1
|
Bugatti A, Zani A, Bardelli M, Giovanetti M, Ravelli C, Ciccozzi M, Caruso A, Caccuri F. Heparan sulfate proteoglycans remodel SARS-CoV-2 spike conformation to allow integrin interaction and infection of endothelial cells. Front Cell Infect Microbiol 2025; 15:1552116. [PMID: 40248367 PMCID: PMC12003327 DOI: 10.3389/fcimb.2025.1552116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 03/17/2025] [Indexed: 04/19/2025] Open
Abstract
SARS-CoV-2 infects ACE2-negative primary HL-mECs through the interaction of an RGD motif, included in all spike proteins, up to the Omicron BA.1 subvariant, with αvβ3 integrin. Following its entry, SARS-CoV-2 remodels ECs phenotype and promotes angiogenesis in the absence of productive viral replication. Moreover, lack of spike/αvβ3 interaction, occurring in Omicron BA.5 which contains the D405N mutation in the RGD motif, inhibits HL-mECs infection and dysfunction. It is worth noting that anti-spike antibodies do not impact SARS-CoV-2 entry into HL-mECs. This data highlights the fact that i) the RGD motif is not exposed in the entire spike protein and ii) the need of a cofactor favoring spike/αvβ3 interaction. HSPGs are used by different viruses as receptors and coreceptors for their entry into host cells. Here, we use different approaches to scrutinize the role exerted by HSPGs in favoring SARS-CoV-2 infection of ECs. We highlight HSPGs as key molecules responsible for RGD exposure allowing its binding to the αvβ3 integrin as the first step toward viral entry by endocytosis. Indeed, SPR analysis showed lack of spike/αvβ3 interaction in the absence of heparin. This data was further corroborated by immunofluorescence and infectivity assays. Interestingly, the use of Heparinase III or sodium chlorate counteracts the release of proangiogenic molecules and inhibits signaling pathways induced by SARS-CoV-2 infection. Thus, HSPGs may represent a target for preventing SARS-CoV-2 infection of ECs and EC dysfunction-related COVID-19 severity.
Collapse
Affiliation(s)
- Antonella Bugatti
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Alberto Zani
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marta Bardelli
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marta Giovanetti
- Unit of Medical Statistics and Molecular Epidemiology, University Campus Bio-Medico of Rome, Rome, Italy
| | - Cosetta Ravelli
- Section of General Pathology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Massimo Ciccozzi
- Unit of Medical Statistics and Molecular Epidemiology, University Campus Bio-Medico of Rome, Rome, Italy
| | - Arnaldo Caruso
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Centre for Advanced Medical and Pharmaceutical Research, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, Targu Mures, Romania
| | - Francesca Caccuri
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Centre for Advanced Medical and Pharmaceutical Research, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, Targu Mures, Romania
| |
Collapse
|
2
|
Pandian K, Postma R, van Zonneveld AJ, Harms A, Hankemeier T. Microvessels-on-chip: Exploring endothelial cells and COVID-19 plasma interaction with nitric oxide metabolites. Nitric Oxide 2025; 155:12-19. [PMID: 39788280 DOI: 10.1016/j.niox.2025.01.002] [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: 09/14/2024] [Revised: 11/25/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
COVID-19, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), primarily manifests as a flu-like illness with lung injury, often necessitating supplemental oxygen. Elderly individuals and those with pre-existing cardiovascular diseases are at increased risk of mortality. The endothelial barrier disruption observed in patients indicates systemic viral invasion and widespread endotheliitis. Endothelial dysfunction, characterized by impaired nitric oxide (NO) production, contributes to vasoconstriction, inflammation, and coagulation abnormalities seen in COVID-19. In this study, we investigated the impact of COVID-19 patient-derived plasma on the endothelium through NO metabolite analysis using an in vitro 3D micro vessel model. Our experiments revealed alterations in NO metabolites in response to COVID-19 patient plasma perfusion, with BH4+BH2 supplementation improving citrulline levels in severe COVID-19 patient models. Positive correlation between arginase activity and eNOS activity was observed in the severe COVID-19 patient model but not in the mild COVID-19 patient model. These findings underscore the importance of endothelial dysfunction in COVID-19 pathogenesis and highlight potential therapeutic targets for mitigating vascular complications associated with severe infection.
Collapse
Affiliation(s)
- Kanchana Pandian
- Division of Systems Biomedicine and Pharmacology, LACDR, Leiden University, the Netherlands
| | - Rudmer Postma
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center (LUMC), the Netherlands
| | - Anton Jan van Zonneveld
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center (LUMC), the Netherlands
| | - Amy Harms
- Division of Systems Biomedicine and Pharmacology, LACDR, Leiden University, the Netherlands.
| | - Thomas Hankemeier
- Division of Systems Biomedicine and Pharmacology, LACDR, Leiden University, the Netherlands
| |
Collapse
|
3
|
Baissary J, Koberssy Z, Durieux JC, Atieh O, Daher J, Ailstock K, Labbato D, Foster T, Rodgers MA, Merheb A, Funderburg NT, McComsey GA. The Effect of COVID-19 on Arterial Stiffness and Inflammation: A Longitudinal Prospective Study. Viruses 2025; 17:394. [PMID: 40143322 PMCID: PMC11945347 DOI: 10.3390/v17030394] [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: 12/30/2024] [Revised: 03/05/2025] [Accepted: 03/08/2025] [Indexed: 03/28/2025] Open
Abstract
Data are limited for assessing the effect of COVID infection on endothelial function, pre- and post-pandemic. The objective of this study was to assess changes in pre-pandemic cardiovascular parameters after COVID-19 infection. This prospective cohort study used EndoPAT2000 Itamar Medical Ltd., Caesarea, Israel, to measure the augmentation index (AI; arterial elasticity) and reactive hyperemic index (RHI; endothelial function). Markers of endothelial function, inflammation, and gut integrity were collected at pre- and post-pandemic visits. COVID-negative and COVID-positive participants were matched on pre-pandemic covariates, and AI ≥ 5.0 was defined as having worse AI. Among the 156 participants, 50% had documented COVID-19 infection. Groups were balanced (p > 0.05) on pre-pandemic characteristics. Increases in oxLDL (p = 0.03) were observed in the COVID-positive group, and COVID infection had a negative effect on inflammatory markers (sVCAM-1, sTNF-RI, sTNF-RII, sCD14) and gut integrity (I-FABP, BDG) compared to COVID-negative participants (p < 0.05). There was a 16.7% (p = 0.02) increase in the proportion of COVID-positive participants with AI ≥ 5.0, without a significant change (p = 0.09) among the COVID-negative group. COVID-positive status, female sex, and higher IL-6 and sCD163 were associated (p < 0.05) with an increase in having worse AI. COVID infection is independently associated with arterial stiffness. For COVID survivors, female sex and higher markers of inflammation were associated with arterial stiffness.
Collapse
Affiliation(s)
- Jhony Baissary
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (J.B.); (Z.K.); (O.A.); (J.D.)
| | - Ziad Koberssy
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (J.B.); (Z.K.); (O.A.); (J.D.)
| | - Jared C. Durieux
- Clinical Research Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; (J.C.D.); (D.L.); (T.F.); (M.A.R.)
| | - Ornina Atieh
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (J.B.); (Z.K.); (O.A.); (J.D.)
| | - Joviane Daher
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (J.B.); (Z.K.); (O.A.); (J.D.)
| | - Kate Ailstock
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA; (K.A.); (N.T.F.)
| | - Danielle Labbato
- Clinical Research Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; (J.C.D.); (D.L.); (T.F.); (M.A.R.)
| | - Theresa Foster
- Clinical Research Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; (J.C.D.); (D.L.); (T.F.); (M.A.R.)
| | - Michael A. Rodgers
- Clinical Research Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; (J.C.D.); (D.L.); (T.F.); (M.A.R.)
| | - Alexander Merheb
- Harvard Faculty of Arts and Sciences, Harvard University, Cambridge, MA 02138, USA;
| | - Nicholas T. Funderburg
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA; (K.A.); (N.T.F.)
| | - Grace A. McComsey
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (J.B.); (Z.K.); (O.A.); (J.D.)
