1
|
Wu J, Xu S, Li Z, Cong B, Yang Z, Yang Z, Gao W, Liu S, Yu Z, Xu S, Li N, Hou J, Wang G, Cao X, Liu S. SARS-CoV-2 enhances complement-mediated endothelial injury via the suppression of membrane complement regulatory proteins. Emerg Microbes Infect 2025; 14:2467781. [PMID: 39945674 PMCID: PMC11873982 DOI: 10.1080/22221751.2025.2467781] [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: 10/30/2024] [Revised: 01/29/2025] [Accepted: 02/11/2025] [Indexed: 03/01/2025]
Abstract
Complement hyperactivation and thrombotic microangiopathy are closely associated with severe COVID-19. Endothelial dysfunction is a key mechanism underlying thrombotic microangiopathy. To address the relationship between endothelial injury, complement activation and thrombotic microangiopathy of severe COVID-19, we wonder whether, and if so, what and how SARS-CoV-2 factors make endothelial cells (ECs) sensitive to complement-mediated cytotoxicity. We revealed that multiple SARS-CoV-2 proteins enhanced complement-mediated cytotoxicity to ECs by inhibiting membrane complement regulatory proteins (CRPs) and enhancing the deposition of complement-recognizing component FCN1. By screening with CRISPR/Cas9-gRNA libraries, we identified that ADAMTS9, SYAP1, and HIGD1A as intrinsic regulators of CD59 on ECs, which were inhibited by the SARS-CoV-2 M, NSP16, and ORF9b proteins. IFN-γ, GM-CSF, and IFN-α upregulated CD55 and CD59, while IFN-γ antagonized the inhibition of CD59 by the three SARS-CoV-2 proteins. So, the deficiency of IFN-γ weakened the protection of ECs by CRPs against complement-mediated injury which may be enhanced during infection. Our findings illustrated the regulation of protection against complement-mediated attack on self-cells by SARS-CoV-2 infection and immune responses, providing insights into endothelial injury, thrombotic microangiopathy, and potential targets for treating severe COVID-19.
Collapse
Affiliation(s)
- Jian Wu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, People’s Republic of China
| | - Sanpeng Xu
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, People’s Republic of China
| | - Zhiqing Li
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, People’s Republic of China
| | - Boyi Cong
- Frontier Research Center for Cell Response, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, People’s Republic of China
| | - Zongheng Yang
- Department of Immunology, Center for Immunotherapy, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Zhichao Yang
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, People’s Republic of China
| | - Wanfeng Gao
- Frontier Research Center for Cell Response, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, People’s Republic of China
| | - Shuo Liu
- Department of Immunology, Center for Immunotherapy, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Zhou Yu
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, People’s Republic of China
| | - Sheng Xu
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, People’s Republic of China
| | - Nan Li
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, People’s Republic of China
| | - Jin Hou
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, People’s Republic of China
| | - Guoping Wang
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, People’s Republic of China
| | - Xuetao Cao
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, People’s Republic of China
- Frontier Research Center for Cell Response, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, People’s Republic of China
- Department of Immunology, Center for Immunotherapy, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Shuxun Liu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, People’s Republic of China
| |
Collapse
|
2
|
Baazim H, Koyuncu E, Tuncman G, Burak MF, Merkel L, Bahour N, Karabulut ES, Lee GY, Hanifehnezhad A, Karagoz ZF, Földes K, Engin I, Erman AG, Oztop S, Filazi N, Gul B, Ceylan A, Cinar OO, Can F, Kim H, Al-Hakeem A, Li H, Semerci F, Lin X, Yilmaz E, Ergonul O, Ozkul A, Hotamisligil GS. FABP4 as a therapeutic host target controlling SARS-CoV-2 infection. EMBO Mol Med 2025; 17:414-440. [PMID: 39843629 PMCID: PMC11904229 DOI: 10.1038/s44321-024-00188-x] [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: 05/09/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/24/2025] Open
Abstract
Host metabolic fitness is a critical determinant of infectious disease outcomes. Obesity, aging, and other related metabolic disorders are recognized as high-risk disease modifiers for respiratory infections, including coronavirus infections, though the underlying mechanisms remain unknown. Our study highlights fatty acid-binding protein 4 (FABP4), a key regulator of metabolic dysfunction and inflammation, as a modulator of SARS-CoV-2 pathogenesis, correlating strongly with disease severity in COVID-19 patients. We demonstrate that loss of FABP4 function, by genetic or pharmacological means, reduces SARS-CoV-2 replication and disrupts the formation of viral replication organelles in adipocytes and airway epithelial cells. Importantly, FABP4 inhibitor treatment of infected hamsters diminished lung viral titers, alleviated lung damage and reduced collagen deposition. These findings highlight the therapeutic potential of targeting host metabolism in limiting coronavirus replication and mitigating the pathogenesis of infection.
Collapse
Affiliation(s)
- Hatoon Baazim
- Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Gürol Tuncman
- Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - M Furkan Burak
- Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lea Merkel
- Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Nadine Bahour
- Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Ezgi Simay Karabulut
- Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Grace Yankun Lee
- Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Alireza Hanifehnezhad
- Ankara University, Faculty of Veterinary Medicine, Department of Virology, Ankara, Türkiye
| | | | | | - Ilayda Engin
- Ankara University, Biotechnology Institute, Ankara, Türkiye
| | | | - Sidika Oztop
- Ankara Medipol University, School of Medicine, Department of Medical Biology, Ankara, Türkiye
| | - Nazlican Filazi
- Mustafa Kemal University, Faculty of Veterinary Medicine, Department of Virology, Hatay, Türkiye
| | - Buket Gul
- Ankara University, Faculty of Veterinary Medicine, Department of Virology, Ankara, Türkiye
| | - Ahmet Ceylan
- Ankara University, Faculty of Veterinary Medicine, Department of Histology and Embryology, Ankara, Türkiye
| | - Ozge Ozgenc Cinar
- Ankara University, Faculty of Veterinary Medicine, Department of Histology and Embryology, Ankara, Türkiye
| | - Fusun Can
- Koç University, School of Medicine, Department of Infectious Diseases, Istanbul, Türkiye
| | - Hahn Kim
- Crescenta Biosciences Inc, Irvine, CA, USA
- Princeton University Small Molecule Screening Center, Princeton University, Princeton, NJ, USA
- Department of Chemistry, Princeton University, Princeton, NJ, USA
| | | | - Hui Li
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Xihong Lin
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Erkan Yilmaz
- Ankara University, Biotechnology Institute, Ankara, Türkiye
| | - Onder Ergonul
- Koç University, School of Medicine, Department of Infectious Diseases, Istanbul, Türkiye
| | - Aykut Ozkul
- Ankara University, Faculty of Veterinary Medicine, Department of Virology, Ankara, Türkiye.
| | - Gökhan S Hotamisligil
- Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Harvard-MIT Broad Institute, Cambridge, MA, USA.
| |
Collapse
|
3
|
Fekete M, Lehoczki A, Szappanos Á, Toth A, Mahdi M, Sótonyi P, Benyó Z, Yabluchanskiy A, Tarantini S, Ungvari Z. Cerebromicrovascular mechanisms contributing to long COVID: implications for neurocognitive health. GeroScience 2025; 47:745-779. [PMID: 39777702 PMCID: PMC11872997 DOI: 10.1007/s11357-024-01487-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Long COVID (also known as post-acute sequelae of SARS-CoV-2 infection [PASC] or post-COVID syndrome) is characterized by persistent symptoms that extend beyond the acute phase of SARS-CoV-2 infection, affecting approximately 10% to over 30% of those infected. It presents a significant clinical challenge, notably due to pronounced neurocognitive symptoms such as brain fog. The mechanisms underlying these effects are multifactorial, with mounting evidence pointing to a central role of cerebromicrovascular dysfunction. This review investigates key pathophysiological mechanisms contributing to cerebrovascular dysfunction in long COVID and their impacts on brain health. We discuss how endothelial tropism of SARS-CoV-2 and direct vascular infection trigger endothelial dysfunction, impaired neurovascular coupling, and blood-brain barrier disruption, resulting in compromised cerebral perfusion. Furthermore, the infection appears to induce mitochondrial dysfunction, enhancing oxidative stress and inflammation within cerebral endothelial cells. Autoantibody formation following infection also potentially exacerbates neurovascular injury, contributing to chronic vascular inflammation and ongoing blood-brain barrier compromise. These factors collectively contribute to the emergence of white matter hyperintensities, promote amyloid pathology, and may accelerate neurodegenerative processes, including Alzheimer's disease. This review also emphasizes the critical role of advanced imaging techniques in assessing cerebromicrovascular health and the need for targeted interventions to address these cerebrovascular complications. A deeper understanding of the cerebrovascular mechanisms of long COVID is essential to advance targeted treatments and mitigate its long-term neurocognitive consequences.
Collapse
Affiliation(s)
- Monika Fekete
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Andrea Lehoczki
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary.
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary.
| | - Ágnes Szappanos
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Department of Rheumatology and Clinical Immunology, Semmelweis University, Budapest, Hungary
| | - Attila Toth
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
- Research Centre for Molecular Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Mohamed Mahdi
- Laboratory of Retroviral Biochemistry, Department of Biochemistry and Molecular Biology, University of Debrecen, 4032, Debrecen, Hungary
- Infectology Clinic, University of Debrecen Clinical Centre, 4031, Debrecen, Hungary
| | - Péter Sótonyi
- Department of Vascular and Endovascular Surgery, Heart and Vascular Centre, Semmelweis University, 1122, Budapest, Hungary
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, 1094, Budapest, Hungary
- Cerebrovascular and Neurocognitive Disorders Research Group, HUN-REN , Semmelweis University, 1094, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
4
|
Owens CD, Pinto CB, Szarvas Z, Muranyi M, da C. Pinaffi-Langley AC, Peterfi A, Mukli P, Detwiler S, Olay L, Kaposzta Z, Smith K, Kirkpatrick AC, Saleh Velez F, Tarantini S, Csiszar A, Ungvari ZI, Prodan CI, Yabluchanskiy A. COVID-19 Exacerbates Neurovascular Uncoupling and Contributes to Endothelial Dysfunction in Patients with Mild Cognitive Impairment. Biomolecules 2024; 14:1621. [PMID: 39766328 PMCID: PMC11726736 DOI: 10.3390/biom14121621] [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: 09/30/2024] [Revised: 12/04/2024] [Accepted: 12/12/2024] [Indexed: 01/15/2025] Open
Abstract
Mild cognitive impairment (MCI) affects nearly 20% of older adults worldwide, with no targetable interventions for prevention. COVID-19 adversely affects cognition, with >70% of older adults with Long COVID presenting with cognitive complaints. Neurovascular coupling (NVC), an essential mechanism of cognitive function, declines with aging and is further attenuated in neurocognitive disorders. The effect of COVID-19 on NVC responses has yet to be addressed in older adults who are vulnerable to dementia progression. Participants with MCI and a history of COVID-19 (COV+, N = 31) and MCI participants with no history of infection (COV- N = 11) participated in this cross-sectional study to determine if COVID-19 affects cerebrocortical NVC responses and vascular function. Functional near-infrared spectroscopy was used to measure cerebrocortical NVC responses, and endothelial function was assessed via insonation of the brachial artery during a flow-mediated dilation protocol. NVC responses were elicited by the working memory n-back paradigm. NVC in the left dorsolateral prefrontal cortex and endothelial function was decreased in the COV+ group compared to the COV- group. These data provide mechanistic insight into how COVID-19 may exacerbate long-term cognitive sequela seen in older adults, highlighting the urgent need for further research and clinical trials to explore novel therapeutic interventions aimed at preserving/restoring NVC.
