Vinayak S, Shah A, Doolub G, Kimeu R, Horn KP, Bowen SR, Jeilan M, Lee KK, Gachoka S, Riunga F, Adam RD, Vesselle H, Joshi N, Obino M, Makhdomi K, Ombati K, Nganga E, Gitau S, Chung MH, Shah ASV. Assessment of Cardiac, Vascular, and Pulmonary Pathobiology In Vivo During Acute COVID‐19. J Am Heart Assoc 2022; 11:e026399. [DOI: 10.1161/jaha.122.026399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background
Acute COVID‐19–related myocardial, pulmonary, and vascular pathology and how these relate to each other remain unclear. To our knowledge, no studies have used complementary imaging techniques, including molecular imaging, to elucidate this. We used multimodality imaging and biochemical sampling in vivo to identify the pathobiology of acute COVID‐19. Specifically, we investigated the presence of myocardial inflammation and its association with coronary artery disease, systemic vasculitis, and pneumonitis.
Methods and Results
Consecutive patients presenting with acute COVID‐19 were prospectively recruited during hospital admission in this cross‐sectional study. Imaging involved computed tomography coronary angiography (identified coronary disease), cardiac 2‐deoxy‐2‐[fluorine‐18]fluoro‐D‐glucose positron emission tomography/computed tomography (identified vascular, cardiac, and pulmonary inflammatory cell infiltration), and cardiac magnetic resonance (identified myocardial disease) alongside biomarker sampling. Of 33 patients (median age 51 years, 94% men), 24 (73%) had respiratory symptoms, with the remainder having nonspecific viral symptoms. A total of 9 patients (35%, n=9/25) had cardiac magnetic resonance–defined myocarditis. Of these patients, 53% (n=5/8) had myocardial inflammatory cell infiltration. A total of 2 patients (5%) had elevated troponin levels. Cardiac troponin concentrations were not significantly higher in patients with and without myocarditis (8.4 ng/L [interquartile range, IQR: 4.0–55.3] versus 3.5 ng/L [IQR: 2.5–5.5];
P
=0.07) or myocardial cell infiltration (4.4 ng/L [IQR: 3.4–8.3] versus 3.5 ng/L [IQR: 2.8–7.2];
P
=0.89). No patients had obstructive coronary artery disease or vasculitis. Pulmonary inflammation and consolidation (percentage of total lung volume) was 17% (IQR: 5%–31%) and 11% (IQR: 7%–18%), respectively. Neither were associated with the presence of myocarditis.
Conclusions
Myocarditis was present in a third patients with acute COVID‐19, and the majority had inflammatory cell infiltration. Pneumonitis was ubiquitous, but this inflammation was not associated with myocarditis. The mechanism of cardiac pathology is nonischemic and not attributable to a vasculitic process.
Registration
URL:
https://www.isrctn.com
; Unique identifier: ISRCTN12154994.
Collapse
Affiliation(s)
- Shirjel R. Alam
- Department of Cardiology Manchester University Manchester United Kingdom
- Department of Cardiology North Bristol Trust Bristol United Kingdom
- Non‐communicable Disease Epidemiology London School of Hygiene and Tropical Medicine London United Kingdom
| | - Sudhir Vinayak
- Department of Radiology and Department of Medicine Aga Khan University Nairobi Kenya
| | - Adeel Shah
- Department of Radiology and Department of Medicine Aga Khan University Nairobi Kenya
| | - Gemina Doolub
- Department of Cardiology University of Bristol Bristol United Kingdom
| | - Redemptar Kimeu
- Department of Radiology and Department of Medicine Aga Khan University Nairobi Kenya
| | - Kevin P. Horn
- Department of Radiology University of Washington Seattle WA
| | | | - Mohamed Jeilan
- Department of Radiology and Department of Medicine Aga Khan University Nairobi Kenya
| | - Kuan Ken Lee
- Department of Cardiology University of Edinburgh Edinburgh UK
| | - Sylvia Gachoka
- Department of Radiology and Department of Medicine Aga Khan University Nairobi Kenya
| | - Felix Riunga
- Department of Radiology and Department of Medicine Aga Khan University Nairobi Kenya
| | - Rodney D. Adam
- Department of Radiology and Department of Medicine Aga Khan University Nairobi Kenya