- Clinical Research Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; (J.C.D.); (D.L.); (T.F.); (M.A.R.)
| |
Collapse
|
4
|
Liang KH, Chen YC, Hsu CY, Kao ZK, Tsai PH, Huang HY, Chu YC, Ho HL, Liao YC, Lee YC, Huang CC, Wei TC, Liao YJ, Lu YH, Kuo CT, Chiou SH. Predictive biosignatures for hospitalization in patients with virologically confirmed COVID-19. J Chin Med Assoc 2025; 88:246-252. [PMID: 39730204 DOI: 10.1097/jcma.0000000000001203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus, presents with varying severity among individuals. Both viral and host factors can influence the severity of acute and chronic COVID-19, with chronic COVID-19 commonly referred to as long COVID. SARS-CoV-2 infection can be properly diagnosed by performing real-time reverse transcription polymerase chain reaction analysis of nasal swab samples. Pulse oximetry, chest X-ray, and complete blood count (CBC) analysis can be used to assess the condition of the patient to ensure that the appropriate medical care is delivered. This study aimed to develop biosignatures that can be used to distinguish between patients who are likely to develop severe disease and require hospitalization from patients who can be safely monitored in less intensive settings. METHODS A retrospective investigation was conducted on 7897 adult patients with virologically confirmed SARS-CoV-2 infection between January 26, 2020, and November 30, 2023; all patients underwent comprehensive CBC testing at Taipei Veterans General Hospital. Among them, 1867 patients were independently recruited for a population study involving genome-wide genotyping of approximately 424 000 genomic variants. Therefore, the participants were divided into two patient cohorts, one with genomic data (n = 1867) and one without (n = 6030) for model validation and training, respectively. RESULTS We constructed and validated a biosignature model by using a combination of CBC measurements to predict subsequent hospitalization events (hazard ratio = 3.38, 95% confidence interval: 3.07-3.73 for the training cohort and 3.03 [2.46-3.73] for the validation cohort; both p < 10 -8 ). The obtained scores were used to identify the top quartile of patients, who formed the "very high risk" group with a significantly higher cumulative incidence of hospitalization (log-rank p < 10 -8 in both the training and validation cohorts). The "very high risk" group exhibited a cumulative hospitalization rate of >60%, whereas the rate for the other patients was approximately 30% over a 1.5-year period, providing a binary classification of patients with distinct hospitalization risks. To investigate the genetic factors mediating this risk, we conducted a genome-wide association study. Specific regions in chromosomes 7 and 10 and the mitochondrial chromosome (M), harboring IKAROS family zinc finger 1 ( IKZF1 ), actin binding LIM protein 1 ( ABLIM1 ), and mitochondrially encoded NADH:ubiquinone oxidoreductase core subunit 3 ( MT-ND3 ), exhibited prominent associations with binary risk classification. The identified exonic variants of IKZF1 are linked to several autoimmune diseases. Notably, people with different genotypes of the leading variants (rs4132601, rs141492519, and Affx-120744614) exhibited varying cumulative hospitalization rates after infection. CONCLUSION We successfully developed and validated a biosignature model of COVID-19 severe disease in virologically confirmed patients. The identified genomic variants provide new insights for infectious disease research and medical care.
Collapse
Affiliation(s)
- Kung-Hao Liang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Biosafety Level 3 Laboratory, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- College of Medicine, Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- School of Pharmaceutical Sciences, Institute of Food Safety and Health Risk Assessment, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Yu-Chun Chen
- Department of Family Medicine, Taipei Veterans General Hospital, Yuli Branch, Hualien, Taiwan, ROC
- Department of Medical Research, Big Data Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Chun-Yi Hsu
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Medical Research, Big Data Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Zih-Kai Kao
- Department of Medical Research, Big Data Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Information Management, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Ping-Hsing Tsai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Medical Research, Big Data Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Hsin-Yi Huang
- Department of Medical Research, Big Data Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Information Management, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yuan-Chia Chu
- Department of Medical Research, Big Data Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Information Management, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Information Management, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan, ROC
| | - Hsiang-Ling Ho
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Yi-Chu Liao
- Departments of Neurology, Taipei Veterans General Hospital, Taiwan, ROC
- Department of Neurology, School of Medicine, National Yang Ming Chao Tung University, Taipei, Taiwan, ROC
- College of Medicine, Brain Research Center, National Yang Ming Chao Tung University School of Medicine, Taipei, Taiwan, ROC
| | - Yi-Chung Lee
- Departments of Neurology, Taipei Veterans General Hospital, Taiwan, ROC
- Department of Neurology, School of Medicine, National Yang Ming Chao Tung University, Taipei, Taiwan, ROC
- College of Medicine, Brain Research Center, National Yang Ming Chao Tung University School of Medicine, Taipei, Taiwan, ROC
| | - Chi-Cheng Huang
- Department of Surgery, Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Division of Breast Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- College of Public Health, Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Tzu-Chun Wei
- Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Urology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- College of Medicine, Shu-Tien Urological Institute, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Yi-Jia Liao
- Departments of Neurology, Taipei Veterans General Hospital, Taiwan, ROC
| | - Yung-Hsiu Lu
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Chen-Tsung Kuo
- Department of Information Management, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| |
Collapse
|
5
|
Shah FH, Bang JY, Nam YS, Hwang IS, Kim DH, Ki M, Salman S, Lee HW. Understanding the Impact of SARS-CoV-2 on Lung Endothelial Cells: Brief Mechanisms Unveiled. Cell Biochem Biophys 2025; 83:221-227. [PMID: 39312156 DOI: 10.1007/s12013-024-01529-w] [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] [Accepted: 09/07/2024] [Indexed: 03/03/2025]
Abstract
As the world grapples with the coronavirus-19 (COVID) pandemic, more reports are coming in regarding Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) in endotheliopathy. It is a vascular condition in which endothelial cell injury or damage inflicts anatomical and functional changes in the endothelium, significantly impacting the physiological process and function. Previously, it was assumed that SARS-CoV-2 infects respiratory epithelial cells via spike glycoproteins present on the surface of the virus. However, severe cases and different autopsy studies described the clandestine role of this virus in infecting endothelial cells other than epithelial cells. It was observed that SARS-CoV-2 targets the pulmonary and extrapulmonary systems to damage the microvasculature and affect respiratory functioning, resulting in the onset of endotheliopathy, thrombosis, inflammation, pulmonary edema, and fibrosis. Such deleterious events are the consequence of the hyperactive immune response initiated by the SARS-CoV-2 infection, leading to pulmonary and extrapulmonary complications. However, the molecular mechanism behind endotheliopathy and other complications caused by this virus is elusive and will be unraveled by covering recent literature in this mini-review.
Collapse
Affiliation(s)
- Fahad Hassan Shah
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Jun Young Bang
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
- Department of Biochemical Engineering, Chosun University, Gwangju, 61452, Republic of Korea
| | - Yoon Seok Nam
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - In Seo Hwang
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
- Department of Biochemical Engineering, Chosun University, Gwangju, 61452, Republic of Korea
| | - Dae Hong Kim
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
- Department of Biochemical Engineering, Chosun University, Gwangju, 61452, Republic of Korea
| | - Minkyoung Ki
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Saad Salman
- Department of Pharmacy, CECOS University of IT & Emerging Sciences, Peshawar, Pakistan
| | - Heon-Woo Lee
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea.