Collapse
Affiliation(s)
- Cameron D. Owens
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
| | - Camila B. Pinto
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
| | - Zsofia Szarvas
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Institute of Preventive Medicine and Public Health, Semmelweis University, 1089 Budapest, Hungary
| | - Mihaly Muranyi
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
| | - Ana Clara da C. Pinaffi-Langley
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Department of Nutritional Sciences, College of Allied Health, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA
| | - Anna Peterfi
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Institute of Preventive Medicine and Public Health, Semmelweis University, 1089 Budapest, Hungary
| | - Peter Mukli
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Institute of Preventive Medicine and Public Health, Semmelweis University, 1089 Budapest, Hungary
- Department of Physiology, Faculty of Medicine, Semmelweis University, 1087 Budapest, Hungary
| | - Sam Detwiler
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
| | - Lauren Olay
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
| | - Zalan Kaposzta
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Institute of Preventive Medicine and Public Health, Semmelweis University, 1089 Budapest, Hungary
- Department of Physiology, Faculty of Medicine, Semmelweis University, 1087 Budapest, Hungary
| | - Kenneth Smith
- Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA;
| | - Angelia C. Kirkpatrick
- Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA; (A.C.K.); (C.I.P.)
- Cardiovascular Disease Section, Department of Medicine, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA
| | - Faddi Saleh Velez
- Department of Neurology, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA;
| | - Stefano Tarantini
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Institute of Preventive Medicine and Public Health, Semmelweis University, 1089 Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Anna Csiszar
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Institute of Preventive Medicine and Public Health, Semmelweis University, 1089 Budapest, Hungary
| | - Zoltan I. Ungvari
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Institute of Preventive Medicine and Public Health, Semmelweis University, 1089 Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
| | - Calin I. Prodan
- Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA; (A.C.K.); (C.I.P.)
- Department of Neurology, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA;
| | - Andriy Yabluchanskiy
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Institute of Preventive Medicine and Public Health, Semmelweis University, 1089 Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
5
|
Russell SJ, Parker K, Lehoczki A, Lieberman D, Partha IS, Scott SJ, Phillips LR, Fain MJ, Nikolich JŽ. Post-acute sequelae of SARS-CoV-2 infection (Long COVID) in older adults. GeroScience 2024; 46:6563-6581. [PMID: 38874693 PMCID: PMC11493926 DOI: 10.1007/s11357-024-01227-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 05/26/2024] [Indexed: 06/15/2024] Open
Abstract
Long COVID, also known as PASC (post-acute sequelae of SARS-CoV-2), is a complex infection-associated chronic condition affecting tens of millions of people worldwide. Many aspects of this condition are incompletely understood. Among them is how this condition may manifest itself in older adults and how it might impact the older population. Here, we briefly review the current understanding of PASC in the adult population and examine what is known on its features with aging. Finally, we outline the major gaps and areas for research most germane to older adults.
Collapse
Affiliation(s)
- Samantha J Russell
- Division of General Internal Medicine, Geriatrics, and Palliative Medicine, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Center of Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Banner University Medicine-Tucson, Tucson, AZ, USA
| | - Karen Parker
- Division of General Internal Medicine, Geriatrics, and Palliative Medicine, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Center of Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Banner University Medicine-Tucson, Tucson, AZ, USA
| | - Andrea Lehoczki
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary
- Department of Haematology and Stem Cell Transplantation, National Institute for Haematology and Infectious Diseases, South Pest Central Hospital, 1097, Budapest, Hungary
- Department of Public Health, Semmelweis University, Budapest, Hungary
| | - David Lieberman
- Division of General Internal Medicine, Geriatrics, and Palliative Medicine, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Center of Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Banner University Medicine-Tucson, Tucson, AZ, USA
| | - Indu S Partha
- Division of General Internal Medicine, Geriatrics, and Palliative Medicine, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Banner University Medicine-Tucson, Tucson, AZ, USA
| | - Serena J Scott
- Division of General Internal Medicine, Geriatrics, and Palliative Medicine, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Center of Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Banner University Medicine-Tucson, Tucson, AZ, USA
| | - Linda R Phillips
- Division of General Internal Medicine, Geriatrics, and Palliative Medicine, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Center of Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- College of Nursing, University of Arizona, Tucson, AZ, USA
| | - Mindy J Fain
- Division of General Internal Medicine, Geriatrics, and Palliative Medicine, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA.
- Arizona Center of Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA.
- Banner University Medicine-Tucson, Tucson, AZ, USA.
- College of Nursing, University of Arizona, Tucson, AZ, USA.
| | - Janko Ž Nikolich
- Arizona Center of Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA.
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA.
- The Aegis Consortium for Pandemic-Free Future, University of Arizona Health Sciences, Tucson, AZ, USA.
| |
Collapse
|
6
|
Ma L, Cai L, Pan J, Cheng Z, Lv Y, Zheng J, Xu P, Zhang H, Chen X, Huang Y, Luo X, Zhao J, Xu L. The immunopathology of coronary microembolization and the underlying inflammopathophysiological mechanisms. Allergol Immunopathol (Madr) 2024; 52:137-146. [PMID: 39515808 DOI: 10.15586/aei.v52i6.1170] [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: 06/09/2024] [Accepted: 08/22/2024] [Indexed: 11/16/2024]
Abstract
In coronary microembolization, inflammatory cell infiltration, patchy necrosis, and extensive intra-myocardial hemorrhage are dominant, which induce myocardial dysfunction with clinical symptoms of chronic ischemic cardiomyopathy. Microembolization can lead to obstruction of the coronary microvessels and result in the micro-infarction of the heart. The inflammation and elevated expression of the tumor necrosis factor in cardiomyocytes and the activation of extracellular ERK are involved in initiating the inflammatory response mechanism. The PI3K/Akt signaling pathway is the enriched pathway, and for controlling, inhibition of PI3K/Akt is necessary. Furthermore, the release of cytokines and the activation of inflammasomes contribute to the enhancement of vascular permeability, which results in edema within the myocardium. The immune response and inflammation represent the primary triggers in this process. The ability to control immune response and inflammation reactions may lead to the development of new therapies for microembolization.
Collapse
Affiliation(s)
- Li Ma
- Department of Cardiovascular Medicine, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Liping Cai
- Health Management Center, Wuhan Third Hospital, Wuhan, China
| | - Jiayue Pan
- Xiangtao College of Medicine, Xiangtao College Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Zimin Cheng
- Department of Cardiovascular Medicine, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Yuanyuan Lv
- Department of Cardiovascular Medicine, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Jie Zheng
- Department of Cardiovascular Medicine, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Peicheng Xu
- Department of Cardiovascular Medicine, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Hong Zhang
- Department of Cardiovascular Medicine, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Xinyu Chen
- Department of Cardiovascular Medicine, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Yimeng Huang
- Department of Cardiovascular Medicine, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Xiaolei Luo
- Department of Cardiovascular Medicine, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Jinhe Zhao
- Department of Cardiovascular Medicine, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China;
| | - Liang Xu
- Department of ICU, Wuhan Wuchang Hospital, Wuhan, China;
| |
Collapse
|
7
|
Molnar T, Lehoczki A, Fekete M, Varnai R, Zavori L, Erdo-Bonyar S, Simon D, Berki T, Csecsei P, Ezer E. Mitochondrial dysfunction in long COVID: mechanisms, consequences, and potential therapeutic approaches. GeroScience 2024; 46:5267-5286. [PMID: 38668888 PMCID: PMC11336094 DOI: 10.1007/s11357-024-01165-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 04/15/2024] [Indexed: 08/22/2024] Open
Abstract
The COVID-19 pandemic, caused by the SARS-CoV-2 virus, has introduced the medical community to the phenomenon of long COVID, a condition characterized by persistent symptoms following the resolution of the acute phase of infection. Among the myriad of symptoms reported by long COVID sufferers, chronic fatigue, cognitive disturbances, and exercise intolerance are predominant, suggesting systemic alterations beyond the initial viral pathology. Emerging evidence has pointed to mitochondrial dysfunction as a potential underpinning mechanism contributing to the persistence and diversity of long COVID symptoms. This review aims to synthesize current findings related to mitochondrial dysfunction in long COVID, exploring its implications for cellular energy deficits, oxidative stress, immune dysregulation, metabolic disturbances, and endothelial dysfunction. Through a comprehensive analysis of the literature, we highlight the significance of mitochondrial health in the pathophysiology of long COVID, drawing parallels with similar clinical syndromes linked to post-infectious states in other diseases where mitochondrial impairment has been implicated. We discuss potential therapeutic strategies targeting mitochondrial function, including pharmacological interventions, lifestyle modifications, exercise, and dietary approaches, and emphasize the need for further research and collaborative efforts to advance our understanding and management of long COVID. This review underscores the critical role of mitochondrial dysfunction in long COVID and calls for a multidisciplinary approach to address the gaps in our knowledge and treatment options for those affected by this condition.
Collapse
Affiliation(s)
- Tihamer Molnar
- Department of Anaesthesiology and Intensive Care, Medical School, University of Pecs, Pecs, Hungary
| | - Andrea Lehoczki
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary
- Department of Haematology and Stem Cell Transplantation, National Institute for Haematology and Infectious Diseases, South Pest Central Hospital, 1097, Budapest, Hungary
- Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Monika Fekete
- Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Reka Varnai
- Department of Primary Health Care, Medical School University of Pecs, Pecs, Hungary
| | | | - Szabina Erdo-Bonyar
- Department of Immunology and Biotechnology, Medical School, University of Pecs, Pecs, Hungary
| | - Diana Simon
- Department of Immunology and Biotechnology, Medical School, University of Pecs, Pecs, Hungary
| | - Tímea Berki
- Department of Immunology and Biotechnology, Medical School, University of Pecs, Pecs, Hungary
| | - Peter Csecsei
- Department of Neurosurgery, Medical School, University of Pecs, Ret U 2, 7624, Pecs, Hungary.
| | - Erzsebet Ezer
- Department of Anaesthesiology and Intensive Care, Medical School, University of Pecs, Pecs, Hungary
| |
Collapse
|
8
|
Clarke M, Falcione S, Boghozian R, Todoran R, Zhang Y, C. Real MG, StPierre A, Joy T, Jickling GC. Viral Infection and Ischemic Stroke: Emerging Trends and Mechanistic Insights. J Am Heart Assoc 2024; 13:e035892. [PMID: 39258541 PMCID: PMC11935600 DOI: 10.1161/jaha.124.035892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 07/22/2024] [Indexed: 09/12/2024]
Abstract
Population studies have suggested that viral infections may be contributing to risk of ischemic stroke, although the mechanisms for this are unclear. In this review, we examine the epidemiological evidence supporting the involvement of viral diseases, including influenza, COVID-19, chronic herpesvirus infections, and hepatitis C in current trends of stroke incidence. To support these associations, we highlight the virus-host interactions that are critical in the context of stroke, including direct effects of acute and persistent viral infections on vascular function, inflammation, and thrombosis. Additionally, we evaluate the systemic changes that occur during viral infection that can predispose individuals to ischemic stroke, including alterations in blood pressure regulation, coagulation, and lipid metabolism. Our review emphasizes the need to further elucidate precise mechanisms involved in viral infections and stroke risk. Future research will inform the development of targeted interventions for stroke prevention in the context of viral diseases.