| | | | - Nikhil Joshi
- Department of Cardiology University of Bristol Bristol United Kingdom
| | - Mariah Obino
- Department of Radiology and Department of Medicine Aga Khan University Nairobi Kenya
| | - Khalid Makhdomi
- Department of Radiology and Department of Medicine Aga Khan University Nairobi Kenya
| | - Kevin Ombati
- Department of Radiology and Department of Medicine Aga Khan University Nairobi Kenya
| | - Edward Nganga
- Department of Radiology and Department of Medicine Aga Khan University Nairobi Kenya
| | - Samuel Gitau
- Department of Radiology and Department of Medicine Aga Khan University Nairobi Kenya
| | - Michael H. Chung
- Division of Infectious Diseases of the Department of Medicine Emory University Atlanta Georgia
| | - Anoop S. V. Shah
- Department of Radiology and Department of Medicine Aga Khan University Nairobi Kenya
| |
Collapse
|
40
|
Zuin M, Rigatelli G, Bilato C, Imazio M, Roncon L. Autoptic prevalence of pericarditis in coronavirus disease 2019 patients. J Cardiovasc Med (Hagerstown) 2022; 23:623-625. [PMID: 35904993 PMCID: PMC9593321 DOI: 10.2459/jcm.0000000000001306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 03/17/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Marco Zuin
- Department of Translational Medicine, University of Ferrara, Ferrara
- Department of Cardiology, West Vincenza Hospitals, Arzignano
| | - Gianluca Rigatelli
- Department of Cardiology, Santa Maria della Misericordia Hospital, Rovigo
| | - Claudio Bilato
- Department of Cardiology, West Vincenza Hospitals, Arzignano
| | - Massimo Imazio
- Cardiothoracic department University Hospital Santa Maria della Misericordia, Udine, Italy
| | - Loris Roncon
- Department of Cardiology, Santa Maria della Misericordia Hospital, Rovigo
| |
Collapse
|
41
|
Bollano E, Polte CL, Mäyränpää MI, Oldfors A, Bergh N, Lehtonen J, Kandolin R. Cardiac sarcoidosis and giant cell myocarditis after COVID-19 infection. ESC Heart Fail 2022; 9:4298-4303. [PMID: 35996829 PMCID: PMC9539072 DOI: 10.1002/ehf2.14088] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 06/28/2022] [Accepted: 07/18/2022] [Indexed: 01/19/2023] Open
Abstract
Patients infected with SARS-CoV-2 have varying manifestations of cardiac involvement. We report four patients presenting with symptomatic cardiac sarcoidosis (CS) or giant cell myocarditis (GCM) 1-8 months after mild COVID-19. All patients received immunosuppressive therapy and improved gradually within the following months. The possible temporal association between the CS/GCM and COVID-19 infection might suggest that COVID-19 could be a trigger for granulomatous myocarditis.
Collapse
Affiliation(s)
- Entela Bollano
- Department of CardiologySahlgrenska University Hospital413 45GothenburgSweden,Institute of Medicine at Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Christian Lars Polte
- Institute of Medicine at Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden,Department of Clinical Physiology and RadiologySahlgrenska University HospitalGothenburgSweden
| | | | - Anders Oldfors
- Department of Clinical PathologySahlgrenska University HospitalGothenburgSweden
| | - Niklas Bergh
- Department of CardiologySahlgrenska University Hospital413 45GothenburgSweden,Institute of Medicine at Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Jukka Lehtonen
- Department of Cardiology, Heart and Lung CenterHelsinki University HospitalHelsinkiFinland
| | - Riina Kandolin
- Department of Cardiology, Heart and Lung CenterHelsinki University HospitalHelsinkiFinland
| |
Collapse
|
42
|