| |
Collapse
|
6
|
Salazar AS, Vincent L, Ebner B, Nogueira NF, Krauss L, Meyer MS, Grant J, Aguilar N, Pester MS, Parker M, Gonzalez A, Mendez A, Carrico A, Hurwitz BE, Alcaide ML, Martinez C. Macrovascular Function in People with HIV After Recent SARS-CoV-2 Infection. JOURNAL OF VASCULAR DISEASES 2025; 4:4. [PMID: 40110487 PMCID: PMC11922556 DOI: 10.3390/jvd4010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Background People with HIV (PWH) are at increased risk of vascular dysfunction and cardiovascular disease (CVD). SARS-CoV-2 infection has been associated with acute CVD complications. The aim of the study was to as-sess macrovascular function as an early indicator of CVD risk in PWH after mild SARS-CoV-2 infection. Methods PWH aged 20-60 years, with undetectable viral load (RNA < 20 copies/mL), on stable antiretroviral therapy (≥6 months) and history of mild COVID-19 (≥30 days) without any CVD manifestations prior to enrollment were recruited. Participants were excluded if they had history of diabetes mellitus, end-stage renal disease, heart or respiratory disease. Participants were matched 1:1 to pre-pandemic PWH. A health survey, surrogate measures of CVD risk, and macrovascular function (brachial artery flow-mediated vasodilation and arterial stiffness assessments via applanation tonometry) were compared between group. Results A total of 17 PWH and history of COVID-19 (PWH/COV+) were matched with 17 PWH without COVID-19 (PWH/COV-) pre-pandemic. Mean age (45.5 years), sex (76.5% male), body mass index (27.3), and duration of HIV infection (12.2 years) were not different between groups. Both groups had comparable CVD risk factors (total cholesterol, LDL, HDL, systolic and diastolic blood pressure). There were no differences in measures of flow mediated arterial dilatation or arterial stiffness after 30 days of SARS-CoV-2 infection. Conclusions After recent SARS-CoV-2 infection, PWH did not demonstrate evidence of macrovascular dysfunction and increased CVD risk. Results suggest that CVD risk may not be increased in people with well-controlled HIV who did not manifest CVD complications SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Ana S Salazar
- Division of Infectious Diseases, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Internal Medicine, University of Miami/Jackson Memorial Hospital, Miami, FL 33101, USA
| | - Louis Vincent
- Division of Cardiovascular Medicine, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Bertrand Ebner
- Division of Cardiovascular Medicine, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Nicholas Fonseca Nogueira
- Division of Infectious Diseases, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Leah Krauss
- Division of Infectious Diseases, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Madison S Meyer
- Division of Infectious Diseases, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Medicine, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Jelani Grant
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Natalie Aguilar
- Division of Cardiovascular Medicine, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Mollie S Pester
- Behavioral Medicine Research Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Psychology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Meela Parker
- Behavioral Medicine Research Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Psychology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Alex Gonzalez
- Behavioral Medicine Research Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Psychology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Armando Mendez
- Division of Endocrinology, Diabetes and Metabolism, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Adam Carrico
- Department of Public Health, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Barry E Hurwitz
- Behavioral Medicine Research Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Psychology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Division of Endocrinology, Diabetes and Metabolism, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Maria L Alcaide
- Division of Infectious Diseases, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Claudia Martinez
- Division of Cardiovascular Medicine, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
7
|
du Preez HN, Lin J, Maguire GEM, Aldous C, Kruger HG. COVID-19 vaccine adverse events: Evaluating the pathophysiology with an emphasis on sulfur metabolism and endotheliopathy. Eur J Clin Invest 2024; 54:e14296. [PMID: 39118373 DOI: 10.1111/eci.14296] [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: 04/02/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024]
Abstract
In this narrative review, we assess the pathophysiology of severe adverse events that presented after vaccination with DNA and mRNA vaccines against COVID-19. The focus is on the perspective of an undersulfated and degraded glycocalyx, considering its impact on immunomodulation, inflammatory responses, coagulation and oxidative stress. The paper explores various factors that lead to glutathione and inorganic sulfate depletion and their subsequent effect on glycocalyx sulfation and other metabolites, including hormones. Components of COVID-19 vaccines, such as DNA and mRNA material, spike protein antigen and lipid nanoparticles, are involved in possible cytotoxic effects. The common thread connecting these adverse events is endotheliopathy or glycocalyx degradation, caused by depleted glutathione and inorganic sulfate levels, shear stress from circulating nanoparticles, aggregation and formation of protein coronas; leading to imbalanced immune responses and chronic release of pro-inflammatory cytokines, ultimately resulting in oxidative stress and systemic inflammatory response syndrome. By understanding the underlying pathophysiology of severe adverse events, better treatment options can be explored.
Collapse
Affiliation(s)
- Heidi N du Preez
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Durban, South Africa
- College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Johnson Lin
- School of Life Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Glenn E M Maguire
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Durban, South Africa
- School of Chemistry and Physics, University of KwaZulu-Natal, Durban, South Africa
| | - Colleen Aldous
- College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Hendrik G Kruger
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
8
|
Georgieva E, Ananiev J, Yovchev Y, Arabadzhiev G, Abrashev H, Abrasheva D, Atanasov V, Kostandieva R, Mitev M, Petkova-Parlapanska K, Karamalakova Y, Koleva-Korkelia I, Tsoneva V, Nikolova G. COVID-19 Complications: Oxidative Stress, Inflammation, and Mitochondrial and Endothelial Dysfunction. Int J Mol Sci 2023; 24:14876. [PMID: 37834324 PMCID: PMC10573237 DOI: 10.3390/ijms241914876] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 09/28/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
SARS-CoV-2 infection, discovered and isolated in Wuhan City, Hubei Province, China, causes acute atypical respiratory symptoms and has led to profound changes in our lives. COVID-19 is characterized by a wide range of complications, which include pulmonary embolism, thromboembolism and arterial clot formation, arrhythmias, cardiomyopathy, multiorgan failure, and more. The disease has caused a worldwide pandemic, and despite various measures such as social distancing, various preventive strategies, and therapeutic approaches, and the creation of vaccines, the novel coronavirus infection (COVID-19) still hides many mysteries for the scientific community. Oxidative stress has been suggested to play an essential role in the pathogenesis of COVID-19, and determining free radical levels in patients with coronavirus infection may provide an insight into disease severity. The generation of abnormal levels of oxidants under a COVID-19-induced cytokine storm causes the irreversible oxidation of a wide range of macromolecules and subsequent damage to cells, tissues, and organs. Clinical studies have shown that oxidative stress initiates endothelial damage, which increases the risk of complications in COVID-19 and post-COVID-19 or long-COVID-19 cases. This review describes the role of oxidative stress and free radicals in the mediation of COVID-19-induced mitochondrial and endothelial dysfunction.
Collapse
Affiliation(s)
- Ekaterina Georgieva
- Department of General and Clinical Pathology, Forensic Medicine, Deontology and Dermatovenerology, Medical Faculty, Trakia University, 11 Armeiska Str., 6000 Stara Zagora, Bulgaria;
| | - Julian Ananiev
- Department of General and Clinical Pathology, Forensic Medicine, Deontology and Dermatovenerology, Medical Faculty, Trakia University, 11 Armeiska Str., 6000 Stara Zagora, Bulgaria;
| | - Yovcho Yovchev
- Department of Surgery and Anesthesiology, University Hospital “Prof. Dr. St. Kirkovich”, 6000 Stara Zagora, Bulgaria; (Y.Y.); (G.A.)
| | - Georgi Arabadzhiev
- Department of Surgery and Anesthesiology, University Hospital “Prof. Dr. St. Kirkovich”, 6000 Stara Zagora, Bulgaria; (Y.Y.); (G.A.)
| | - Hristo Abrashev
- Department of Vascular Surgery, Medical Faculty, Trakia University, 11 Armeiska Str., 6000 Stara Zagora, Bulgaria;
| | - Despina Abrasheva
- II Department of Internal Medicine Therapy: Cardiology, Rheumatology, Hematology and Gastroenterology, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria;
| | - Vasil Atanasov
- Forensic Toxicology Laboratory, Military Medical Academy, 3 G. Sofiiski, 1606 Sofia, Bulgaria; (V.A.); (R.K.)
| | - Rositsa Kostandieva
- Forensic Toxicology Laboratory, Military Medical Academy, 3 G. Sofiiski, 1606 Sofia, Bulgaria; (V.A.); (R.K.)
| | - Mitko Mitev
- Department of Diagnostic Imaging, University Hospital “Prof. Dr. St. Kirkovich”, 6000 Stara Zagora, Bulgaria;
| | - Kamelia Petkova-Parlapanska
- Department of Medical Chemistry and Biochemistry, Medical Faculty, Trakia University, 11 Armeiska Str., 6000 Stara Zagora, Bulgaria; (K.P.-P.); (Y.K.)
| | - Yanka Karamalakova
- Department of Medical Chemistry and Biochemistry, Medical Faculty, Trakia University, 11 Armeiska Str., 6000 Stara Zagora, Bulgaria; (K.P.-P.); (Y.K.)
| | - Iliana Koleva-Korkelia
- Department of Obstetrics and Gynaecology Clinic, University Hospital “Prof. St. Kirkovich”, 6000 Stara Zagora, Bulgaria;
| | - Vanya Tsoneva
- Department of Propaedeutics of Internal Medicine and Clinical Laboratory, Medical Faculty, Trakia University, 11 Armeiska Str., 6000 Stara Zagora, Bulgaria;
| | - Galina Nikolova
- Department of Medical Chemistry and Biochemistry, Medical Faculty, Trakia University, 11 Armeiska Str., 6000 Stara Zagora, Bulgaria; (K.P.-P.); (Y.K.)