Collapse
Affiliation(s)
- Michael Clarke
- Faculty of Medicine and DentistryDepartment of Medical Microbiology and ImmunologyUniversity of AlbertaEdmontonABCanada
| | - Sarina Falcione
- Faculty of Medicine and DentistryDepartment of MedicineDivision of NeurologyUniversity of AlbertaEdmontonABCanada
| | - Roobina Boghozian
- Faculty of Medicine and DentistryDepartment of MedicineDivision of NeurologyUniversity of AlbertaEdmontonABCanada
| | - Raluca Todoran
- Faculty of Medicine and DentistryDepartment of MedicineDivision of NeurologyUniversity of AlbertaEdmontonABCanada
| | - Yiran Zhang
- Faculty of Medicine and DentistryDepartment of MedicineDivision of NeurologyUniversity of AlbertaEdmontonABCanada
| | - Maria Guadalupe C. Real
- Faculty of Medicine and DentistryDepartment of MedicineDivision of NeurologyUniversity of AlbertaEdmontonABCanada
| | - Alexis StPierre
- Faculty of Medicine and DentistryDepartment of MedicineDivision of NeurologyUniversity of AlbertaEdmontonABCanada
| | - Twinkle Joy
- Faculty of Medicine and DentistryDepartment of MedicineDivision of NeurologyUniversity of AlbertaEdmontonABCanada
| | - Glen C. Jickling
- Faculty of Medicine and DentistryDepartment of Medical Microbiology and ImmunologyUniversity of AlbertaEdmontonABCanada
- Faculty of Medicine and DentistryDepartment of MedicineDivision of NeurologyUniversity of AlbertaEdmontonABCanada
| |
Collapse
|
9
|
Chidambaram V, Kumar A, Sadaf MI, Lu E, Al’Aref SJ, Tarun T, Galiatsatos P, Gulati M, Blumenthal RS, Leucker TM, Karakousis PC, Mehta JL. COVID-19 in the Initiation and Progression of Atherosclerosis: Pathophysiology During and Beyond the Acute Phase. JACC. ADVANCES 2024; 3:101107. [PMID: 39113913 PMCID: PMC11304887 DOI: 10.1016/j.jacadv.2024.101107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 04/24/2024] [Accepted: 06/01/2024] [Indexed: 08/10/2024]
Abstract
The incidence of atherosclerotic cardiovascular disease is increasing globally, especially in low- and middle-income countries, despite significant efforts to reduce traditional risk factors. Premature subclinical atherosclerosis has been documented in association with several viral infections. The magnitude of the recent COVID-19 pandemic has highlighted the need to understand the association between SARS-CoV-2 and atherosclerosis. This review examines various pathophysiological mechanisms, including endothelial dysfunction, platelet activation, and inflammatory and immune hyperactivation triggered by SARS-CoV-2 infection, with specific attention on their roles in initiating and promoting the progression of atherosclerotic lesions. Additionally, it addresses the various pathogenic mechanisms by which COVID-19 in the post-acute phase may contribute to the development of vascular disease. Understanding the overlap of these syndromes may enable novel therapeutic strategies. We further explore the need for guidelines for closer follow-up for the often-overlooked evidence of atherosclerotic cardiovascular disease among patients with recent COVID-19, particularly those with cardiometabolic risk factors.
Collapse
Affiliation(s)
- Vignesh Chidambaram
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Amudha Kumar
- Division of Cardiology, Department of Medicine, Loyola University Medical Center, Maywood, Illinois, USA
| | - Murrium I. Sadaf
- Division of Cardiovascular Medicine, Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Emily Lu
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Subhi J. Al’Aref
- Division of Cardiovascular Medicine, Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Tushar Tarun
- Division of Cardiovascular Medicine, Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Panagis Galiatsatos
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Martha Gulati
- Barbra Streisand Women's Heart Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Roger S. Blumenthal
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Thorsten M. Leucker
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Petros C. Karakousis
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jawahar L. Mehta
- Division of Cardiovascular Medicine, Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Division of Cardiovascular Medicine, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| |
Collapse
|
10
|
Keramidas P, Pitou M, Papachristou E, Choli-Papadopoulou T. Insights into the Activation of Unfolded Protein Response Mechanism during Coronavirus Infection. Curr Issues Mol Biol 2024; 46:4286-4308. [PMID: 38785529 PMCID: PMC11120126 DOI: 10.3390/cimb46050261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/24/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
Coronaviruses represent a significant class of viruses that affect both animals and humans. Their replication cycle is strongly associated with the endoplasmic reticulum (ER), which, upon virus invasion, triggers ER stress responses. The activation of the unfolded protein response (UPR) within infected cells is performed from three transmembrane receptors, IRE1, PERK, and ATF6, and results in a reduction in protein production, a boost in the ER's ability to fold proteins properly, and the initiation of ER-associated degradation (ERAD) to remove misfolded or unfolded proteins. However, in cases of prolonged and severe ER stress, the UPR can also instigate apoptotic cell death and inflammation. Herein, we discuss the ER-triggered host responses after coronavirus infection, as well as the pharmaceutical targeting of the UPR as a potential antiviral strategy.
Collapse
Affiliation(s)
| | | | | | - Theodora Choli-Papadopoulou
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (P.K.); (M.P.); (E.P.)
| |
Collapse
|
11
|
Colzani M, Bargehr J, Mescia F, Williams EC, Knight-Schrijver V, Lee J, Summers C, Mohorianu I, Smith KGC, Lyons PA, Sinha S. Proinflammatory cytokines driving cardiotoxicity in COVID-19. Cardiovasc Res 2024; 120:174-187. [PMID: 38041432 PMCID: PMC10936751 DOI: 10.1093/cvr/cvad174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 10/09/2023] [Accepted: 10/20/2023] [Indexed: 12/03/2023] Open
Abstract
AIMS Cardiac involvement is common in patients hospitalized with COVID-19 and correlates with an adverse disease trajectory. While cardiac injury has been attributed to direct viral cytotoxicity, serum-induced cardiotoxicity secondary to serological hyperinflammation constitutes a potentially amenable mechanism that remains largely unexplored. METHODS AND RESULTS To investigate serological drivers of cardiotoxicity in COVID-19 we have established a robust bioassay that assessed the effects of serum from COVID-19 confirmed patients on human embryonic stem cell (hESC)-derived cardiomyocytes. We demonstrate that serum from COVID-19 positive patients significantly reduced cardiomyocyte viability independent of viral transduction, an effect that was also seen in non-COVID-19 acute respiratory distress syndrome (ARDS). Serum from patients with greater disease severity led to worse cardiomyocyte viability and this significantly correlated with levels of key inflammatory cytokines, including IL-6, TNF-α, IL1-β, IL-10, CRP, and neutrophil to lymphocyte ratio with a specific reduction of CD4+ and CD8+ cells. Combinatorial blockade of IL-6 and TNF-α partly rescued the phenotype and preserved cardiomyocyte viability and function. Bulk RNA sequencing of serum-treated cardiomyocytes elucidated specific pathways involved in the COVID-19 response impacting cardiomyocyte viability, structure, and function. The observed effects of serum-induced cytotoxicity were cell-type selective as serum exposure did not adversely affect microvascular endothelial cell viability but resulted in endothelial activation and a procoagulant state. CONCLUSION These results provide direct evidence that inflammatory cytokines are at least in part responsible for the cardiovascular damage seen in COVID-19 and characterise the downstream activated pathways in human cardiomyocytes. The serum signature of patients with severe disease indicates possible targets for therapeutic intervention.
Collapse
Affiliation(s)
- Maria Colzani
- Wellcome – MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Puddicombe Way, CB2 0AW Cambridge, UK
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Addenbrooke's Hospital, Hills Rd, CB2 0SP Cambridge, UK
| | - Johannes Bargehr
- Wellcome – MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Puddicombe Way, CB2 0AW Cambridge, UK
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Addenbrooke's Hospital, Hills Rd, CB2 0SP Cambridge, UK
| | - Federica Mescia
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Addenbrooke's Hospital, Hills Rd, CB2 0SP Cambridge, UK
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Puddicombe Way, CB2 0AW Cambridge, UK
| | - Eleanor C Williams
- Wellcome – MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Puddicombe Way, CB2 0AW Cambridge, UK
| | - Vincent Knight-Schrijver
- Wellcome – MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Puddicombe Way, CB2 0AW Cambridge, UK
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Addenbrooke's Hospital, Hills Rd, CB2 0SP Cambridge, UK
| | - Jonathan Lee
- Wellcome – MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Puddicombe Way, CB2 0AW Cambridge, UK
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Addenbrooke's Hospital, Hills Rd, CB2 0SP Cambridge, UK
| | - Charlotte Summers
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Addenbrooke's Hospital, Hills Rd, CB2 0SP Cambridge, UK
- Wolfson Lung Injury Unit, Heart and Lung Research Institute, Cambridge Biomedical Campus, Papworth Road, CB2 0BB Cambridge, UK
| | - Irina Mohorianu
- Wellcome – MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Puddicombe Way, CB2 0AW Cambridge, UK
| | - Kenneth G C Smith
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Addenbrooke's Hospital, Hills Rd, CB2 0SP Cambridge, UK
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Puddicombe Way, CB2 0AW Cambridge, UK
| | - Paul A Lyons
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Addenbrooke's Hospital, Hills Rd, CB2 0SP Cambridge, UK
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Puddicombe Way, CB2 0AW Cambridge, UK
| | - Sanjay Sinha
- Wellcome – MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Puddicombe Way, CB2 0AW Cambridge, UK
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Addenbrooke's Hospital, Hills Rd, CB2 0SP Cambridge, UK
| |
Collapse
|
12
|
Lui KO, Ma Z, Dimmeler S. SARS-CoV-2 induced vascular endothelial dysfunction: direct or indirect effects? Cardiovasc Res 2024; 120:34-43. [PMID: 38159046 DOI: 10.1093/cvr/cvad191] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 10/18/2023] [Accepted: 11/03/2023] [Indexed: 01/03/2024] Open
Abstract
Clinical evidence reveals that manifestations of endothelial dysfunction are widely observed in COVID-19 and long-COVID patients. However, whether these detrimental effects are caused by direct infection of the endothelium or are indirectly mediated by systemic inflammation has been a matter of debate. It has been well acknowledged that endothelial cells (ECs) of the cardiovascular system ubiquitously express the SARS-CoV-2 entry receptor angiotensin-converting enzyme 2 (ACE2), yet accumulating evidence suggests that it is more predominantly expressed by pericytes and vascular smooth muscle cells of the mammalian blood vessel. Besides, replicative infection of ECs by SARS-CoV-2 has yet to be demonstrated both in vitro and in vivo. In this study, we review latest research on endothelial ACE2 expression in different vascular beds, and the heterogeneity in various EC subsets with differential ACE2 expression in response to SARS-CoV-2. We also discuss ACE2-independent alternative mechanisms underlying endothelial activation in COVID-19, and the clinical manifestations of SARS-CoV-2-induced endothelial dysfunction. Altogether, understanding ACE2-dependent and ACE2-independent mechanisms driving SARS-CoV-2-induced vascular dysfunction would shed light on strategies of more effective therapies targeting cardiovascular complications associated with COVID-19.