Almamlouk R, Kashour T, Obeidat S, Bois MC, Maleszewski JJ, Omrani OA, Tleyjeh R, Berbari E, Chakhachiro Z, Zein-Sabatto B, Gerberi D, Tleyjeh IM, Paniz Mondolfi AE, Finn AV, Duarte-Neto AN, Rapkiewicz AV, Frustaci A, Keresztesi AA, Hanley B, Märkl B, Lardi C, Bryce C, Lindner D, Aguiar D, Westermann D, Stroberg E, Duval EJ, Youd E, Bulfamante GP, Salmon I, Auer J, Maleszewski JJ, Hirschbühl K, Absil L, Barton LM, Ferraz da Silva LF, Moore L, Dolhnikoff M, Lammens M, Bois MC, Osborn M, Remmelink M, Nascimento Saldiva PH, Jorens PG, Craver R, Aparecida de Almeida Monteiro R, Scendoni R, Mukhopadhyay S, Suzuki T, Mauad T, Fracasso T, Grimes Z. COVID-19-Associated cardiac pathology at the postmortem evaluation: a collaborative systematic review. Clin Microbiol Infect 2022; 28:1066-1075. [PMID: 35339672 PMCID: PMC8941843 DOI: 10.1016/j.cmi.2022.03.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/10/2022] [Accepted: 03/14/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Many postmortem studies address the cardiovascular effects of COVID-19 and provide valuable information, but are limited by their small sample size. OBJECTIVES The aim of this systematic review is to better understand the various aspects of the cardiovascular complications of COVID-19 by pooling data from a large number of autopsy studies. DATA SOURCES We searched the online databases Ovid EBM Reviews, Ovid Embase, Ovid Medline, Scopus, and Web of Science for concepts of autopsy or histopathology combined with COVID-19, published between database inception and February 2021. We also searched for unpublished manuscripts using the medRxiv services operated by Cold Spring Harbor Laboratory. STUDY ELIGIBILITY CRITERIA Articles were considered eligible for inclusion if they reported human postmortem cardiovascular findings among individuals with a confirmed SARS coronavirus type 2 (CoV-2) infection. PARTICIPANTS Confirmed COVID-19 patients with post-mortem cardiovascular findings. INTERVENTIONS None. METHODS Studies were individually assessed for risk of selection, detection, and reporting biases. The median prevalence of different autopsy findings with associated interquartile ranges (IQRs). RESULTS This review cohort contained 50 studies including 548 hearts. The median age of the deceased was 69 years. The most prevalent acute cardiovascular findings were myocardial necrosis (median: 100.0%; IQR, 20%-100%; number of studies = 9; number of patients = 64) and myocardial oedema (median: 55.5%; IQR, 19.5%-92.5%; number of studies = 4; number of patients = 46). The median reported prevalence of extensive, focal active, and multifocal myocarditis were all 0.0%. The most prevalent chronic changes were myocyte hypertrophy (median: 69.0%; IQR, 46.8%-92.1%) and fibrosis (median: 35.0%; IQR, 35.0%-90.5%). SARS-CoV-2 was detected in the myocardium with median prevalence of 60.8% (IQR 40.4-95.6%). CONCLUSIONS Our systematic review confirmed the high prevalence of acute and chronic cardiac pathologies in COVID-19 and SARS-CoV-2 cardiac tropism, as well as the low prevalence of myocarditis in COVID-19.
Collapse
Affiliation(s)
| | - Tarek Kashour
- Department of Cardiac Sciences, King Fahad Cardiac Center, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Sawsan Obeidat
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Melanie C Bois
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Joseph J Maleszewski
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA; Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Osama A Omrani
- The Royal London Hospital, Barts Health NHS Trust, London, UK; Barts and the London School of Medicine and Dentistry, Queen Mary University, London, UK
| | - Rana Tleyjeh
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Elie Berbari
- Division of Infectious Diseases, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Zaher Chakhachiro
- Department of Pathology and Laboratory Medicine, American University of Beirut, Beirut, Lebanon
| | - Bassel Zein-Sabatto
- Department of Pathology and Laboratory Medicine, American University of Beirut, Beirut, Lebanon
| | - Dana Gerberi
- Mayo Clinic Libraries, Mayo Clinic, Rochester, MN, USA
| | - Imad M Tleyjeh
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia; Division of Infectious Diseases, Mayo Clinic College of Medicine and Science, Rochester, MN, USA; Infectious Diseases Section, Department of Medical Specialties King Fahad Medical City, Riyadh, Saudi Arabia; Division of Epidemiology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA.