| |
Collapse
|
9
|
Bele A, Wagh V, Munjewar PK. A Comprehensive Review on Cardiovascular Complications of COVID-19: Unraveling the Link to Bacterial Endocarditis. Cureus 2023; 15:e44019. [PMID: 37746510 PMCID: PMC10517725 DOI: 10.7759/cureus.44019] [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: 08/04/2023] [Accepted: 08/23/2023] [Indexed: 09/26/2023] Open
Abstract
The global pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has ushered in a new era of understanding the multifaceted nature of infectious diseases. Beyond its well-documented respiratory impact, COVID-19 has unveiled intricate interactions with the cardiovascular system, with potential implications that extend to bacterial endocarditis. This review explores the complex interplay between COVID-19 and bacterial endocarditis, elucidating shared risk factors, theoretical mechanisms, and clinical implications. We examine the diverse cardiovascular manifestations of COVID-19, ranging from myocarditis and thromboembolic events to arrhythmias, and delve into the pathogenesis, clinical features, and diagnostic challenges of bacterial endocarditis. By analyzing potential connections, such as viral-induced endothelial disruption and immune modulation, we shed light on the plausible relationship between COVID-19 and bacterial endocarditis. Our synthesis highlights the significance of accurate diagnosis, optimal management, and interdisciplinary collaboration in addressing the challenges posed by these intricate interactions. In addition, we underscore the importance of future research, emphasizing prospective studies on bacterial endocarditis incidence and investigations into the long-term cardiovascular effects of COVID-19. As the boundaries of infectious diseases and cardiovascular complications converge, this review calls for continued research, vigilance, and coordinated efforts to enhance patient care and public health strategies in a rapidly evolving landscape.
Collapse
Affiliation(s)
- Anurag Bele
- Internal Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Vasant Wagh
- Community Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Pratiksha K Munjewar
- Medical Surgical Nursing, Smt. Radhikabai Meghe Memorial College of Nursing, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
10
|
Russo C, Valle MS, Malaguarnera L, Romano IR, Malaguarnera L. Comparison of Vitamin D and Resveratrol Performances in COVID-19. Nutrients 2023; 15:nu15112639. [PMID: 37299603 DOI: 10.3390/nu15112639] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023] Open
Abstract
Over the last few years, we have experienced the infection generated by severe respiratory syndrome coronavirus 2 (SARS-CoV-2) often resulting in an exaggerated immune reaction and systemic inflammation. The preferred treatments against SARS-CoV-2 were those that mitigated immunological/inflammatory dysfunction. A variety of observational epidemiological studies have reported that vitamin D deficiency is often a crucial factor in many inflammatory diseases and autoimmune diseases, as well as the susceptibility to contract infectious diseases, including acute respiratory infections. Similarly, resveratrol regulates immunity, modifying the gene expression and the release of proinflammatory cytokines in the immune cells. Therefore, it plays an immunomodulatory role that can be beneficial in the prevention and development of non-communicable diseases associated with inflammation. Since both vitamin D and resveratrol also act as immunomodulators in inflammatory pathologies, many studies have paid particular attention to an integrated treatment of either vitamin D or resveratrol in the immune reaction against SARS-CoV-2 infections. This article offers a critical evaluation of published clinical trials that have examined the use of vitamin D or resveratrol as adjuncts in COVID-19 management. Furthermore, we aimed to compare the anti-inflammatory and antioxidant properties linked to the modulation of the immune system, along with antiviral properties of both vitamin D and resveratrol.
Collapse
Affiliation(s)
- Cristina Russo
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| | - Maria Stella Valle
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| | - Luisa Malaguarnera
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| | - Ivana Roberta Romano
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| | - Lucia Malaguarnera
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| |
Collapse
|
11
|
Kukreja RC, Wang R, Koka S, Das A, Samidurai A, Xi L. Treating diabetes with combination of phosphodiesterase 5 inhibitors and hydroxychloroquine-a possible prevention strategy for COVID-19? Mol Cell Biochem 2023; 478:679-696. [PMID: 36036333 PMCID: PMC9421626 DOI: 10.1007/s11010-022-04520-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 06/30/2022] [Indexed: 01/09/2023]
Abstract
Type 2 diabetes (T2D) is one of the major risk factors for developing cardiovascular disease and the resultant devastating morbidity and mortality. The key features of T2D are hyperglycemia, hyperlipidemia, insulin resistance, and impaired insulin secretion. Patients with diabetes and myocardial infarction have worse prognosis than those without T2D. Moreover, obesity and T2D are recognized risk factors in developing severe form of COVID-19 with higher mortality rate. The current lines of drug therapy are insufficient to control T2D and its serious cardiovascular complications. Phosphodiesterase 5 (PDE5) is a cGMP specific enzyme, which is the target of erectile dysfunction drugs including sildenafil, vardenafil, and tadalafil. Cardioprotective effects of PDE5 inhibitors against ischemia/reperfusion (I/R) injury were reported in normal and diabetic animals. Hydroxychloroquine (HCQ) is a widely used antimalarial and anti-inflammatory drug and its hyperglycemia-controlling effect in diabetic patients is also under investigation. This review provides our perspective of a potential use of combination therapy of PDE5 inhibitor with HCQ to reduce cardiovascular risk factors and myocardial I/R injury in T2D. We previously observed that diabetic mice treated with tadalafil and HCQ had significantly reduced fasting blood glucose and lipid levels, increased plasma insulin and insulin-like growth factor-1 levels, and improved insulin sensitivity, along with smaller myocardial infarct size following I/R. The combination treatment activated Akt/mTOR cellular survival pathway, which was likely responsible for the salutary effects. Therefore, pretreatment with PDE5 inhibitor and HCQ may be a potentially useful therapy not only for controlling T2D but also reducing the rate and severity of COVID-19 infection in the vulnerable population of diabetics.
Collapse
Affiliation(s)
- Rakesh C Kukreja
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-020D, Box 980204, Richmond, VA, 23298-0204, USA.
| | - Rui Wang
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-020D, Box 980204, Richmond, VA, 23298-0204, USA
| | - Saisudha Koka
- Department of Microbiology, Immunology and Pharmacology, Arkansas College of Osteopathic Medicine, Fort Smith, AR, 72916-6024, USA
| | - Anindita Das
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-020D, Box 980204, Richmond, VA, 23298-0204, USA
| | - Arun Samidurai
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-020D, Box 980204, Richmond, VA, 23298-0204, USA
| | - Lei Xi
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-020D, Box 980204, Richmond, VA, 23298-0204, USA.
| |
Collapse
|
12
|
Differential transcriptomic landscapes of multiple organs from SARS-CoV-2 early infected rhesus macaques. Protein Cell 2022; 13:920-939. [PMID: 35377064 PMCID: PMC8978510 DOI: 10.1007/s13238-022-00915-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 03/08/2022] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2 infection causes complicated clinical manifestations with variable multi-organ injuries, however, the underlying mechanism, in particular immune responses in different organs, remains elusive. In this study, comprehensive transcriptomic alterations of 14 tissues from rhesus macaque infected with SARS-CoV-2 were analyzed. Compared to normal controls, SARS-CoV-2 infection resulted in dysregulation of genes involving diverse functions in various examined tissues/organs, with drastic transcriptomic changes in cerebral cortex and right ventricle. Intriguingly, cerebral cortex exhibited a hyperinflammatory state evidenced by significant upregulation of inflammation response-related genes. Meanwhile, expressions of coagulation, angiogenesis and fibrosis factors were also up-regulated in cerebral cortex. Based on our findings, neuropilin 1 (NRP1), a receptor of SARS-CoV-2, was significantly elevated in cerebral cortex post infection, accompanied by active immune response releasing inflammatory factors and signal transmission among tissues, which enhanced infection of the central nervous system (CNS) in a positive feedback way, leading to viral encephalitis. Overall, our study depicts a multi-tissue/organ transcriptomic landscapes of rhesus macaque with early infection of SARS-CoV-2, and provides important insights into the mechanistic basis for COVID-19-associated clinical complications.
Collapse
|
13
|
Perera MR, Greenwood EJD, Crozier TWM, Elder EG, Schmitt J, Crump CM, Lehner PJ, Wills MR, Sinclair JH. Human Cytomegalovirus Infection of Epithelial Cells Increases SARS-CoV-2 Superinfection by Upregulating the ACE2 Receptor. J Infect Dis 2022; 227:543-553. [PMID: 36408607 PMCID: PMC9927080 DOI: 10.1093/infdis/jiac452] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 11/14/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), has caused widespread morbidity and mortality since its onset in late 2019. Here, we demonstrate that prior infection with human cytomegalovirus (HCMV) substantially increases infection with SARS-CoV-2 in vitro. HCMV is a common herpesvirus carried by 40%-100% of the population, which can reactivate in the lung under inflammatory conditions, such as those resulting from SARS-CoV-2 infection. We show in both endothelial and epithelial cell types that HCMV infection upregulates ACE2, the SARS-CoV-2 cell entry receptor. These observations suggest that HCMV reactivation events in the lung of healthy HCMV carriers could exacerbate SARS-CoV-2 infection and subsequent COVID-19 symptoms. This effect could contribute to the disparity of disease severity seen in ethnic minorities and those with lower socioeconomic status, due to their higher CMV seroprevalence. Our results warrant further clinical investigation as to whether HCMV infection influences the pathogenesis of SARS-CoV-2.