Collapse
Affiliation(s)
- Kathy O Lui
- Department of Chemical Pathology, and Li Ka Shing Institute of Health Science, Prince of Wales Hospital, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Sha Tin, New Territories, 999077 Hong Kong, China
| | - Zhangjing Ma
- Department of Chemical Pathology, and Li Ka Shing Institute of Health Science, Prince of Wales Hospital, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Sha Tin, New Territories, 999077 Hong Kong, China
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, and Faculty of Biological Sciences, Goethe University Frankfurt, Theodor Stern Kai 7, 60590 Frankfurt, Germany
| |
Collapse
|
13
|
Smadja DM, Jannot AS, Philippe A, Lu E, Rancic J, Sanchez O, Chocron R, Gendron N, Diehl JL. Circulating Von Willebrand factor: a consistent biomarker predicting in-hospital mortality across different waves of the COVID-19 pandemic. Angiogenesis 2024; 27:1-4. [PMID: 38070063 DOI: 10.1007/s10456-023-09901-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/25/2023] [Indexed: 02/22/2024]
Affiliation(s)
- David M Smadja
- Paris Cité University, Innovative Therapies in Hemostasis, INSERM, 75006, Paris, France.
- Hematology department, AP-HP, Georges Pompidou European Hospital, 20 rue Leblanc, 75015, Paris, France.
| | - Anne-Sophie Jannot
- Medical Informatics, Biostatistics and Public Health Department, Centre de Recherche des Cordeliers, AP-HP, Georges Pompidou European Hospital, European Georges Pompidou Hospital, AP-HP.CUP, University Paris Cité, Paris, France
| | - Aurélien Philippe
- Paris Cité University, Innovative Therapies in Hemostasis, INSERM, 75006, Paris, France
- Hematology department, AP-HP, Georges Pompidou European Hospital, 20 rue Leblanc, 75015, Paris, France
| | - Estelle Lu
- Medical Informatics, Biostatistics and Public Health Department, Centre de Recherche des Cordeliers, AP-HP, Georges Pompidou European Hospital, European Georges Pompidou Hospital, AP-HP.CUP, University Paris Cité, Paris, France
| | - Jeanne Rancic
- Paris Cité University, Innovative Therapies in Hemostasis, INSERM, 75006, Paris, France
- Hematology department, AP-HP, Georges Pompidou European Hospital, 20 rue Leblanc, 75015, Paris, France
| | - Olivier Sanchez
- Paris Cité University, Innovative Therapies in Hemostasis, INSERM, 75006, Paris, France
- Respiratory Medicine Department, Georges Pompidou European Hospital, AP-HP, 75015, Paris, France
| | - Richard Chocron
- Emergency department, AP-HP, Georges Pompidou European Hospital, 75015, Paris, France
- Paris Cité University, PARCC, INSERM, 75015, Paris, France
| | - Nicolas Gendron
- Paris Cité University, Innovative Therapies in Hemostasis, INSERM, 75006, Paris, France
- Hematology department, AP-HP, Georges Pompidou European Hospital, 20 rue Leblanc, 75015, Paris, France
| | - Jean-Luc Diehl
- Paris Cité University, Innovative Therapies in Hemostasis, INSERM, 75006, Paris, France
- Intensive care unit, AP-HP, Georges Pompidou European Hospital, 75015, Paris, France
| |
Collapse
|
14
|
Vieceli Dalla Sega F, Fortini F, Licastro D, Monego SD, Degasperi M, Ascierto A, Marracino L, Severi P, D'Accolti M, Soffritti I, Brambilla M, Camera M, Tremoli E, Contoli M, Spadaro S, Campo G, Ferrari R, Caselli E, Rizzo P. Serum from COVID-19 patients promotes endothelial cell dysfunction through protease-activated receptor 2. Inflamm Res 2024; 73:117-130. [PMID: 38117300 DOI: 10.1007/s00011-023-01823-y] [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: 05/31/2023] [Revised: 10/06/2023] [Accepted: 11/13/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Endothelial dysfunction plays a central role in the pathophysiology of COVID-19 and is closely linked to the severity and mortality of the disease. The inflammatory response to SARS-CoV-2 infection can alter the capacity of the endothelium to regulate vascular tone, immune responses, and the balance between anti-thrombotic and pro-thrombotic properties. However, the specific endothelial pathways altered during COVID-19 still need to be fully understood. OBJECTIVE In this study, we sought to identify molecular changes in endothelial cells induced by circulating factors characteristic of COVID-19. METHODS AND RESULTS To this aim, we cultured endothelial cells with sera from patients with COVID-19 or non-COVID-19 pneumonia. Through transcriptomic analysis, we were able to identify a distinctive endothelial phenotype that is induced by sera from COVID-19 patients. We confirmed and expanded this observation in vitro by showing that COVID-19 serum alters functional properties of endothelial cells leading to increased apoptosis, loss of barrier integrity, and hypercoagulability. Furthermore, we demonstrated that these endothelial dysfunctions are mediated by protease-activated receptor 2 (PAR-2), as predicted by transcriptome network analysis validated by in vitro functional assays. CONCLUSION Our findings provide the rationale for further studies to evaluate whether targeting PAR-2 may be a clinically effective strategy to counteract endothelial dysfunction in COVID-19.
Collapse
Affiliation(s)
| | | | | | | | | | - Alessia Ascierto
- Department of Translational Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Luisa Marracino
- Department of Translational Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Paolo Severi
- Department of Translational Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Maria D'Accolti
- Department of Chemical, Pharmaceutical, and Agricultural Sciences, and LTTA, Section of Microbiology, University of Ferrara, Ferrara, Italy
| | - Irene Soffritti
- Department of Chemical, Pharmaceutical, and Agricultural Sciences, and LTTA, Section of Microbiology, University of Ferrara, Ferrara, Italy
| | | | - Marina Camera
- Centro Cardiologico Monzino IRCCS, Milan, Italy
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Elena Tremoli
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
| | - Marco Contoli
- Respiratory Section, Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Savino Spadaro
- Intensive Care Unit, Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Gianluca Campo
- Cardiology Unit, Azienda Ospedaliero-Universitaria di Ferrara, University of Ferrara, Ferrara, Italy
| | - Roberto Ferrari
- Department of Translational Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Elisabetta Caselli
- Department of Chemical, Pharmaceutical, and Agricultural Sciences, and LTTA, Section of Microbiology, University of Ferrara, Ferrara, Italy
| | - Paola Rizzo
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
- Department of Translational Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| |
Collapse
|
15
|
Perico L, Benigni A, Remuzzi G. SARS-CoV-2 and the spike protein in endotheliopathy. Trends Microbiol 2024; 32:53-67. [PMID: 37393180 PMCID: PMC10258582 DOI: 10.1016/j.tim.2023.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 07/03/2023]
Abstract
SARS-CoV-2, the causative agent of COVID-19, primarily affects the epithelial compartment in the upper and lower airways. There is evidence that the microvasculature in both the pulmonary and extrapulmonary systems is a major target of SARS-CoV-2. Consistent with this, vascular dysfunction and thrombosis are the most severe complications in COVID-19. The proinflammatory milieu triggered by the hyperactivation of the immune system by SARS-CoV-2 has been suggested to be the main trigger for endothelial dysfunction during COVID-19. More recently, a rapidly growing number of reports have indicated that SARS-CoV-2 can interact directly with endothelial cells through the spike protein, leading to multiple instances of endothelial dysfunction. Here, we describe all the available findings showing the direct effect of the SARS-CoV-2 spike protein on endothelial cells and offer mechanistic insights into the molecular basis of vascular dysfunction in severe COVID-19.
Collapse
Affiliation(s)
- Luca Perico
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy.
| | - Ariela Benigni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy
| |
Collapse
|
16
|
Jones EAV. Mechanism of COVID-19-Induced Cardiac Damage from Patient, In Vitro and Animal Studies. Curr Heart Fail Rep 2023; 20:451-460. [PMID: 37526812 PMCID: PMC10589152 DOI: 10.1007/s11897-023-00618-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/07/2023] [Indexed: 08/02/2023]
Abstract
PURPOSE OF REVIEW Though patient studies have been important for understanding the disease, research done in animals and cell culture complement our knowledge from patient data and provide insight into the mechanism of the disease. Understanding how COVID causes damage to the heart is essential to understanding possible long-term consequences. RECENT FINDINGS COVID-19 is primarily a disease that attacks the lungs; however, it is known to have important consequences in many other tissues including the heart. Though myocarditis does occur in some patients, for most cases of cardiac damage, the injury arises from scarring either due to myocardial infarction or micro-infarction. The main focus is on how COVID affects blood flow through the coronaries. We review how endothelial activation leads to a hypercoagulative state in COVID-19. We also emphasize the effects that the cytokine storm can directly have on the regulation of coronary blood flow. Since the main two cell types that can be infected in the heart are pericytes and cardiomyocytes, we further describe the known effects on pericyte function and how that can further lead to microinfarcts within the heart. Though many of these effects are systemic, this review focuses on the consequences on cardiac tissue of this dysregulation and the role that it has in the formation of myocardial scarring.
Collapse
Affiliation(s)
- Elizabeth A V Jones
- Centre for Molecular and Vascular Biology, Herestraat 49, Bus 911, 3000, KU, Leuven, Belgium.
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, Netherlands.
| |
Collapse
|
17
|
Furini G, De Carli A, Fonnesu R, Spezia PG, Scebba F, Pistello M, Lai M, Lionetti V. Gene silencing of endothelial von Willebrand factor reduces the susceptibility of human endothelial cells to SARS-CoV-2 infection. FEBS J 2023; 290:4300-4315. [PMID: 37098810 DOI: 10.1111/febs.16808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 03/03/2023] [Accepted: 04/25/2023] [Indexed: 04/27/2023]
Abstract
Mechanisms underlying vascular endothelial susceptibility to infection by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are not fully understood. Emerging evidence indicates that patients lacking von Willebrand factor (vWF), an endothelial hallmark, are less severely affected by SARS-CoV-2 infection, yet the precise role of endothelial vWF in modulating coronavirus entry into endothelial cells is unknown. In the present study, we demonstrated that effective gene silencing by short interfering RNA (siRNA) for vWF expression in resting human umbilical vein endothelial cells (HUVECs) significantly reduced by 56% the cellular levels of SARS-CoV-2 genomic RNA. Similar reduction in intracellular SARS-CoV-2 genomic RNA levels was observed in non-activated HUVECs treated with siRNA targeting angiotensin-converting enzyme 2 (ACE2), the cellular gateway to coronavirus. By integrating quantitative information from real-time PCR and high-resolution confocal imaging, we demonstrated that ACE2 gene expression and its plasma membrane localization in HUVECs were both markedly reduced after treatment with siRNA anti-vWF or anti-ACE2. Conversely, siRNA anti-ACE2 did not reduce endothelial vWF gene expression and protein levels. Finally, SARS-CoV-2 infection of viable HUVECs was enhanced by overexpression of vWF, which increased ACE2 levels. Of note, we found a similar increase in interferon-β mRNA levels following transfection with untargeted, anti-vWF or anti-ACE2 siRNA and pcDNA3.1-WT-VWF. We envision that siRNA targeting endothelial vWF will protect against productive endothelial infection by SARS-CoV-2 through downregulation of ACE2 expression and might serve as a novel tool to induce disease resistance by modulating the regulatory role of vWF on ACE2 expression.