| | | | - Aloke V Finn
- CVPath Institute, Inc., Gaithersburg, and University of Maryland, Baltimore, MD, USA
| | - Amaro Nunes Duarte-Neto
- Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Amy V Rapkiewicz
- NYU Winthrop Hospital, Department of Pathology, Long Island School of Medicine, Long Island, NY, USA
| | - Andrea Frustaci
- Department of Clinical, Internal, Anesthesiologist and Cardiovascular Sciences, La Sapienza University, Rome, Italy; Cellular and Molecular Cardiology Lab, IRCCS L. Spallanzani, Rome, Italy
| | - Arthur-Atilla Keresztesi
- Fogolyan Kristof Emergency County Hospital, Covasna County Institution of Forensic Medicine, Covasna, Romania
| | - Brian Hanley
- Department of Cellular Pathology, Northwest London Pathology, Imperial College London NHS Trust, London, UK; Centre for Inflammatory Disease, Imperial College London, London, UK
| | - Bruno Märkl
- Institute of Pathology and Molecular Diagnostics, University Medical Center Augsburg, Augsburg, Germany
| | - Christelle Lardi
- University Center of Legal Medicine, Geneva University Hospital, Geneva, Switzerland
| | - Clare Bryce
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Diana Lindner
- Department of Cardiology, University Heart and Vascular Centre, Hamburg, Germany; DZHK-German Center for Cardiovascular Research, Partner site, Hamburg/Kiel/Lübeck, Germany
| | - Diego Aguiar
- University Center of Legal Medicine, Geneva University Hospital, Geneva, Switzerland
| | - Dirk Westermann
- Department of Cardiology, University Heart and Vascular Centre, Hamburg, Germany; DZHK-German Center for Cardiovascular Research, Partner site, Hamburg/Kiel/Lübeck, Germany
| | - Edana Stroberg
- Office of the Chief Medical Examiner, Oklahoma City, OK, USA
| | - Eric J Duval
- Office of the Chief Medical Examiner, Oklahoma City, OK, USA
| | - Esther Youd
- Forensic Medicine and Science, University of Glasgow, Glasgow, UK
| | - Gaetano Pietro Bulfamante
- Unità di Anatomia Patologica, Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan, Italy; Struttura Complessa di Anatomia Patologica e Genetica Medica, ASST Santi Paolo e Carlo, Milan, Italy
| | - Isabelle Salmon
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium; Centre Universitaire inter Régional d'expertise en Anatomie Pathologique Hospitalière, Jumet, Belgium; DIAPath, Center for Microscopy and Molecular Imaging, ULB, Gosselies, Belgium
| | - Johann Auer
- Department of Cardiology and Intensive Care, St. Josef Hospital Braunau, Austria; Department of Cardiology and Intensive Care, Kepler University of Medicine Linz, Austria
| | - Joseph J Maleszewski
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA; Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Klaus Hirschbühl
- Department of Hematology and Clinical Oncology, University Medical Center Augsburg, Augsburg, Germany
| | - Lara Absil
- Department of Pathology, Erasme Hospital, ULB, Brussels, Belgium
| | - Lisa M Barton
- Office of the Chief Medical Examiner, Oklahoma City, OK, USA
| | - Luiz Fernando Ferraz da Silva
- Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; Serviço de Verificação de Óbitos da Capital, Universidade de São Paulo, São Paulo, Brazil
| | - Luiza Moore
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; Ageing and Somatic Mutation, Wellcome Sanger Institute, Cambridge, UK
| | - Marisa Dolhnikoff
- Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Martin Lammens
- Department of Pathology, Antwerp University Hospital, University of Antwerp, Edegem, Belgium
| | - Melanie C Bois
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Michael Osborn
- Department of Cellular Pathology, Northwest London Pathology, Imperial College London NHS Trust, London, UK; Death Investigation Committee, Royal College of Pathologists, London, UK; Nightingale NHS Hospital, London, UK
| | - Myriam Remmelink
- Department of Pathology, Erasme Hospital, ULB, Brussels, Belgium
| | | | - Philippe G Jorens
- Infla-Med Research Consortium of Excellence, University of Antwerp, Antwerp, Belgium; Department of Medicine and Health Sciences, Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium; Department of Intensive Care Medicine, Antwerp University Hospital, University of Antwerp, Edegem, Belgium
| | - Randall Craver
- Children's Hospital of New Orleans and Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | | | - Roberto Scendoni
- Institute of Legal Medicine, Department of Law, University of Macerata, Macerata, Italy
| | | | - Tadaki Suzuki
- National Institute of Infectious Diseases, Tokyo, Japan
| | - Thais Mauad
- Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Tony Fracasso
- University Center of Legal Medicine, Geneva University Hospital, Geneva, Switzerland
| | - Zachary Grimes
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
43
|
Wang L, Western D, Timsina J, Repaci C, Song WM, Norton J, Kohlfeld P, Budde J, Climer S, Butt OH, Jacobson D, Garvin M, Templeton AR, Campagna S, O’Halloran J, Presti R, Goss CW, Mudd PA, Ances BM, Zhang B, Sung YJ, Cruchaga C. Plasma proteomics of SARS-CoV-2 infection and severity reveals impact on Alzheimer and coronary disease pathways. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.07.25.22278025. [PMID: 35923315 PMCID: PMC9347279 DOI: 10.1101/2022.07.25.22278025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Identification of the plasma proteomic changes of Coronavirus disease 2019 (COVID-19) is essential to understanding the pathophysiology of the disease and developing predictive models and novel therapeutics. We performed plasma deep proteomic profiling from 332 COVID-19 patients and 150 controls and pursued replication in an independent cohort (297 cases and 76 controls) to find potential biomarkers and causal proteins for three COVID-19 outcomes (infection, ventilation, and death). We identified and replicated 1,449 proteins associated with any of the three outcomes (841 for infection, 833 for ventilation, and 253 for death) that can be query on a web portal ( https://covid.proteomics.wustl.edu/ ). Using those proteins and machine learning approached we created and validated specific prediction models for ventilation (AUC>0.91), death (AUC>0.95) and either outcome (AUC>0.80). These proteins were also enriched in specific biological processes, including immune and cytokine signaling (FDR ≤ 3.72×10 -14 ), Alzheimer's disease (FDR ≤ 5.46×10 -10 ) and coronary artery disease (FDR ≤ 4.64×10 -2 ). Mendelian randomization using pQTL as instrumental variants nominated BCAT2 and GOLM1 as a causal proteins for COVID-19. Causal gene network analyses identified 141 highly connected key proteins, of which 35 have known drug targets with FDA-approved compounds. Our findings provide distinctive prognostic biomarkers for two severe COVID-19 outcomes (ventilation and death), reveal their relationship to Alzheimer's disease and coronary artery disease, and identify potential therapeutic targets for COVID-19 outcomes.