Collapse
Affiliation(s)
- Marianne R Perera
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Edward J D Greenwood
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Thomas W M Crozier
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Elizabeth G Elder
- Department of Microbiology, National Veterinary Institute Uppsala, Sweden
| | - Janika Schmitt
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Colin M Crump
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Paul J Lehner
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Mark R Wills
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - John H Sinclair
- Correspondence: John H. Sinclair, PhD, Box 157, Level 5, Addenbrookes Hospital, Hills Road, Cambridge, CB2 0QQ, UK ()
| | | |
Collapse
|
14
|
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
|
15
|
Falcinelli E, Petito E, Gresele P. The role of platelets, neutrophils and endothelium in COVID-19 infection. Expert Rev Hematol 2022; 15:727-745. [PMID: 35930267 DOI: 10.1080/17474086.2022.2110061] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION COVID-19 is associated to an increased risk of thrombosis, as a result of a complex process that involves the activation of vascular and circulating cells, the release of soluble inflammatory and thrombotic mediators and blood clotting activation. AREAS COVERED This article reviews the pathophysiological role of platelets, neutrophils and the endothelium, and of their interactions, in the thrombotic complications of COVID-19 patients, and the current and future therapeutic approaches targeting these cell types. EXPERT OPINION Virus-induced platelet, neutrophil and endothelial cell changes are crucial triggers of the thrombotic complications and of the adverse evolution of COVID-19. Both the direct interaction with the virus and the associated cytokine storm concur to trigger cell activation in a classical thromboinflammatory vicious circle. Although heparin has proven to be an effective prophylactic and therapeutic weapon for the prevention and treatment of COVID-19-associated thrombosis, it acts downstream of the cascade of events triggered by SARS-CoV-2. The identification of specific molecular targets interrupting the thromboinflammatory cascade upstream, and more specifically acting either on the interaction of SARS-CoV-2 with blood and vascular cells or on the specific signalling mechanisms associated with their COVID-19-associated activation, might theoretically offer greater protection with potentially lesser side effects.
Collapse
Affiliation(s)
- E Falcinelli
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - E Petito
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - P Gresele
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
16
|
Ghasemzadeh M, Ahmadi J, Hosseini E. Platelet-leukocyte crosstalk in COVID-19: How might the reciprocal links between thrombotic events and inflammatory state affect treatment strategies and disease prognosis? Thromb Res 2022; 213:179-194. [PMID: 35397313 PMCID: PMC8969450 DOI: 10.1016/j.thromres.2022.03.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 03/11/2022] [Accepted: 03/28/2022] [Indexed: 01/09/2023]
Abstract
Platelet-leukocyte crosstalk is commonly manifested by reciprocal links between thrombosis and inflammation. Platelet thrombus acts as a reactive matrix that recruits leukocytes to the injury site where their massive accumulation, activation and migration promote thrombotic events while triggering inflammatory responses. As a life-threatening condition with the associations between inflammation and thrombosis, COVID-19 presents diffuse alveolar damage due to exaggerated macrophage activity and cytokine storms. These events, together with direct intracellular virus invasion lead to pulmonary vascular endothelialitis, cell membranes disruption, severe endothelial injury, and thrombosis. The developing pre-alveolar thrombus provides a hyper-reactive milieu that recruits circulating leukocytes to the injury site where their activation contributes to thrombus stabilization and thrombosis propagation, primarily through the formation of Neutrophil extracellular trap (NET). NET fragments can also circulate and deposit in further distance where they may disseminate intravascular thrombosis in severe cases of disease. Thrombi may also facilitate leukocytes migration into alveoli where their accumulation and activation exacerbate cytokine storms and tissue damage, further complicating the disease. Based on these mechanisms, whether an effective anti-inflammatory protocol can prevent thrombotic events, or on the other hand; efficient antiplatelet or anticoagulant regimens may be associated with reduced cytokine storms and tissue damage, is now of interests for several ongoing researches. Thus shedding more light on platelet-leukocyte crosstalk, the review presented here discusses the detailed mechanisms by which platelets may contribute to the pathogenesis of COVID-19, especially in severe cases where their interaction with leukocytes can intensify both inflammatory state and thrombosis in a reciprocal manner.
Collapse
Affiliation(s)
- Mehran Ghasemzadeh
- Corresponding authors at: Blood Transfusion Research Centre, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization Building, Hemmat Exp. Way, Next to the Milad Tower, Tehran, Iran
| | | | - Ehteramolsadat Hosseini
- Corresponding authors at: Blood Transfusion Research Centre, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization Building, Hemmat Exp. Way, Next to the Milad Tower, Tehran, Iran
| |
Collapse
|
17
|
COVID-19, the Pandemic of the Century and Its Impact on Cardiovascular Diseases. CARDIOLOGY DISCOVERY 2021; 1:233-258. [PMID: 34888547 PMCID: PMC8638821 DOI: 10.1097/cd9.0000000000000038] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 09/19/2021] [Indexed: 01/08/2023]
Abstract
COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection likely ranks among the deadliest diseases in human history. As with other coronaviruses, SARS-CoV-2 infection damages not only the lungs but also the heart and many other organs that express angiotensin-converting enzyme 2 (ACE2), a receptor for SARS-CoV-2. COVID-19 has upended lives worldwide. Dietary behaviors have been altered such that they favor metabolic and cardiovascular complications, while patients have avoided hospital visits because of limited resources and the fear of infection, thereby increasing out-hospital mortality due to delayed diagnosis and treatment. Clinical observations show that sex, age, and race all influence the risk for SARS-CoV-2 infection, as do hypertension, obesity, and pre-existing cardiovascular conditions. Many hospitalized COVID-19 patients suffer cardiac injury, acute coronary syndromes, or cardiac arrhythmia. SARS-CoV-2 infection may lead to cardiomyocyte apoptosis and necrosis, endothelial cell damage and dysfunction, oxidative stress and reactive oxygen species production, vasoconstriction, fibrotic and thrombotic protein expression, vascular permeability and microvascular dysfunction, heart inflammatory cell accumulation and activation, and a cytokine storm. Current data indicate that COVID-19 patients with cardiovascular diseases should not discontinue many existing cardiovascular therapies such as ACE inhibitors, angiotensin receptor blockers, steroids, aspirin, statins, and PCSK9 inhibitors. This review aims to furnish a framework relating to COVID-19 and cardiovascular pathophysiology.
Collapse
|
18
|
Carbone ML, Failla CM. Interleukin role in the regulation of endothelial cell pathological activation. VASCULAR BIOLOGY (BRISTOL, ENGLAND) 2021; 3:R96-R105. [PMID: 34870094 PMCID: PMC8630758 DOI: 10.1530/vb-21-0010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/18/2021] [Indexed: 02/06/2023]
Abstract
Interleukins (ILs) are the group of cytokines firstly identified as expressed by leukocytes and playing different immunomodulatory functions. With increasing evidence of a constant crosstalk between leukocytes and endothelial cells in the regulation of immune cell differentiation and activation, a role of ILs also in endothelial cell stimulation and vascular inflammation has been shown. ILs act on endothelial cells both in an autocrine and a paracrine manner. In fact, a cross regulation is present among ILs expressed by different cell types, leading to amplification or blocking of the initial inflammatory signal with the secretion of additional ILs or involvement of other adjacent cells and tissues. Based on selective structural features, ILs can be divided into four major groups, a fifth group comprises ILs that do not fit into any of the other four. Most of the ILs playing a role in endothelial cell activation belong to the IL1-like cytokine group, but the number of ILs involved in vascular inflammation is constantly growing, and a special contribution of IL6, IL8, and IL17 has been underlined. This review aims at presenting current knowledge and at underling missing information about the role of IL in activating endothelial cells in selected pathological settings such as tumours, psoriasis, systemic sclerosis, and viral infection.