Collapse
Affiliation(s)
- Giulia Furini
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Pisa, Italy
- UOSVD Anesthesia and Resuscitation, Fondazione Toscana "G. Monasterio", Pisa, Italy
| | - Alessandro De Carli
- Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, Retrovirus Center, University of Pisa, Italy
| | - Rossella Fonnesu
- Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, Retrovirus Center, University of Pisa, Italy
| | - Pietro Giorgio Spezia
- Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, Retrovirus Center, University of Pisa, Italy
| | - Francesca Scebba
- BioMedicine Laboratory, Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Pisa, Italy
| | - Mauro Pistello
- Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, Retrovirus Center, University of Pisa, Italy
- Virology Unit, Pisa University Hospital, Italy
| | - Michele Lai
- Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, Retrovirus Center, University of Pisa, Italy
- CISUP - Centre for Instrumentation Sharing - University of Pisa, Italy
| | - Vincenzo Lionetti
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Pisa, Italy
- UOSVD Anesthesia and Resuscitation, Fondazione Toscana "G. Monasterio", Pisa, Italy
- BioMedicine Laboratory, Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Pisa, Italy
| |
Collapse
|
18
|
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
|
19
|
Escaffre O, Szaniszlo P, Törő G, Vilas CL, Servantes BJ, Lopez E, Juelich TL, Levine CB, McLellan SLF, Cardenas JC, Freiberg AN, Módis K. Hydrogen Sulfide Ameliorates SARS-CoV-2-Associated Lung Endothelial Barrier Disruption. Biomedicines 2023; 11:1790. [PMID: 37509430 PMCID: PMC10376201 DOI: 10.3390/biomedicines11071790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/14/2023] [Accepted: 06/17/2023] [Indexed: 07/30/2023] Open
Abstract
Recent studies have confirmed that lung microvascular endothelial injury plays a critical role in the pathophysiology of COVID-19. Our group and others have demonstrated the beneficial effects of H2S in several pathological processes and provided a rationale for considering the therapeutic implications of H2S in COVID-19 therapy. Here, we evaluated the effect of the slow-releasing H2S donor, GYY4137, on the barrier function of a lung endothelial cell monolayer in vitro, after challenging the cells with plasma samples from COVID-19 patients or inactivated SARS-CoV-2 virus. We also assessed how the cytokine/chemokine profile of patients' plasma, endothelial barrier permeability, and disease severity correlated with each other. Alterations in barrier permeability after treatments with patient plasma, inactivated virus, and GYY4137 were monitored and assessed by electrical impedance measurements in real time. We present evidence that GYY4137 treatment reduced endothelial barrier permeability after plasma challenge and completely reversed the endothelial barrier disruption caused by inactivated SARS-CoV-2 virus. We also showed that disease severity correlated with the cytokine/chemokine profile of the plasma but not with barrier permeability changes in our assay. Overall, these data demonstrate that treatment with H2S-releasing compounds has the potential to ameliorate SARS-CoV-2-associated lung endothelial barrier disruption.
Collapse
Affiliation(s)
- Olivier Escaffre
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Institute for Human Infections & Immunity, Sealy & Smith Foundation, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Peter Szaniszlo
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Gabor Törő
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Caitlyn L. Vilas
- John Sealy School of Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Brenna J. Servantes
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Ernesto Lopez
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Terry L. Juelich
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Corri B. Levine
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Susan L. F. McLellan
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jessica C. Cardenas
- The Center for Translational Injury Research, Department of Surgery, UTHealth McGovern Medical School, Houston, TX 77030, USA
| | - Alexander N. Freiberg
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Institute for Human Infections & Immunity, Sealy & Smith Foundation, University of Texas Medical Branch, Galveston, TX 77555, USA
- The Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Katalin Módis
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
20
|
Sykes RA, Neves KB, Alves-Lopes R, Caputo I, Fallon K, Jamieson NB, Kamdar A, Legrini A, Leslie H, McIntosh A, McConnachie A, Morrow A, McFarlane RW, Mangion K, McAbney J, Montezano AC, Touyz RM, Wood C, Berry C. Vascular mechanisms of post-COVID-19 conditions: Rho-kinase is a novel target for therapy. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2023; 9:371-386. [PMID: 37019821 PMCID: PMC10236521 DOI: 10.1093/ehjcvp/pvad025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/24/2023] [Accepted: 04/04/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND In post-coronavirus disease-19 (post-COVID-19) conditions (long COVID), systemic vascular dysfunction is implicated, but the mechanisms are uncertain, and the treatment is imprecise. METHODS AND RESULTS Patients convalescing after hospitalization for COVID-19 and risk factor matched controls underwent multisystem phenotyping using blood biomarkers, cardiorenal and pulmonary imaging, and gluteal subcutaneous biopsy (NCT04403607). Small resistance arteries were isolated and examined using wire myography, histopathology, immunohistochemistry, and spatial transcriptomics. Endothelium-independent (sodium nitroprusside) and -dependent (acetylcholine) vasorelaxation and vasoconstriction to the thromboxane A2 receptor agonist, U46619, and endothelin-1 (ET-1) in the presence or absence of a RhoA/Rho-kinase inhibitor (fasudil), were investigated. Thirty-seven patients, including 27 (mean age 57 years, 48% women, 41% cardiovascular disease) 3 months post-COVID-19 and 10 controls (mean age 57 years, 20% women, 30% cardiovascular disease), were included. Compared with control responses, U46619-induced constriction was increased (P = 0.002) and endothelium-independent vasorelaxation was reduced in arteries from COVID-19 patients (P < 0.001). This difference was abolished by fasudil. Histopathology revealed greater collagen abundance in COVID-19 arteries {Masson's trichrome (MT) 69.7% [95% confidence interval (CI): 67.8-71.7]; picrosirius red 68.6% [95% CI: 64.4-72.8]} vs. controls [MT 64.9% (95% CI: 59.4-70.3) (P = 0.028); picrosirius red 60.1% (95% CI: 55.4-64.8), (P = 0.029)]. Greater phosphorylated myosin light chain antibody-positive staining in vascular smooth muscle cells was observed in COVID-19 arteries (40.1%; 95% CI: 30.9-49.3) vs. controls (10.0%; 95% CI: 4.4-15.6) (P < 0.001). In proof-of-concept studies, gene pathways associated with extracellular matrix alteration, proteoglycan synthesis, and viral mRNA replication appeared to be upregulated. CONCLUSION Patients with post-COVID-19 conditions have enhanced vascular fibrosis and myosin light change phosphorylation. Rho-kinase activation represents a novel therapeutic target for clinical trials.
Collapse
Affiliation(s)
- Robert A Sykes
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow, UK
| | - Karla B Neves
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
- Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Rhéure Alves-Lopes
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
| | - Ilaria Caputo
- Università degli Studi di Padova, 35122 Padova, Italy
| | - Kirsty Fallon
- Clinical Research Facility, Queen Elizabeth University Hospital, NHS Greater Glasgow & Clyde Health Board, Glasgow, UK
| | - Nigel B Jamieson
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Anna Kamdar
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
| | - Assya Legrini
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Holly Leslie
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Alasdair McIntosh
- Robertson Centre for Biostatistics, School of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Alex McConnachie
- Robertson Centre for Biostatistics, School of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Andrew Morrow
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow, UK
| | | | - Kenneth Mangion
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
- Department of Cardiology, Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde Health Board, Glasgow, UK
| | - John McAbney
- Institute of Biomedical and Life Sciences (FBLS), University of Glasgow, Glasgow G12 8QQ, UK
| | - Augusto C Montezano
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, QC H4A 3J1, Canada
| | - Rhian M Touyz
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, QC H4A 3J1, Canada
| | - Colin Wood
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Colin Berry
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow, UK
- Department of Cardiology, Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde Health Board, Glasgow, UK
| |
Collapse
|
21
|
Abstract
Viral infections are a leading cause of myocarditis and pericarditis worldwide, conditions that frequently coexist. Myocarditis and pericarditis were some of the early comorbidities associated with SARS-CoV-2 infection and COVID-19. Many epidemiologic studies have been conducted since that time concluding that SARS-CoV-2 increased the incidence of myocarditis/pericarditis at least 15× over pre-COVID levels although the condition remains rare. The incidence of myocarditis pre-COVID was reported at 1 to 10 cases/100 000 individuals and with COVID ranging from 150 to 4000 cases/100 000 individuals. Before COVID-19, some vaccines were reported to cause myocarditis and pericarditis in rare cases, but the use of novel mRNA platforms led to a higher number of reported cases than with previous platforms providing new insight into potential pathogenic mechanisms. The incidence of COVID-19 vaccine-associated myocarditis/pericarditis covers a large range depending on the vaccine platform, age, and sex examined. Importantly, the findings highlight that myocarditis occurs predominantly in male patients aged 12 to 40 years regardless of whether the cause was due to a virus-like SARS-CoV-2 or associated with a vaccine-a demographic that has been reported before COVID-19. This review discusses findings from COVID-19 and COVID-19 vaccine-associated myocarditis and pericarditis considering the known symptoms, diagnosis, management, treatment, and pathogenesis of disease that has been gleaned from clinical research and animal models. Sex differences in the immune response to COVID-19 are discussed, and theories for how mRNA vaccines could lead to myocarditis/pericarditis are proposed. Additionally, gaps in our understanding that need further research are raised.
Collapse
Affiliation(s)
- DeLisa Fairweather
- Department of Cardiovascular Medicine (D.F., D.J.B., D.N.D., L.T.C.), Mayo Clinic, Jacksonville, FL
- Department of Environmental Health Sciences and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (D.F.,)
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN (D.F., D.J.B., D.N.D.)
| | - Danielle J. Beetler
- Department of Cardiovascular Medicine (D.F., D.J.B., D.N.D., L.T.C.), Mayo Clinic, Jacksonville, FL
- Mayo Clinic Graduate School of Biomedical Sciences (D.J.B., D.N.D.), Mayo Clinic, Jacksonville, FL
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN (D.F., D.J.B., D.N.D.)
| | - Damian N. Di Florio
- Department of Cardiovascular Medicine (D.F., D.J.B., D.N.D., L.T.C.), Mayo Clinic, Jacksonville, FL
- Mayo Clinic Graduate School of Biomedical Sciences (D.J.B., D.N.D.), Mayo Clinic, Jacksonville, FL
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN (D.F., D.J.B., D.N.D.)
| | - Nicolas Musigk
- Deutsches Herzzentrum der Charité, Berlin, Germany (N.M., B.H.)
| | | | - Leslie T. Cooper
- Department of Cardiovascular Medicine (D.F., D.J.B., D.N.D., L.T.C.), Mayo Clinic, Jacksonville, FL
| |
Collapse
|
22
|
Matsumoto C, Shibata S, Kishi T, Morimoto S, Mogi M, Yamamoto K, Kobayashi K, Tanaka M, Asayama K, Yamamoto E, Nakagami H, Hoshide S, Mukoyama M, Kario K, Node K, Rakugi H. Long COVID and hypertension-related disorders: a report from the Japanese Society of Hypertension Project Team on COVID-19. Hypertens Res 2023; 46:601-619. [PMID: 36575228 PMCID: PMC9793823 DOI: 10.1038/s41440-022-01145-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/28/2022]
Abstract
The coronavirus disease 2019 (COVID-19) affects infected patients even after the acute phase and impairs their health and quality of life by causing a wide variety of symptoms, referred to as long COVID. Although the evidence is still insufficient, hypertension is suspected to be a potential risk factor for long COVID, and the occurrence of cardiovascular diseases seems to be a key facet of multiple conditions observed in long COVID. Nonetheless, there are few reports that comprehensively review the impacts of long COVID on hypertension and related disorders. As a sequel to our previous report in 2020 which reviewed the association of COVID-19 and hypertension, we summarize the possible influences of long COVID on hypertension-related organs, including the cardiovascular system, kidney, and endocrine system, as well as the pathophysiological mechanisms associated with the disorders in this review. Given that the clinical course of COVID-19 is highly affected by age and sex, we also review the impacts of these factors on long COVID. Lastly, we discuss areas of uncertainty and future directions, which may lead to better understanding and improved prognosis of clinical problems associated with COVID-19.