Collapse
Affiliation(s)
- Lihua Wang
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, MO, USA
| | - Dan Western
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, MO, USA
| | - Jigyasha Timsina
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, MO, USA
| | - Charlie Repaci
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, MO, USA
| | - Won-Min Song
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Joanne Norton
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, MO, USA
| | - Pat Kohlfeld
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, MO, USA
| | - John Budde
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, MO, USA
| | - Sharlee Climer
- Department of Computer Science, University of Missouri-St. Louis, St. Louis, MO, USA
| | - Omar H. Butt
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Daniel Jacobson
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Michael Garvin
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Alan R Templeton
- Department of Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Shawn Campagna
- Department of Chemistry, University of Tennessee, Knoxville, TN, USA
| | - Jane O’Halloran
- Division of Infectious Diseases, Washington University School of Medicine, St Louis, MO, USA
| | - Rachel Presti
- Division of Infectious Diseases, Washington University School of Medicine, St Louis, MO, USA
| | - Charles W. Goss
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Philip A. Mudd
- Department of Emergency Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Beau M. Ances
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Yun Ju Sung
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, MO, USA
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, MO, USA
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
44
|
Das BB, Akam-Venkata J, Abdulkarim M, Hussain T. Parametric Mapping Cardiac Magnetic Resonance Imaging for the Diagnosis of Myocarditis in Children in the Era of COVID-19 and MIS-C. CHILDREN (BASEL, SWITZERLAND) 2022; 9:1061. [PMID: 35884045 PMCID: PMC9320921 DOI: 10.3390/children9071061] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/08/2022] [Accepted: 07/14/2022] [Indexed: 12/03/2022]
Abstract
Myocarditis comprises many clinical presentations ranging from asymptomatic to sudden cardiac death. The history, physical examination, cardiac biomarkers, inflammatory markers, and electrocardiogram are usually helpful in the initial assessment of suspected acute myocarditis. Echocardiography is the primary tool to detect ventricular wall motion abnormalities, pericardial effusion, valvular regurgitation, and impaired function. The advancement of cardiac magnetic resonance (CMR) imaging has been helpful in clinical practice for diagnosing myocarditis. A recent Scientific Statement by the American Heart Association suggested CMR as a confirmatory test to diagnose acute myocarditis in children. However, standard CMR parametric mapping parameters for diagnosing myocarditis are unavailable in pediatric patients for consistency and reliability in the interpretation. The present review highlights the unmet clinical needs for standard CMR parametric criteria for diagnosing acute and chronic myocarditis in children and differentiating dilated chronic myocarditis phenotype from idiopathic dilated cardiomyopathy. Of particular relevance to today's practice, we also assess the potential and limitations of CMR to diagnose acute myocarditis in children exposed to severe acute respiratory syndrome coronavirus-2 infections. The latter section will discuss the multi-inflammatory syndrome in children (MIS-C) and mRNA coronavirus disease 19 vaccine-associated myocarditis.