Collapse
|
19
|
Deer RR, Rock MA, Vasilevsky N, Carmody L, Rando H, Anzalone AJ, Basson MD, Bennett TD, Bergquist T, Boudreau EA, Bramante CT, Byrd JB, Callahan TJ, Chan LE, Chu H, Chute CG, Coleman BD, Davis HE, Gagnier J, Greene CS, Hillegass WB, Kavuluru R, Kimble WD, Koraishy FM, Köhler S, Liang C, Liu F, Liu H, Madhira V, Madlock-Brown CR, Matentzoglu N, Mazzotti DR, McMurry JA, McNair DS, Moffitt RA, Monteith TS, Parker AM, Perry MA, Pfaff E, Reese JT, Saltz J, Schuff RA, Solomonides AE, Solway J, Spratt H, Stein GS, Sule AA, Topaloglu U, Vavougios GD, Wang L, Haendel MA, Robinson PN. Characterizing Long COVID: Deep Phenotype of a Complex Condition. EBioMedicine 2021; 74:103722. [PMID: 34839263 PMCID: PMC8613500 DOI: 10.1016/j.ebiom.2021.103722] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/22/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Numerous publications describe the clinical manifestations of post-acute sequelae of SARS-CoV-2 (PASC or "long COVID"), but they are difficult to integrate because of heterogeneous methods and the lack of a standard for denoting the many phenotypic manifestations. Patient-led studies are of particular importance for understanding the natural history of COVID-19, but integration is hampered because they often use different terms to describe the same symptom or condition. This significant disparity in patient versus clinical characterization motivated the proposed ontological approach to specifying manifestations, which will improve capture and integration of future long COVID studies. METHODS The Human Phenotype Ontology (HPO) is a widely used standard for exchange and analysis of phenotypic abnormalities in human disease but has not yet been applied to the analysis of COVID-19. FUNDING We identified 303 articles published before April 29, 2021, curated 59 relevant manuscripts that described clinical manifestations in 81 cohorts three weeks or more following acute COVID-19, and mapped 287 unique clinical findings to HPO terms. We present layperson synonyms and definitions that can be used to link patient self-report questionnaires to standard medical terminology. Long COVID clinical manifestations are not assessed consistently across studies, and most manifestations have been reported with a wide range of synonyms by different authors. Across at least 10 cohorts, authors reported 31 unique clinical features corresponding to HPO terms; the most commonly reported feature was Fatigue (median 45.1%) and the least commonly reported was Nausea (median 3.9%), but the reported percentages varied widely between studies. INTERPRETATION Translating long COVID manifestations into computable HPO terms will improve analysis, data capture, and classification of long COVID patients. If researchers, clinicians, and patients share a common language, then studies can be compared/pooled more effectively. Furthermore, mapping lay terminology to HPO will help patients assist clinicians and researchers in creating phenotypic characterizations that are computationally accessible, thereby improving the stratification, diagnosis, and treatment of long COVID. FUNDING U24TR002306; UL1TR001439; P30AG024832; GBMF4552; R01HG010067; UL1TR002535; K23HL128909; UL1TR002389; K99GM145411.
Collapse
Affiliation(s)
- Rachel R Deer
- University of Texas Medical Branch, Galveston, TX, USA.
| | | | - Nicole Vasilevsky
- Center for Health AI, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Monarch Initiative
| | - Leigh Carmody
- Monarch Initiative; The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Halie Rando
- Center for Health AI, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Alfred J Anzalone
- Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Marc D Basson
- Department of Surgery, University of North Dakota School of Medicine and Health Sciences
| | - Tellen D Bennett
- Section of Informatics and Data Science, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Eilis A Boudreau
- Department of Neurology; Department of Medical Informatics & Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239
| | - Carolyn T Bramante
- Departments of Internal Medicine and Pediatrics, University of Minnesota Medical School, Minneapolis, MN 55455
| | - James Brian Byrd
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109
| | - Tiffany J Callahan
- Center for Health AI, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Lauren E Chan
- Monarch Initiative; College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, USA
| | - Haitao Chu
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN USA
| | - Christopher G Chute
- Johns Hopkins University, Schools of Medicine, Public Health, and Nursing, Baltimore, MD, USA
| | - Ben D Coleman
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA; Institute for Systems Genomics, University of Connecticut, Farmington, CT 06032, USA
| | | | - Joel Gagnier
- Departments of Orthopaedic Surgery & Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Casey S Greene
- Center for Health AI, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - William B Hillegass
- University of Mississippi Medical Center, University of Mississippi Medical Center, Jackson, MS, USA; Departments of Data Science and Medicine
| | | | - Wesley D Kimble
- West Virginia Clinical and Translational Science Institute, West Virginia University, Morgantown, WV, USA
| | | | | | - Chen Liang
- Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Feifan Liu
- Department of Population and Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, MA, USA
| | - Hongfang Liu
- Department of Artificial Intelligence and Informatics, Mayo Clinic, MN, USA
| | | | - Charisse R Madlock-Brown
- Department of Diagnostic and Health Sciences, University of Tennessee Health Science Center, 920 Madison Ave. Suite 518N, Memphis TN 38613
| | - Nicolas Matentzoglu
- Monarch Initiative; Semanticly Ltd; European Bioinformatics Institute (EMBL-EBI)
| | - Diego R Mazzotti
- Division of Medical Informatics, Department of Internal Medicine, University of Kansas Medical Center
| | - Julie A McMurry
- Center for Health AI, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Monarch Initiative
| | - Douglas S McNair
- Quantitative Sciences, Global Health Div., Gates Foundation, Seattle, WA 98109, USA
| | | | | | - Ann M Parker
- Pulmonary and Critical Care Medicine, Johns Hopkins University, Schools of Medicine, Baltimore, MD, USA
| | - Mallory A Perry
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | | | - Justin T Reese
- Monarch Initiative; Lawrence Berkeley National Laboratory
| | - Joel Saltz
- Stony Brook University; Biomedical Informatics
| | | | - Anthony E Solomonides
- Outcomes Research Network, Research Institute, NorthShore University HealthSystem, Evanston, IL 60201, USA; Institute for Translational Medicine, University of Chicago, Chicago, IL, USA
| | - Julian Solway
- Institute for Translational Medicine, University of Chicago, Chicago, IL, USA
| | - Heidi Spratt
- University of Texas Medical Branch, Galveston, TX, USA
| | - Gary S Stein
- University of Vermont Larner College of Medicine, Departments of Biochemistry and Surgery, Burlington, Vermont 05405
| | | | | | - George D Vavougios
- Department of Computer Science and Telecommunications, University of Thessaly, Papasiopoulou 2 - 4, P.C.; 131 - Galaneika, Lamia, Greece; Department of Neurology, Athens Naval Hospital 70 Deinokratous Street, P.C. 115 21 Athens, Greece; Department of Respiratory Medicine, Faculty of Medicine, University of Thessaly, Biopolis, P.C. 41500 Larissa, Greece
| | - Liwei Wang
- Department of Artificial Intelligence and Informatics, Mayo Clinic, MN, USA
| | - Melissa A Haendel
- Center for Health AI, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Monarch Initiative.
| | - Peter N Robinson
- Monarch Initiative; The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA; Institute for Systems Genomics, University of Connecticut, Farmington, CT 06032, USA.
| |
Collapse
|
20
|
Cumpstey AF, Clark AD, Santolini J, Jackson AA, Feelisch M. COVID-19: A Redox Disease-What a Stress Pandemic Can Teach Us About Resilience and What We May Learn from the Reactive Species Interactome About Its Treatment. Antioxid Redox Signal 2021; 35:1226-1268. [PMID: 33985343 DOI: 10.1089/ars.2021.0017] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Significance: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus causing coronavirus disease 2019 (COVID-19), affects every aspect of human life by challenging bodily, socioeconomic, and political systems at unprecedented levels. As vaccines become available, their distribution, safety, and efficacy against emerging variants remain uncertain, and specific treatments are lacking. Recent Advances: Initially affecting the lungs, COVID-19 is a complex multisystems disease that disturbs the whole-body redox balance and can be long-lasting (Long-COVID). Numerous risk factors have been identified, but the reasons for variations in susceptibility to infection, disease severity, and outcome are poorly understood. The reactive species interactome (RSI) was recently introduced as a framework to conceptualize how cells and whole organisms sense, integrate, and accommodate stress. Critical Issues: We here consider COVID-19 as a redox disease, offering a holistic perspective of its effects on the human body, considering the vulnerability of complex interconnected systems with multiorgan/multilevel interdependencies. Host/viral glycan interactions underpin SARS-CoV-2's extraordinary efficiency in gaining cellular access, crossing the epithelial/endothelial barrier to spread along the vascular/lymphatic endothelium, and evading antiviral/antioxidant defences. An inflammation-driven "oxidative storm" alters the redox landscape, eliciting epithelial, endothelial, mitochondrial, metabolic, and immune dysfunction, and coagulopathy. Concomitantly reduced nitric oxide availability renders the sulfur-based redox circuitry vulnerable to oxidation, with eventual catastrophic failure in redox communication/regulation. Host nutrient limitations are crucial determinants of resilience at the individual and population level. Future Directions: While inflicting considerable damage to health and well-being, COVID-19 may provide the ultimate testing ground to improve the diagnosis and treatment of redox-related stress diseases. "Redox phenotyping" of patients to characterize whole-body RSI status as the disease progresses may inform new therapeutic approaches to regain redox balance, reduce mortality in COVID-19 and other redox diseases, and provide opportunities to tackle Long-COVID. Antioxid. Redox Signal. 35, 1226-1268.