Collapse
Affiliation(s)
- Chisa Matsumoto
- Department of Cardiology, Preventive medicine, Tokyo Medical University, Tokyo, Japan.
| | - Shigeru Shibata
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan.
| | - Takuya Kishi
- Department of Graduate School of Medicine (Cardiology), International University of Health and Welfare, Okawa, Japan
| | - Satoshi Morimoto
- Department of Endocrinology and Hypertension, Tokyo Women's Medical University, Tokyo, Japan
| | - Masaki Mogi
- Department of Pharmacology, Ehime University Graduate School of Medicine, Toon, Japan
| | - Koichi Yamamoto
- Department of Geriatric and General Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Kazuo Kobayashi
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Kobayashi Internal Medicine Clinic, Sagamihara, Japan
| | - Masami Tanaka
- Department of Internal Medicine, Adachi Medical Center, Tokyo Women's Medical University, Tokyo, Japan
| | - Kei Asayama
- Department of Hygiene and Public Health, Teikyo University School of Medicine, Tokyo, Japan
| | - Eiichiro Yamamoto
- Department of Cardiovascular Medicine, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Hironori Nakagami
- Department of Health Development and Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
| | - Satoshi Hoshide
- Division of Cardiovascular Medicine, Department of Medicine, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Masashi Mukoyama
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Kazuomi Kario
- Division of Cardiovascular Medicine, Department of Medicine, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Koichi Node
- Department of Cardiovascular Medicine, Saga University, Saga, Japan
| | - Hiromi Rakugi
- Department of Geriatric and General Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
23
|
Maruhashi T, Higashi Y. Current topic of vascular function in hypertension. Hypertens Res 2023; 46:630-637. [PMID: 36604472 PMCID: PMC9813887 DOI: 10.1038/s41440-022-01147-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/23/2022] [Accepted: 12/05/2022] [Indexed: 01/06/2023]
Abstract
Vascular function assessment is useful for the evaluation of atherosclerosis severity, which may provide additional information for cardiovascular risk stratification. In addition, vascular function assessment is helpful for a better understanding of pathophysiological associations between vascular dysfunction and cardiometabolic disorders. In 2020 and 2021, although coronavirus disease 2019 (COVID-19) was still a worldwide challenge for health care systems, many excellent articles regarding vascular function were published in Hypertension Research and other major cardiovascular and hypertension journals. In this review, we summarize new findings on vascular function and discuss the association between vascular function and COVID-19, the importance of lifestyle modifications for the maintenance of vascular function, and the usefulness of vascular function tests for cardiovascular risk assessment. We hope this review will be helpful for the management of cardiovascular risk factors, including hypertension and cardiovascular diseases, in clinical practice.
Collapse
Affiliation(s)
- Tatsuya Maruhashi
- Department of Regenerative Medicine, Division of Radiation Medical Science, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan.
| | - Yukihito Higashi
- Department of Regenerative Medicine, Division of Radiation Medical Science, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan.,Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| |
Collapse
|
24
|
Yao S, Yu Y, Xu L, Pan X. Genomic and clinical features of endoplasmic reticulum stress factor in digestive system pan-cancer studies. Front Oncol 2023; 12:1072576. [PMID: 36698399 PMCID: PMC9868864 DOI: 10.3389/fonc.2022.1072576] [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: 10/17/2022] [Accepted: 11/29/2022] [Indexed: 01/11/2023] Open
Abstract
Introduction Digestive system pan-cancer is one of the lethal malignant tumors, which have the propensity for poor prognosis and difficult treatment. Endoplasmic reticulum (ER) stress has served as a pivotal role in the progression of the tumor, while the implication of ER stress on digestive system pan-cancers still needs elucidation, especially from the perspective of clinical outcome and that of genomic features. Methods First, Among the ER STRESS factors from the REACTOME_UNFOLDED_PROTEIN_RESPONSE_UPR (113 genes) and HALLMARK_UNFOLDED_PROTEIN_RESPONSE (92 genes) terms, 153 ER STRESS regulators were identified after removing replicates. The somatic mutation data and copy number variation data of gastrointestinal pan-cancer were downloaded from The Cancer Genome Atlas (TCGA) database. Then, we explored the clinical outcome and genetic mutation of ER stress-related differentially expressed genes (DEGs) by multiple bioinformatics analysis. Subsequently, we analyzed the Spearman correlation between the drug sensitivity of 179 gastrointestinal anticancer drugs and the transcriptional expression of 153 ER stress factors in 769 cancer cell lines of the GDSC2 cohort. Next, ssGSEA method was used to quantify the immune cell infiltration scores in the tumor microenvironment, and Spearman correlation was used to calculate the correlation between ER stress scores and immune cell infiltration. Finally, we analyzed the cellular origin of ER stress factor dysregulation. Results We analyzed the genomic changes and clinical outcomes of ER stress factors in different tumors of gastrointestinal pan-cancer. Endoplasmic reticulum stress factor (ER) in digestive tract tumors showed high SNV mutation frequency, less methylation dysregulation and was associated with multiple oncogenic pathways. Endoplasmic reticulum stress factor (ER) is a risk factor for many cancers, but the effect on overall survival in rectal adenocarcinoma is opposite to that in other gastrointestinal tumors. And ER stress factors are highly correlated with drugs that target important pathways. Discussion Based on the clinical prognosis and genomic analysis of ER stress-related factors in patients with gastrointestinal pan-cancer, this study provides a new direction for further research on gastrointestinal pan-cancer.
Collapse
Affiliation(s)
- Sheng Yao
- Department of Gastroenterology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yuanquan Yu
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China,Laboratory of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Liyi Xu
- Department of Gastroenterology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China,*Correspondence: Liyi Xu, ; Xiang Pan,
| | - Xiang Pan
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China,Laboratory of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China,*Correspondence: Liyi Xu, ; Xiang Pan,
| |
Collapse
|
25
|
Innate Immunity in Cardiovascular Diseases-Identification of Novel Molecular Players and Targets. J Clin Med 2023; 12:jcm12010335. [PMID: 36615135 PMCID: PMC9821340 DOI: 10.3390/jcm12010335] [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: 11/01/2022] [Revised: 12/20/2022] [Accepted: 12/25/2022] [Indexed: 01/03/2023] Open
Abstract
During the past few years, unexpected developments have driven studies in the field of clinical immunology. One driver of immense impact was the outbreak of a pandemic caused by the novel virus SARS-CoV-2. Excellent recent reviews address diverse aspects of immunological re-search into cardiovascular diseases. Here, we specifically focus on selected studies taking advantage of advanced state-of-the-art molecular genetic methods ranging from genome-wide epi/transcriptome mapping and variant scanning to optogenetics and chemogenetics. First, we discuss the emerging clinical relevance of advanced diagnostics for cardiovascular diseases, including those associated with COVID-19-with a focus on the role of inflammation in cardiomyopathies and arrhythmias. Second, we consider newly identified immunological interactions at organ and system levels which affect cardiovascular pathogenesis. Thus, studies into immune influences arising from the intestinal system are moving towards therapeutic exploitation. Further, powerful new research tools have enabled novel insight into brain-immune system interactions at unprecedented resolution. This latter line of investigation emphasizes the strength of influence of emotional stress-acting through defined brain regions-upon viral and cardiovascular disorders. Several challenges need to be overcome before the full impact of these far-reaching new findings will hit the clinical arena.
Collapse
|
26
|
Abstract
Although coronary artery dilation is associated with both multisystem inflammatory syndrome in children (MIS-C) and Kawaski disease, there is currently no evidence of long-term coronary artery damage in MIS-C2. The SARS-CoV-2 spike protein does not seem to act as a superantigen that binds the Vβ-chain of the T cell receptor6; the skewing of Vβ21.3 expression on T cells in children with MIS-C is most likely attributable to a non-SARS-CoV-2 related antigen7. The incidence and severity of MIS-C declined during the Omicron wave of the COVID-19 pandemic as compared with earlier waves, although the precise reason for this decline remains to be determined10.
Collapse
|
27
|
Strong attenuation of SARS-CoV-2 Omicron BA.1 and increased replication of the BA.5 subvariant in human cardiomyocytes. Signal Transduct Target Ther 2022; 7:395. [PMID: 36566244 PMCID: PMC9789943 DOI: 10.1038/s41392-022-01256-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 11/15/2022] [Accepted: 11/20/2022] [Indexed: 12/26/2022] Open
|
28
|
Pelisek J, Reutersberg B, Greber UF, Zimmermann A. Vascular dysfunction in COVID-19 patients: update on SARS-CoV-2 infection of endothelial cells and the role of long non-coding RNAs. Clin Sci (Lond) 2022; 136:1571-1590. [PMID: 36367091 PMCID: PMC9652506 DOI: 10.1042/cs20220235] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 08/16/2023]
Abstract
Although COVID-19 is primarily a respiratory disease, it may affect also the cardiovascular system. COVID-19 patients with cardiovascular disorder (CVD) develop a more severe disease course with a significantly higher mortality rate than non-CVD patients. A common denominator of CVD is the dysfunction of endothelial cells (ECs), increased vascular permeability, endothelial-to-mesenchymal transition, coagulation, and inflammation. It has been assumed that clinical complications in COVID-19 patients suffering from CVD are caused by SARS-CoV-2 infection of ECs through the angiotensin-converting enzyme 2 (ACE2) receptor and the cellular transmembrane protease serine 2 (TMPRSS2) and the consequent dysfunction of the infected vascular cells. Meanwhile, other factors associated with SARS-CoV-2 entry into the host cells have been described, including disintegrin and metalloproteinase domain-containing protein 17 (ADAM17), the C-type lectin CD209L or heparan sulfate proteoglycans (HSPG). Here, we discuss the current data about the putative entry of SARS-CoV-2 into endothelial and smooth muscle cells. Furthermore, we highlight the potential role of long non-coding RNAs (lncRNAs) affecting vascular permeability in CVD, a process that might exacerbate disease in COVID-19 patients.
Collapse
Affiliation(s)
- Jaroslav Pelisek
- Department of Vascular Surgery, University Hospital Zürich, Zürich, Switzerland
| | | | - Urs F Greber
- Department of Molecular Life Sciences, University of Zürich, Switzerland
| | | |
Collapse
|
29
|
Solimando AG, Marziliano D, Ribatti D. SARS-CoV-2 and Endothelial Cells: Vascular Changes, Intussusceptive Microvascular Growth and Novel Therapeutic Windows. Biomedicines 2022; 10:2242. [PMID: 36140343 PMCID: PMC9496230 DOI: 10.3390/biomedicines10092242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/16/2022] Open
Abstract
Endothelial activation in infectious diseases plays a crucial role in understanding and predicting the outcomes and future treatments of several clinical conditions. COVID-19 is no exception. Moving from basic principles to novel approaches, an evolving view of endothelial activation provides insights into a better knowledge of the upstream actors in COVID-19 as a crucial future direction for managing SARS-CoV-2 and other infections. Assessing the function of resting and damaged endothelial cells in infection, particularly in COVID-19, five critical processes emerged controlling thrombo-resistance: vascular integrity, blood flow regulation, immune cell trafficking, angiogenesis and intussusceptive microvascular growth. Endothelial cell injury is associated with thrombosis, increased vessel contraction and a crucial phenomenon identified as intussusceptive microvascular growth, an unprecedented event of vessel splitting into two lumens through the integration of circulating pro-angiogenic cells. An essential awareness of endothelial cells and their phenotypic changes in COVID-19 inflammation is pivotal to understanding the vascular biology of infections and may offer crucial new therapeutic windows.