Collapse
Affiliation(s)
- Bibhuti B. Das
- Department of Pediatrics, Children’s of Mississippi Heart Center, University of Mississippi Medical Center, Jackson, MS 39216, USA;
| | - Jyothsna Akam-Venkata
- Department of Pediatrics, Children’s of Mississippi Heart Center, University of Mississippi Medical Center, Jackson, MS 39216, USA;
| | - Mubeena Abdulkarim
- Pediatric Cardiology, Nicklaus Children’s Hospital, Miami, FL 33155, USA;
| | - Tarique Hussain
- Pediatric Cardiology, Children’s Health, UTSW Medical Center, Dallas, TX 75235, USA;
| |
Collapse
|
45
|
Huang X, Huang B, He Y, Feng L, Shi J, Wang L, Peng J, Chen Y. Sars-Cov-2 Spike Protein-Induced Damage of hiPSC-Derived Cardiomyocytes. Adv Biol (Weinh) 2022; 6:e2101327. [PMID: 35523737 PMCID: PMC9347759 DOI: 10.1002/adbi.202101327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 04/12/2022] [Indexed: 01/28/2023]
Abstract
Sars-Cov-2 may trigger molecular and functional alterations of cardiomyocytes (CMs) of the heart due to the presence of receptor angiotensin-converting enzyme 2 (ACE2) of the host cells. While the endocytic itinerary of the virus via cleavage of the spike protein of Sars-Cov-2 is well understood, the role of the remaining part of the spike protein subunit and ACE2 complex is still elusive. Herein, the possible effects of this complex are investigated by using synthetic spike proteins of Sars-Cov-2, human-induced pluripotent stem cells (hiPSC), and a culture device made of an arrayed monolayer of cross-linked nanofibers. hiPSCs are first differentiated into CMs that form cardiac tissue-like constructs with regular beating and expression of both ACE2 and gap junction protein Connexin 43. When incubated with the spike proteins, the hiPSC-CMs undergo a rhythmic fluctuation with overstretched sarcomere structures and dispersed gap junction proteins. When incubated with the spike proteins and supplementary angiotensin II, the damage of the spike protein on hiPSC-CMs is enhanced due to downregulated ACE2, chromatin margination, altered Connexin 43 expression, sarcomere disruption, and beating break. This discovery may imply latent effects of the spike proteins on the heart.
Collapse
Affiliation(s)
- Xiaochen Huang
- École Normale Supérieure‐PSL Research University, Département de Chimie, Sorbonne Universités‐UPMC Univ Paris 06, CNRS UMR 8640, PASTEUR, 24, rue LhomondParis75005France
| | - Boxin Huang
- École Normale Supérieure‐PSL Research University, Département de Chimie, Sorbonne Universités‐UPMC Univ Paris 06, CNRS UMR 8640, PASTEUR, 24, rue LhomondParis75005France
| | - Yong He
- École Normale Supérieure‐PSL Research University, Département de Chimie, Sorbonne Universités‐UPMC Univ Paris 06, CNRS UMR 8640, PASTEUR, 24, rue LhomondParis75005France
| | - Liang Feng
- École Normale Supérieure‐PSL Research University, Département de Chimie, Sorbonne Universités‐UPMC Univ Paris 06, CNRS UMR 8640, PASTEUR, 24, rue LhomondParis75005France
| | - Jian Shi
- MesoBioTech, 231 Rue Saint‐HonoréParis75001France
| | - Li Wang
- MesoBioTech, 231 Rue Saint‐HonoréParis75001France
| | - Juan Peng
- École Normale Supérieure‐PSL Research University, Département de Chimie, Sorbonne Universités‐UPMC Univ Paris 06, CNRS UMR 8640, PASTEUR, 24, rue LhomondParis75005France
| | - Yong Chen
- École Normale Supérieure‐PSL Research University, Département de Chimie, Sorbonne Universités‐UPMC Univ Paris 06, CNRS UMR 8640, PASTEUR, 24, rue LhomondParis75005France
| |
Collapse
|
46
|
Jum'ah H, Kundrapu S, Jabri A, Kondapaneni M, Tomashefski JF, Loeffler AG. Cardiac Macrophage Density in Covid-19 Infection: Relationship to Myocyte Necrosis and Acute Lung Injury. Cardiovasc Pathol 2022; 60:107447. [PMID: 35718082 PMCID: PMC9212794 DOI: 10.1016/j.carpath.2022.107447] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/27/2022] [Accepted: 06/12/2022] [Indexed: 11/30/2022] Open
Abstract
SARS-Cov-2 infection is not limited to the respiratory tract and can involve other organs including the heart, blood vessels, kidneys, liver, gastrointestinal tract, placenta, and skin. Covid-19 patients with cardiac involvement usually have higher morbidity and mortality compared to those without cardiac involvement. The frequency and the specificity of the myocardial pathological changes in patients who die after documented infection with SARS-Cov-2 is uncertain. Macrophages can be found in the normal heart (interstitium, around the endothelial cells and in the epicardial adipose tissue), and they are considered part of the major immune cell population in the heart. In this case-control autopsy study, we compare the gross and microscopic cardiac findings, and the available clinical characteristics between a group of 10 Covid-19 decedents and a control group of 20 patients who died with non-SARS-Cov-2 severe bronchopneumonia and/or diffuse alveolar damage. The objectives of this semi-quantitative study are to study single myocyte necrosis and its relation to the strain on the heart caused by lung injury as a causative mechanism, and to study the density of myocardial and epicardial macrophages in Covid-19 hearts in comparison to the control group, and in Covid-19 hearts with single myocyte necrosis in comparison to Covid-19 hearts without single myocyte necrosis. Lymphocytic myocarditis was not identified in any of the hearts from the Covid-19 or the control group. Single myocyte necrosis is more frequent in the Covid-19 group compared to the control group, suggesting that it is unrelated to the strain on the heart caused by underlying lung injury. The density of the macrophages in the epicardium and myocardium in the hearts of the Covid-19 group is higher compared to those in the control group. The density of epicardial macrophages is higher in the Covid-19 hearts with single myocyte necrosis than in those without. These observations contribute to our increasing appreciation of the role of macrophages in the pathophysiologic response to infection by SARS-CoV-2.