Collapse
Affiliation(s)
- Andrew F Cumpstey
- Respiratory and Critical Care Research Group, Southampton NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Anna D Clark
- Respiratory and Critical Care Research Group, Southampton NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Jérôme Santolini
- Institute for Integrative Biology of the Cell (I2BC), Biochemistry, Biophysics and Structural Biology, CEA, CNRS, Université Paris-Sud, Universite Paris-Saclay, Gif-sur-Yvette, France
| | - Alan A Jackson
- Human Nutrition, University of Southampton and University Hospital Southampton, Southampton, United Kingdom
| | - Martin Feelisch
- Respiratory and Critical Care Research Group, Southampton NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
21
|
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
|
22
|
Zhang L, Zhou L, Bao L, Liu J, Zhu H, Lv Q, Liu R, Chen W, Tong W, Wei Q, Xu Y, Deng W, Gao H, Xue J, Song Z, Yu P, Han Y, Zhang Y, Sun X, Yu X, Qin C. SARS-CoV-2 crosses the blood-brain barrier accompanied with basement membrane disruption without tight junctions alteration. Signal Transduct Target Ther 2021; 6:337. [PMID: 34489403 PMCID: PMC8419672 DOI: 10.1038/s41392-021-00719-9] [Citation(s) in RCA: 174] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/05/2021] [Accepted: 07/30/2021] [Indexed: 02/06/2023] Open
Abstract
SARS-CoV-2 has been reported to show a capacity for invading the brains of humans and model animals. However, it remains unclear whether and how SARS-CoV-2 crosses the blood–brain barrier (BBB). Herein, SARS-CoV-2 RNA was occasionally detected in the vascular wall and perivascular space, as well as in brain microvascular endothelial cells (BMECs) in the infected K18-hACE2 transgenic mice. Moreover, the permeability of the infected vessel was increased. Furthermore, disintegrity of BBB was discovered in the infected hamsters by administration of Evans blue. Interestingly, the expression of claudin5, ZO-1, occludin and the ultrastructure of tight junctions (TJs) showed unchanged, whereas, the basement membrane was disrupted in the infected animals. Using an in vitro BBB model that comprises primary BMECs with astrocytes, SARS-CoV-2 was found to infect and cross through the BMECs. Consistent with in vivo experiments, the expression of MMP9 was increased and collagen IV was decreased while the markers for TJs were not altered in the SARS-CoV-2-infected BMECs. Besides, inflammatory responses including vasculitis, glial activation, and upregulated inflammatory factors occurred after SARS-CoV-2 infection. Overall, our results provide evidence supporting that SARS-CoV-2 can cross the BBB in a transcellular pathway accompanied with basement membrane disrupted without obvious alteration of TJs.
Collapse
Affiliation(s)
- Ling Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Li Zhou
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Linlin Bao
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Jiangning Liu
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Hua Zhu
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Qi Lv
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Ruixue Liu
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Wei Chen
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Wei Tong
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Qiang Wei
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Yanfeng Xu
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Wei Deng
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Hong Gao
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Jing Xue
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Zhiqi Song
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Pin Yu
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Yunlin Han
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Yu Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Xiuping Sun
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Xuan Yu
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Chuan Qin
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China.
| |
Collapse
|
23
|
Zinellu A, Paliogiannis P, Carru C, Mangoni AA. INR and COVID-19 severity and mortality: A systematic review with meta-analysis and meta-regression. Adv Med Sci 2021; 66:372-380. [PMID: 34315012 PMCID: PMC8292100 DOI: 10.1016/j.advms.2021.07.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/08/2021] [Accepted: 07/18/2021] [Indexed: 12/16/2022]
Abstract
Objectives D-dimer elevations, suggesting a pro-thrombotic state and coagulopathy, predict adverse outcomes in coronavirus disease 2019 (COVID-19). However, the clinical significance of other coagulation markers, particularly the international normalized ratio (INR), is not well established. We conducted a systematic review and meta-analysis of the INR in COVID-19. Methods A literature search was conducted in PubMed, Web of Science and Scopus, between January 2020 and February 2021, for studies reporting INR values, measures of COVID-19 severity, and mortality (PROSPERO registration number: CRD42021241468). Results Thirty-eight studies in 7440 COVID-19 patients with low disease severity or survivor status during follow up (50 % males, mean age 57 years) and 2331 with high severity or non-survivor status (60 % males, mean age 69 years) were identified. The INR was significantly prolonged in patients with severe disease or non-survivor status than in patients with mild disease or survivor status (standard mean difference, SMD, 0.60; 95 % confidence interval, CI 0.42 to 0.77; p < 0.001). There was extreme between-study heterogeneity (I2 = 90.2 %; p < 0.001). Sensitivity analysis, performed by sequentially removing each study and re-assessing the pooled estimates, showed that the magnitude and direction of the effect size was not modified. The Begg's and Egger's t-tests did not show publication bias. In meta-regression, the SMD of the INR was significantly associated with C-reactive protein (p = 0.048) and D-dimer (p = 0.001). Conclusions Prolonged INR values were significantly associated with COVID-19 severity and mortality. Both INR prolongation and D-dimer elevations can be useful in diagnosing COVID-19-associated coagulopathy and predicting clinical outcomes.
Collapse
Affiliation(s)
- Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Panagiotis Paliogiannis
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Arduino A Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, Australia; Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, Australia.
| |
Collapse
|
24
|
Zinellu A, Mangoni AA. Cystatin C, COVID-19 severity and mortality: a systematic review and meta-analysis. J Nephrol 2021; 35:59-68. [PMID: 34390479 PMCID: PMC8363863 DOI: 10.1007/s40620-021-01139-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 08/06/2021] [Indexed: 12/23/2022]
Abstract
Background Combined markers of renal dysfunction and inflammation, e.g., cystatin C, might assist with risk stratification and clinical decisions in patients with coronavirus disease 19 (COVID-19). We conducted a systematic review and meta-analysis with meta-regression of serum cystatin C in COVID-19. Methods We searched PubMed, Web of Science and Scopus, between January 2020 and February 2021, for studies reporting serum cystatin C concentrations, measures of clinical severity and survival outcomes in hospitalized COVID-19 patients (PROSPERO registration number: CRD42021245295). Results Thirteen studies in 2510 COVID-19 patients, 1972 with low severity or survivor status and 538 with high severity or non-survivor status during follow up, were included in the meta-analysis. The pooled results showed that serum cystatin C concentrations were higher in patients with high disease severity or non-survivor status (standard mean deviation, SMD, 1.71, 95% CI 0.95 to 2.46, p < 0.001). Extreme between-study heterogeneity was observed (I2 = 97.5%, p < 0.001). Sensitivity analysis, performed by sequentially removing each study and re-assessing the pooled estimates, showed that the magnitude and direction of the effect size was not substantially modified. The Begg’s and Egger’s t tests did not show publication bias. In meta-regression, the SMD of serum cystatin C was not associated with age, proportion of males, C-reactive protein, neutrophils, lymphocytes, aspartate aminotransferase, alanine aminotransferase, albumin, creatinine, creatine kinase-MB, lactate dehydrogenase, and proportion of patients with diabetes or hypertension. Conclusions Higher concentrations of serum cystatin C were associated with higher COVID-19 severity and mortality. Graphic abstract ![]()
Collapse
Affiliation(s)
- Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Arduino A Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Bedford Park, SA, 5042, Australia. .,Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, Australia.