Collapse
Affiliation(s)
- Antonio Giovanni Solimando
- Guido Baccelli Unit of Internal Medicine, Department of Biomedical Sciences and Human Oncology, School of Medicine, Aldo Moro University of Bari, 70124 Bari, Italy
| | - Donatello Marziliano
- Guido Baccelli Unit of Internal Medicine, Department of Biomedical Sciences and Human Oncology, School of Medicine, Aldo Moro University of Bari, 70124 Bari, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, 70124 Bari, Italy
| |
Collapse
|
30
|
Urata R, Ikeda K, Yamazaki E, Ueno D, Katayama A, Shin-Ya M, Ohgitani E, Mazda O, Matoba S. Senescent endothelial cells are predisposed to SARS-CoV-2 infection and subsequent endothelial dysfunction. Sci Rep 2022; 12:11855. [PMID: 35879338 PMCID: PMC9314328 DOI: 10.1038/s41598-022-15976-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 07/01/2022] [Indexed: 12/11/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19), caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), remains to spread worldwide. COVID-19 is characterized by the striking high mortality in elderly; however, its mechanistic insights remain unclear. Systemic thrombosis has been highlighted in the pathogenesis of COVID-19, and lung microangiopathy in association with endothelial cells (ECs) injury has been reported by post-mortem analysis of the lungs. Here, we experimentally investigated the SARS-CoV-2 infection in cultured human ECs, and performed a comparative analysis for post-infection molecular events using early passage and replicative senescent ECs. We found that; (1) SARS-CoV-2 infects ECs but does not replicate and disappears in 72 hours without causing severe cell damage, (2) Senescent ECs are highly susceptible to SARS-CoV-2 infection, (3) SARS-CoV-2 infection alters various genes expression, which could cause EC dysfunctions, (4) More genes expression is affected in senescent ECs by SARS-CoV-2 infection than in early passage ECs, which might causes further exacerbated dysfunction in senescent ECs. These data suggest that sustained EC dysfunctions due to SARS-CoV-2 infection may contribute to the microangiopathy in the lungs, leading to deteriorated inflammation and thrombosis in COVID-19. Our data also suggest a possible causative role of EC senescence in the aggravated disease in elder COVID-19 patients.
Collapse
Affiliation(s)
- Ryota Urata
- Department of Cardiology, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyo, Kyoto, 602-8566, Japan
| | - Koji Ikeda
- Department of Cardiology, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyo, Kyoto, 602-8566, Japan. .,Department of Epidemiology for Longevity and Regional Health, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyo, Kyoto, 602-8566, Japan.
| | - Ekura Yamazaki
- Department of Cardiology, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyo, Kyoto, 602-8566, Japan
| | - Daisuke Ueno
- Department of Cardiology, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyo, Kyoto, 602-8566, Japan
| | - Akiko Katayama
- Department of Cardiology, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyo, Kyoto, 602-8566, Japan
| | - Masaharu Shin-Ya
- Department of Immunology, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyo, Kyoto, 602-8566, Japan
| | - Eriko Ohgitani
- Department of Immunology, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyo, Kyoto, 602-8566, Japan
| | - Osam Mazda
- Department of Immunology, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyo, Kyoto, 602-8566, Japan
| | - Satoaki Matoba
- Department of Cardiology, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyo, Kyoto, 602-8566, Japan
| |
Collapse
|
31
|
Ghanam AR, Bryant WB, Miano JM. Of mice and human-specific long noncoding RNAs. Mamm Genome 2022; 33:281-292. [PMID: 35106622 PMCID: PMC8806012 DOI: 10.1007/s00335-022-09943-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 01/10/2022] [Indexed: 01/05/2023]
Abstract
The number of human LncRNAs has now exceeded all known protein-coding genes. Most studies of human LncRNAs have been conducted in cell culture systems where various mechanisms of action have been worked out. On the other hand, efforts to elucidate the function of human LncRNAs in an in vivo setting have been limited. In this brief review, we highlight some strengths and weaknesses of studying human LncRNAs in the mouse. Special consideration is given to bacterial artificial chromosome transgenesis and genome editing. The integration of these technical innovations offers an unprecedented opportunity to complement and extend the expansive literature of cell culture models for the study of human LncRNAs. Two different examples of how BAC transgenesis and genome editing can be leveraged to gain insight into human LncRNA regulation and function in mice are presented: the random integration of a vascular cell-enriched LncRNA and a targeted approach for a new LncRNA immediately upstream of the ACE2 gene, which encodes the receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiologic agent underlying the coronavirus disease-19 (COVID-19) pandemic.
Collapse
Affiliation(s)
- Amr R Ghanam
- Vascular Biology Center, Department of Medicine, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, CL-3060, Augusta, GA, 30912, USA
| | - William B Bryant
- Vascular Biology Center, Department of Medicine, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, CL-3060, Augusta, GA, 30912, USA
| | - Joseph M Miano
- Vascular Biology Center, Department of Medicine, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, CL-3060, Augusta, GA, 30912, USA.
| |
Collapse
|
32
|
Péterfi A, Mészáros Á, Szarvas Z, Pénzes M, Fekete M, Fehér Á, Lehoczki A, Csípő T, Fazekas-Pongor V. Comorbidities and increased mortality of COVID-19 among the elderly: A systematic review. Physiol Int 2022; 109:163-176. [PMID: 35575986 DOI: 10.1556/2060.2022.00206] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 02/14/2022] [Accepted: 03/01/2022] [Indexed: 02/18/2024]
Abstract
Purpose The purpose of current review is to conduct a systematic overview of articles published between 2019 and 2021 on the relationship of comorbidities and mortality due to Coronavirus Disease 2019 (COVID-19) among the elderly population. Methods We conducted a systematic search on PubMed for articles published between 2019 and 2021 to identify any cohort and case-control studies that investigated the relationship of comorbidities and COVID-19 mortality among the elderly, defined as 60 years of age and above. Databases were searched independently by two authors. Disagreements were resolved by the inclusion of a third investigator. Reviews, systematic reviews, and meta-analyses were excluded from our systematic review. Results A total of 15 studies were selected for our systematic review. Of the included studies, 3 were case-control, 3 were prospective cohort studies and 9 were retrospective cohort studies. As for size, 10 studies were conducted on populations of <1000 participants, 3 ranging from 1001 to 10,000, and 2 on populations of >10,000 individuals. The included studies found that the presence of certain conditions, such as cardiovascular, respiratory, renal diseases, malignancies, diseases of the nervous system and diabetes are associated to increased mortality in populations that consisted of elderly patients. Conclusion Results of our systematic review suggest that comorbidities contribute to increased COVID-19 mortality among the elderly. The detrimental effect of comorbidities and advanced age on the immune response could lead to a more frequent occurrence of symptomatic and severe infections with COVID-19.
Collapse
Affiliation(s)
- Anna Péterfi
- 1 Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Ágota Mészáros
- 1 Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Zsófia Szarvas
- 1 Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Melinda Pénzes
- 1 Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Mónika Fekete
- 1 Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Ágnes Fehér
- 1 Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Andrea Lehoczki
- 2 National Institute for Hematology and Infectious Diseases, Department of Hematology and Stem Cell Transplantation, South Pest Central Hospital, Budapest, Hungary
| | - Tamás Csípő
- 1 Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Vince Fazekas-Pongor
- 1 Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
33
|
Persistent viral RNA shedding of SARS-CoV-2 is associated with delirium incidence and six-month mortality in hospitalized COVID-19 patients. GeroScience 2022; 44:1241-1254. [PMID: 35538386 PMCID: PMC9090540 DOI: 10.1007/s11357-022-00561-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/17/2022] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Persistent viral RNA shedding of SARS-CoV-2 following COVID-19 has increasingly been recognized, with limited understanding of its implications on outcomes in hospitalized COVID-19 patients. METHODS We retrospectively assessed for persistent viral shedding across Northwestern Medicine Healthcare (NMHC) patients between March and August 2020. We assessed for predictors of persistent viral shedding, in-hospital delirium, and six-month mortality using binary logistic regression. RESULTS Of the 2,518 hospitalized patients with an RT-PCR-confirmed diagnosis of COVID-19, 959 underwent repeat SARS-CoV-2 RT-PCR at least fourteen days from initial positive testing. Of those, 405 (42.2%) patients were found to have persistent viral shedding. Persistent viral shedding was associated with male sex, increased BMI, diabetes mellitus, chronic kidney disease, and exposure to corticosteroids during initial COVID-19 hospitalization. Persistent viral shedding was independently associated with incidence of in-hospital delirium after adjusting for factors including severity of respiratory dysfunction (OR 2.45; 95% CI 1.75, 3.45). Even after adjusting for age, severity of respiratory dysfunction, and occurrence of in-hospital delirium, persistent viral shedding remained significantly associated with increased six-month mortality (OR 2.43; 95% CI 1.42, 4.29). CONCLUSIONS Persistent viral shedding occurs frequently in hospitalized COVID-19 patients and is associated with in-hospital delirium and increased six-month mortality.
Collapse
|
34
|
Raghavan S, Leo MD. Histamine Potentiates SARS-CoV-2 Spike Protein Entry Into Endothelial Cells. Front Pharmacol 2022; 13:872736. [PMID: 35548336 PMCID: PMC9084361 DOI: 10.3389/fphar.2022.872736] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/23/2022] [Indexed: 01/05/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) which causes coronavirus disease (COVID-19) is one of the most serious global health crises in recent history. COVID-19 patient symptoms range from life-threatening to mild and asymptomatic, which presents unique problems in identifying, quarantining, and treating the affected individuals. The emergence of unusual symptoms among survivors, now referred to as “Long COVID”, is concerning, especially since much about the condition and the treatment of it is still relatively unknown. Evidence so far also suggests that some of these symptoms can be attributed to vascular inflammation. Although famotidine, the commonly used histamine H2 receptor (H2R) blocker, was shown to have no antiviral activity, recent reports indicate that it could prevent adverse outcomes in COVID-19 patients. Histamine is a classic proinflammatory mediator, the levels of which increase along with other cytokines during COVID-19 infection. Histamine activates H2R signaling, while famotidine specifically blocks H2R activation. Investigating the effects of recombinant SARS-CoV-2 spike protein S1 Receptor-Binding Domain (Spike) on ACE2 expression in cultured human coronary artery endothelial cells, we found that the presence of histamine potentiated spike-mediated ACE2 internalization into endothelial cells. This effect was blocked by famotidine, protein kinase A inhibition, or by H2 receptor protein knockdown. Together, these results indicate that histamine and histamine receptor signaling is likely essential for spike protein to induce ACE2 internalization in endothelial cells and cause endothelial dysfunction and that this effect can be blocked by the H2R blocker, famotidine.