Collapse
Affiliation(s)
- Husam Jum'ah
- Department of Pathology, MetroHealth Medical Center, Cleveland, OH, US; Case Western Reserve University School of Medicine, Cleveland, OH
| | - Sirisha Kundrapu
- Department of Pathology, MetroHealth Medical Center, Cleveland, OH, US; Case Western Reserve University School of Medicine, Cleveland, OH
| | - Ahmad Jabri
- Case Western Reserve University School of Medicine, Cleveland, OH; Department of Cardiology, MetroHealth Medical Center, Cleveland, OH, US
| | - Meera Kondapaneni
- Case Western Reserve University School of Medicine, Cleveland, OH; Department of Cardiology, MetroHealth Medical Center, Cleveland, OH, US
| | - Joseph F Tomashefski
- Department of Pathology, MetroHealth Medical Center, Cleveland, OH, US; Case Western Reserve University School of Medicine, Cleveland, OH
| | - Agnes G Loeffler
- Department of Pathology, MetroHealth Medical Center, Cleveland, OH, US; Case Western Reserve University School of Medicine, Cleveland, OH.
| |
Collapse
|
47
|
McHenry A, Iyer K, Wang J, Liu C, Harigopal M. Detection of SARS-CoV-2 in tissue: the comparative roles of RT-qPCR, in situ RNA hybridization, and immunohistochemistry. Expert Rev Mol Diagn 2022; 22:559-574. [PMID: 35658709 DOI: 10.1080/14737159.2022.2085508] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The emergence of SARS-CoV-2, the causative agent the COVID-19 pandemic, has led to a rapidly expanding arsenal of molecular diagnostic assays for the detection of viral material in tissue specimens. AREAS COVERED We review the value and shortcomings of available tissue-based assays for SARS-CoV-2 detection in formalin-fixed paraffin-embedded (FFPE) tissue, including immunohistochemistry, in situ hybridization, and quantitative reverse transcription PCR (RT-qPCR). The validation, accuracy, and comparative utility of each method is discussed. Subsequently, we identify commercially available antibodies which render the greatest specificity and reproducibility of staining in FFPE specimens. EXPERT OPINION We offer expert opinion on the efficacy of such techniques and guidance for future implementation, both clinical and experimental.
Collapse
Affiliation(s)
- Austin McHenry
- Yale University School of Medicine, Department of Pathology, New Haven, CT, 06520, United States
| | - Krishna Iyer
- Yale University School of Medicine, Department of Pathology, New Haven, CT, 06520, United States
| | - Jianhi Wang
- Yale University School of Medicine, Department of Pathology, New Haven, CT, 06520, United States
| | - Chen Liu
- Yale University School of Medicine, Department of Pathology, New Haven, CT, 06520, United States
| | - Malini Harigopal
- Yale University School of Medicine, Department of Pathology, New Haven, CT, 06520, United States
| |
Collapse
|
48
|
Cardiovascular Tropism and Sequelae of SARS-CoV-2 Infection. Viruses 2022; 14:v14061137. [PMID: 35746609 PMCID: PMC9228192 DOI: 10.3390/v14061137] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/11/2022] [Accepted: 05/16/2022] [Indexed: 01/18/2023] Open
Abstract
The extrapulmonary manifestation of coronavirus disease-19 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), became apparent early in the ongoing pandemic. It is now recognized that cells of the cardiovascular system are targets of SARS-CoV-2 infection and associated disease pathogenesis. While some details are emerging, much remains to be understood pertaining to the mechanistic basis by which SARS-CoV-2 contributes to acute and chronic manifestations of COVID-19. This knowledge has the potential to improve clinical management for the growing populations of patients impacted by COVID-19. Here, we review the epidemiology and pathophysiology of cardiovascular sequelae of COVID-19 and outline proposed disease mechanisms, including direct SARS-CoV-2 infection of major cardiovascular cell types and pathogenic effects of non-infectious viral particles and elicited inflammatory mediators. Finally, we identify the major outstanding questions in cardiovascular COVID-19 research.