| |
Collapse
|
25
|
Veluswamy P, Wacker M, Stavridis D, Reichel T, Schmidt H, Scherner M, Wippermann J, Michels G. The SARS-CoV-2/Receptor Axis in Heart and Blood Vessels: A Crisp Update on COVID-19 Disease with Cardiovascular Complications. Viruses 2021; 13:1346. [PMID: 34372552 PMCID: PMC8310117 DOI: 10.3390/v13071346] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/02/2021] [Accepted: 07/06/2021] [Indexed: 01/08/2023] Open
Abstract
The SARS-CoV-2 virus causing COVID-19 disease has emerged expeditiously in the world and has been declared pandemic since March 2020, by World Health Organization (WHO). The destructive effects of SARS-CoV-2 infection are increased among the patients with pre-existing chronic conditions and, in particular, this review focuses on patients with underlying cardiovascular complications. The expression pattern and potential functions of SARS-CoV-2 binding receptors and the attributes of SARS-CoV-2 virus tropism in a physio-pathological state of heart and blood vessel are precisely described. Of note, the atheroprotective role of ACE2 receptors is reviewed. A detailed description of the possible detrimental role of SARS-CoV-2 infection in terms of vascular leakage, including endothelial glycocalyx dysfunction and bradykinin 1 receptor stimulation is concisely stated. Furthermore, the potential molecular mechanisms underlying SARS-CoV-2 induced clot formation in association with host defense components, including activation of FXIIa, complements and platelets, endothelial dysfunction, immune cell responses with cytokine-mediated action are well elaborated. Moreover, a brief clinical update on patient with COVID-19 disease with underlying cardiovascular complications and those who had new onset of cardiovascular complications post-COVID-19 disease was also discussed. Taken together, this review provides an overview of the mechanistic aspects of SARS-CoV-2 induced devastating effects, in vital organs such as the heart and vessels.
Collapse
Affiliation(s)
- Priya Veluswamy
- Heart Surgery Research, Department of Cardiothoracic Surgery, Faculty of Medicine, Otto-von-Guericke University, 39120 Magdeburg, Germany; (M.W.); (D.S.); (M.S.); (J.W.)
| | - Max Wacker
- Heart Surgery Research, Department of Cardiothoracic Surgery, Faculty of Medicine, Otto-von-Guericke University, 39120 Magdeburg, Germany; (M.W.); (D.S.); (M.S.); (J.W.)
| | - Dimitrios Stavridis
- Heart Surgery Research, Department of Cardiothoracic Surgery, Faculty of Medicine, Otto-von-Guericke University, 39120 Magdeburg, Germany; (M.W.); (D.S.); (M.S.); (J.W.)
| | - Thomas Reichel
- Department of Cardiology, Diabetology and Infectiology, Klinikum Magdeburg, 39130 Magdeburg, Germany; (T.R.); (H.S.)
| | - Hendrik Schmidt
- Department of Cardiology, Diabetology and Infectiology, Klinikum Magdeburg, 39130 Magdeburg, Germany; (T.R.); (H.S.)
| | - Maximilian Scherner
- Heart Surgery Research, Department of Cardiothoracic Surgery, Faculty of Medicine, Otto-von-Guericke University, 39120 Magdeburg, Germany; (M.W.); (D.S.); (M.S.); (J.W.)
| | - Jens Wippermann
- Heart Surgery Research, Department of Cardiothoracic Surgery, Faculty of Medicine, Otto-von-Guericke University, 39120 Magdeburg, Germany; (M.W.); (D.S.); (M.S.); (J.W.)
| | - Guido Michels
- Department of Acute and Emergency Care, Sankt Antonius-Hospital Eschweiler, 52249 Eschweiler, Germany;
| |
Collapse
|
26
|
Sušić L, Maričić L, Vincelj J, Vadoci M, Sušić T. Understanding the association between endothelial dysfunction and left ventricle diastolic dysfunction in development of coronary artery disease and heart failure. ACTA BIO-MEDICA : ATENEI PARMENSIS 2021; 92:e2021204. [PMID: 34212905 PMCID: PMC8343725 DOI: 10.23750/abm.v92i3.11495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 04/07/2021] [Indexed: 11/23/2022]
Abstract
Cardiovascular diseases (CVDs) have been the most common cause of death worldwide for decades. Until recently the most affected patients were middle-aged and elderly, predominantly men, with more frequent ST elevation myocardial infarction (STEMI) caused by obstructive coronary artery disease (CAD). However, in the last two decades we have noticed an increased incidence of ischemia with non-obstructive coronary arteries (INOCA), which includes myocardial infarction with non-obstructive coronary arteries (MINOCA) and non-myocardial infarction syndromes, such as microvascular and vasospastic angina, conditions that have been particularly pronounced in women and young adults - the population we considered low-risky till than. Therefore, it has become apparent that for this group of patients conventional methods of assessing the risk of future cardiovascular (CV) events are no longer specific and sensitive enough. Heart failure with preserved ejection fraction (HFpEF) is another disease, the incidence of which has been rising rapidly during last two decades, and predominantly affects elderly population. Although the etiology and pathophysiology of INOCA and HFpEF are complex and not fully understood, there is no doubt that the underlying cause of both conditions is endothelial dysfunction (ED) which further promotes the development of left ventricular diastolic dysfunction (LVDD). Plasma biomarkers of ED, as well as natriuretic peptides (NPs), have been intensively investigated recently, and some of them have great potential for early detection and better assessment of CV risk in the future.
Collapse
Affiliation(s)
- Livija Sušić
- Department of Internal Medicine, Osijek-Baranja County Health Center, Osijek, Croatia and Josip Juraj Strossmayer University of Osijek, Faculty of Medicine, Osijek, Croatia.
| | - Lana Maričić
- Cardiology, University Hospital Centre Osijek, Osijek, Croatia; Josip Juraj Strossmayer University of Osijek, Faculty of Medicine, Osijek, Croatia.
| | - Josip Vincelj
- Josip Juraj Strossmayer University of Osijek, Faculty of Medicine, Osijek, Croatia.
| | - Milena Vadoci
- 1Department of Internal Medicine, Osijek-Baranja County Health Center, Osijek, Croatia.
| | | |
Collapse
|
27
|
Giordo R, Zinellu A, Eid AH, Pintus G. Therapeutic Potential of Resveratrol in COVID-19-Associated Hemostatic Disorders. Molecules 2021; 26:856. [PMID: 33562030 PMCID: PMC7915700 DOI: 10.3390/molecules26040856] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 01/30/2021] [Accepted: 02/03/2021] [Indexed: 12/14/2022] Open
Abstract
Coagulation disorders, endotheliopathy and inflammation are the most common hallmarks in SARS-CoV-2 infection, largely determining COVID-19's outcome and severity. Dysfunctions of endothelial cells and platelets are tightly linked in contributing to the systemic inflammatory response that appears to be both a cause and a consequence of COVID-19-associated coagulation disorders and thrombotic events. Indeed, elevated levels of circulating inflammatory cytokines are often associated with abnormal coagulation parameters in COVID-19 patients. Although treatments with low molecular weight heparin (LMWH) have shown beneficial effects in decreasing patient mortality with severe COVID-19, additional therapeutic strategies are urgently needed. Utilizing the anti-inflammatory and anti-thrombotic properties of natural compounds may provide alternative therapeutic approaches to prevent or reduce the risk factors associated with pre-existing conditions and comorbidities that can worsen COVID-19 patients' outcomes. In this regard, resveratrol, a natural compound found in several plants and fruits such as grapes, blueberries and cranberries, may represent a promising coadjuvant for the prevention and treatment of COVID-19. By virtue of its anti-thrombotic and anti-inflammatory properties, resveratrol would be expected to lower COVID-19-associated mortality, which is well known to be increased by thrombosis and inflammation. This review analyzes and discusses resveratrol's ability to modulate vascular hemostasis at different levels targeting both primary hemostasis (interfering with platelet activation and aggregation) and secondary hemostasis (modulating factors involved in coagulation cascade).
Collapse
Affiliation(s)
- Roberta Giordo
- Department of Medical Laboratory Sciences, College of Health Sciences, and Sharjah Institute for Medical Research, University of Sharjah, University City Rd, Sharjah 27272, United Arab Emirates;
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy;
| | - Ali Hussein Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, P.O. Box 2713 Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, P.O. Box 2713 Doha, Qatar
| | - Gianfranco Pintus
- Department of Medical Laboratory Sciences, College of Health Sciences, and Sharjah Institute for Medical Research, University of Sharjah, University City Rd, Sharjah 27272, United Arab Emirates;
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy;
| |
Collapse
|