Collapse
|
35
|
Sulli A, Gotelli E, Bica PF, Schiavetti I, Pizzorni C, Aloè T, Grosso M, Barisione E, Paolino S, Smith V, Cutolo M. Detailed videocapillaroscopic microvascular changes detectable in adult COVID-19 survivors. Microvasc Res 2022; 142:104361. [PMID: 35339493 PMCID: PMC8942583 DOI: 10.1016/j.mvr.2022.104361] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/08/2022] [Accepted: 03/14/2022] [Indexed: 12/12/2022]
Abstract
Objective COVID-19 is a multisystem disease that causes endothelial dysfunction and organ damage. Aim of the study was to evaluate the microvascular status in COVID-19 survivors with past different disease severity, in comparison with age and sex-matched primary Raynaud's phenomenon (PRP) patients and control subjects (CNT), including possible effects of concomitant therapies. Methods Sixty-one COVID-19 survivors (mean age 58 ± 13 years, mean days from disease onset 126 ± 53 and mean days from recovery 104 ± 53), thirty-one PRP patients (mean age 59 ± 15 years, mean disease duration 11 ± 10 years) and thirty CNT (mean age 58 ± 13 years) underwent nailfold videocapillaroscopy (NVC) examination. The following capillaroscopic parameters were searched and scored (0–3): dilated capillaries, giant capillaries, isolated microhemorrhages, capillary ramifications (angiogenesis) and capillary number, including absolute capillary number per linear millimeter at the nailfold bed. Results The mean nailfold capillary number per linear millimeter was significantly lower in COVID-19 survivors when compared with PRP patients and CNT (univariate and multivariate analysis p < 0.001). On the contrary, COVID-19 survivors showed significantly less isolated microhemorrhages than PRP patients and CNT (univariate and multivariate analysis, p = 0.005 and p = 0.012, respectively). No statistically significant difference was observed between COVID-19 survivors and control groups concerning the frequency of dilated capillaries and capillary ramifications. COVID-19 selective therapies showed a promising trend on preserving capillary loss and deserving further investigations. Conclusions SARS-CoV-2 seems to mainly induce a significant loss of capillaries in COVID-19 survivors at detailed NVC analysis in comparison to controls. The presence of a significant reduced score for isolated microhaemorrhages in COVID-19 survivors deserves further analysis.
Collapse
Affiliation(s)
- Alberto Sulli
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Specialties, University of Genova, IRCCS San Martino Polyclinic Hospital, Genova, Italy.
| | - Emanuele Gotelli
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Specialties, University of Genova, IRCCS San Martino Polyclinic Hospital, Genova, Italy
| | - Pietro Francesco Bica
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Specialties, University of Genova, IRCCS San Martino Polyclinic Hospital, Genova, Italy
| | - Irene Schiavetti
- Department of Health Sciences, University of Genova, Genova, Italy
| | - Carmen Pizzorni
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Specialties, University of Genova, IRCCS San Martino Polyclinic Hospital, Genova, Italy.
| | - Teresita Aloè
- Interventional Pneumology Unit, IRCCS San Martino Polyclinic Hospital, Genova, Italy
| | - Marco Grosso
- Interventional Pneumology Unit, IRCCS San Martino Polyclinic Hospital, Genova, Italy
| | - Emanuela Barisione
- Interventional Pneumology Unit, IRCCS San Martino Polyclinic Hospital, Genova, Italy
| | - Sabrina Paolino
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Specialties, University of Genova, IRCCS San Martino Polyclinic Hospital, Genova, Italy.
| | - Vanessa Smith
- Department of Rheumatology, Ghent University Hospital, Ghent, Belgium; Department of Internal Medicine, Ghent University Hospital, Ghent, Belgium; Unit for Molecular Immunology and Inflammation, Vlaams Instituut voor Biotechnologie (VIB), Inflammation Research Center (IRC), Ghent, Belgium.
| | - Maurizio Cutolo
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Specialties, University of Genova, IRCCS San Martino Polyclinic Hospital, Genova, Italy.
| |
Collapse
|
36
|
Martínez-Salazar B, Holwerda M, Stüdle C, Piragyte I, Mercader N, Engelhardt B, Rieben R, Döring Y. COVID-19 and the Vasculature: Current Aspects and Long-Term Consequences. Front Cell Dev Biol 2022; 10:824851. [PMID: 35242762 PMCID: PMC8887620 DOI: 10.3389/fcell.2022.824851] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/20/2022] [Indexed: 12/11/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) was first identified in December 2019 as a novel respiratory pathogen and is the causative agent of Corona Virus disease 2019 (COVID-19). Early on during this pandemic, it became apparent that SARS-CoV-2 was not only restricted to infecting the respiratory tract, but the virus was also found in other tissues, including the vasculature. Individuals with underlying pre-existing co-morbidities like diabetes and hypertension have been more prone to develop severe illness and fatal outcomes during COVID-19. In addition, critical clinical observations made in COVID-19 patients include hypercoagulation, cardiomyopathy, heart arrythmia, and endothelial dysfunction, which are indicative for an involvement of the vasculature in COVID-19 pathology. Hence, this review summarizes the impact of SARS-CoV-2 infection on the vasculature and details how the virus promotes (chronic) vascular inflammation. We provide a general overview of SARS-CoV-2, its entry determinant Angiotensin-Converting Enzyme II (ACE2) and the detection of the SARS-CoV-2 in extrapulmonary tissue. Further, we describe the relation between COVID-19 and cardiovascular diseases (CVD) and their impact on the heart and vasculature. Clinical findings on endothelial changes during COVID-19 are reviewed in detail and recent evidence from in vitro studies on the susceptibility of endothelial cells to SARS-CoV-2 infection is discussed. We conclude with current notions on the contribution of cardiovascular events to long term consequences of COVID-19, also known as “Long-COVID-syndrome”. Altogether, our review provides a detailed overview of the current perspectives of COVID-19 and its influence on the vasculature.
Collapse
Affiliation(s)
- Berenice Martínez-Salazar
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Melle Holwerda
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Chiara Stüdle
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Indre Piragyte
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Nadia Mercader
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Institute of Anatomy, University of Bern, Bern, Switzerland.,Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Bern Center of Precision Medicine BCPM, University of Bern, Bern, Switzerland
| | | | - Robert Rieben
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Yvonne Döring
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
37
|
Multiorgan and Vascular Tropism of SARS-CoV-2. Viruses 2022; 14:v14030515. [PMID: 35336922 PMCID: PMC8955556 DOI: 10.3390/v14030515] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 12/15/2022] Open
Abstract
Although the respiratory tract is the main target of SARS-CoV-2, other tissues and organs are permissive to the infection. In this report, we investigated this wide-spectrum tropism by studying the SARS-CoV-2 genetic intra-host variability in multiple tissues. The virological and histological investigation of multiple specimens from a post-mortem COVID-19 patient was performed. SARS-CoV-2 genome was detected in several tissues, including the lower respiratory system, cardio-vascular biopsies, stomach, pancreas, adrenal gland, mediastinal ganglion and testicles. Subgenomic RNA transcripts were also detected, in favor of an active viral replication, especially in testicles. Ultra-deep sequencing allowed us to highlight several SARS-CoV-2 mutations according to tissue distribution. More specifically, mutations of the spike protein, i.e., V341A (18.3%), E654 (44%) and H655R (30.8%), were detected in the inferior vena cava. SARS-CoV-2 variability can contribute to heterogeneous distributions of viral quasispecies, which may affect the COVID-19 pathogeny.
Collapse
|
38
|
Ning Q, Wu D, Wang X, Xi D, Chen T, Chen G, Wang H, Lu H, Wang M, Zhu L, Hu J, Liu T, Ma K, Han M, Luo X. The mechanism underlying extrapulmonary complications of the coronavirus disease 2019 and its therapeutic implication. Signal Transduct Target Ther 2022; 7:57. [PMID: 35197452 PMCID: PMC8863906 DOI: 10.1038/s41392-022-00907-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/10/2022] [Accepted: 01/17/2022] [Indexed: 02/06/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) is a highly transmissible disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that poses a major threat to global public health. Although COVID-19 primarily affects the respiratory system, causing severe pneumonia and acute respiratory distress syndrome in severe cases, it can also result in multiple extrapulmonary complications. The pathogenesis of extrapulmonary damage in patients with COVID-19 is probably multifactorial, involving both the direct effects of SARS-CoV-2 and the indirect mechanisms associated with the host inflammatory response. Recognition of features and pathogenesis of extrapulmonary complications has clinical implications for identifying disease progression and designing therapeutic strategies. This review provides an overview of the extrapulmonary complications of COVID-19 from immunological and pathophysiologic perspectives and focuses on the pathogenesis and potential therapeutic targets for the management of COVID-19.
Collapse
Affiliation(s)
- Qin Ning
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Di Wu
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaojing Wang
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dong Xi
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Chen
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guang Chen
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongwu Wang
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiling Lu
- National Medical Center for Major Public Health Events, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Wang
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Zhu
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junjian Hu
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tingting Liu
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ke Ma
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meifang Han
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiaoping Luo
- National Medical Center for Major Public Health Events, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
39
|
Abstract
Mechanical stress from haemodynamic perturbations or interventional manipulation of epicardial coronary atherosclerotic plaques with inflammatory destabilization can release particulate debris, thrombotic material and soluble substances into the coronary circulation. The physical material obstructs the coronary microcirculation, whereas the soluble substances induce endothelial dysfunction and facilitate vasoconstriction. Coronary microvascular obstruction and dysfunction result in patchy microinfarcts accompanied by an inflammatory reaction, both of which contribute to progressive myocardial contractile dysfunction. In clinical studies, the benefit of protection devices to retrieve atherothrombotic debris during percutaneous coronary interventions has been modest, and the treatment of microembolization has mostly relied on antiplatelet and vasodilator agents. The past 25 years have witnessed a relative proportional increase in non-ST-segment elevation myocardial infarction in the presentation of acute coronary syndromes. An associated increase in the incidence of plaque erosion rather than rupture has also been recognized as a key mechanism in the past decade. We propose that coronary microembolization is a decisive link between plaque erosion at the culprit lesion and the manifestation of non-ST-segment elevation myocardial infarction. In this Review, we characterize the features and mechanisms of coronary microembolization and discuss the clinical trials of drugs and devices for prevention and treatment.
Collapse
Affiliation(s)
- Petra Kleinbongard
- grid.5718.b0000 0001 2187 5445Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| | - Gerd Heusch
- grid.5718.b0000 0001 2187 5445Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| |
Collapse
|
40
|
Myocardial Damage by SARS-CoV-2: Emerging Mechanisms and Therapies. Viruses 2021; 13:v13091880. [PMID: 34578462 PMCID: PMC8473126 DOI: 10.3390/v13091880] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/06/2021] [Accepted: 09/18/2021] [Indexed: 01/01/2023] Open
Abstract
Evidence is emerging that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can infect various organs of the body, including cardiomyocytes and cardiac endothelial cells in the heart. This review focuses on the effects of SARS-CoV-2 in the heart after direct infection that can lead to myocarditis and an outline of potential treatment options. The main points are: (1) Viral entry: SARS-CoV-2 uses specific receptors and proteases for docking and priming in cardiac cells. Thus, different receptors or protease inhibitors might be effective in SARS-CoV-2-infected cardiac cells. (2) Viral replication: SARS-CoV-2 uses RNA-dependent RNA polymerase for replication. Drugs acting against ssRNA(+) viral replication for cardiac cells can be effective. (3) Autophagy and double-membrane vesicles: SARS-CoV-2 manipulates autophagy to inhibit viral clearance and promote SARS-CoV-2 replication by creating double-membrane vesicles as replication sites. (4) Immune response: Host immune response is manipulated to evade host cell attacks against SARS-CoV-2 and increased inflammation by dysregulating immune cells. Efficiency of immunosuppressive therapy must be elucidated. (5) Programmed cell death: SARS-CoV-2 inhibits programmed cell death in early stages and induces apoptosis, necroptosis, and pyroptosis in later stages. (6) Energy metabolism: SARS-CoV-2 infection leads to disturbed energy metabolism that in turn leads to a decrease in ATP production and ROS production. (7) Viroporins: SARS-CoV-2 creates viroporins that lead to an imbalance of ion homeostasis. This causes apoptosis, altered action potential, and arrhythmia.
Collapse
|
41
|
What the HEC happens around the heart during COVID-19? Basic Res Cardiol 2021; 116:43. [PMID: 34223971 PMCID: PMC8256399 DOI: 10.1007/s00395-021-00883-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 06/01/2021] [Indexed: 12/15/2022]
|