Collapse
|
49
|
Tobler DL, Pruzansky AJ, Naderi S, Ambrosy AP, Slade JJ. Long-Term Cardiovascular Effects of COVID-19: Emerging Data Relevant to the Cardiovascular Clinician. Curr Atheroscler Rep 2022; 24:563-570. [PMID: 35507278 PMCID: PMC9065238 DOI: 10.1007/s11883-022-01032-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2022] [Indexed: 12/15/2022]
Abstract
Purpose of Review COVID-19 is now a global pandemic and the illness affects multiple organ systems, including the cardiovascular system. Long-term cardiovascular consequences of COVID-19 are not yet fully characterized. This review seeks to consolidate available data on long-term cardiovascular complications of COVID-19 infection. Recent Findings Acute cardiovascular complications of COVID-19 infection include myocarditis, pericarditis, acute coronary syndrome, heart failure, pulmonary hypertension, right ventricular dysfunction, and arrhythmia. Long-term follow-up shows increased incidence of arrhythmia, heart failure, acute coronary syndrome, right ventricular dysfunction, myocardial fibrosis, hypertension, and diabetes mellitus. There is increased mortality in COVID-19 patients after hospital discharge, and initial myocardial injury is associated with increased mortality. Summary Emerging data demonstrates increased incidence of cardiovascular illness and structural changes in recovered COVID-19 patients. Future research will be important in understanding the clinical significance of these structural abnormalities, and to determine the effect of vaccines on preventing long-term cardiovascular complications.
Collapse
Affiliation(s)
- Diana L Tobler
- Department of Cardiology, Kaiser Permanente San Francisco Medical Center, San Francisco, CA, USA
| | - Alix J Pruzansky
- Department of Medicine, Kaiser Permanente Santa Clara Medical Center, Santa Clara, CA, USA
| | - Sahar Naderi
- Department of Cardiology, Kaiser Permanente San Francisco Medical Center, San Francisco, CA, USA
| | - Andrew P Ambrosy
- Department of Cardiology, Kaiser Permanente San Francisco Medical Center, San Francisco, CA, USA.,Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Justin J Slade
- Department of Cardiology, Kaiser Permanente San Francisco Medical Center, San Francisco, CA, USA.
| |
Collapse
|
50
|
Cao W, Birkenbach M, Chen S. Patterns of Inflammatory Cell Infiltration and Expression of STAT6 in the Lungs of Patients With COVID-19: An Autopsy Study. Appl Immunohistochem Mol Morphol 2022; 30:350-357. [PMID: 35363626 PMCID: PMC9066507 DOI: 10.1097/pai.0000000000001023] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 02/18/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 causes diffuse alveolar damage (DAD), lymphocyte infiltration in the lungs and a cytokine storm. In this study we examined inflammatory cell infiltrates and the expression of signal transducer and activator of transcription (STAT) 6 in the lungs of patients with coronavirus disease 2019 (COVID-19). METHODS Eighteen COVID-19 autopsy cases, 9 non-COVID cases with DAD, and 11 controls without lung diseases were included. Immunostainings for STAT6, CD3, CD4, CD8, CD68, and broad-spectrum keratins were performed. RESULTS The average age of COVID-19 patients was 64.4±2.1 years. The disease duration was 7 to 53 days. The number of pneumocytes, macrophages or CD3+ T cells was significantly increased in the lungs of patients with COVID-19. Patients' age above 67 years, blood troponin levels >0.2 ng/mL, platelet count >100×109/L, lung macrophages >130/high-power field (HPF), CD3+ T cells >145/HPF, CD8+ T cells <30/HPF, and CD8/CD4 ratio <1 were associated with shorter survival duration after onset of symptoms. In addition, STAT6 staining was much stronger in pneumocytes and lymphocytes in the lungs of patients with COVID-19 than non-COVID DAD patients or controls. CONCLUSION Older age, high blood troponin level and platelet count, more macrophages and fewer CD8+ T cells in the lungs of COVID-19 were associated with poorer outcome. STAT6 expression was increased in pneumocytes and lymphocytes in the lungs of patients with COVID-19, implying a role of STAT6 in cytokine storms.
Collapse
Affiliation(s)
- Weibiao Cao
- Departments of Pathology and Laboratory Medicine
- Medicine, Rhode Island Hospital and The Alpert Medical School of Brown University, Providence, RI
| | | | - Sonja Chen
- Departments of Pathology and Laboratory Medicine
| |
Collapse
|