51
|
Böning D, Kuebler WM, Vogel D, Bloch W. The oxygen dissociation curve of blood in COVID-19-An update. Front Med (Lausanne) 2023; 10:1098547. [PMID: 36923010 PMCID: PMC10008909 DOI: 10.3389/fmed.2023.1098547] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 02/03/2023] [Indexed: 03/02/2023] Open
Abstract
An impressive effect of the infection with SARS-Co-19 is the impairment of oxygen uptake due to lung injury. The reduced oxygen diffusion may potentially be counteracted by an increase in oxygen affinity of hemoglobin. However, hypoxia and anemia associated with COVID-19 usually decrease oxygen affinity due to a rise in [2,3-bisphosphoglycerate]. As such, COVID-19 related changes in the oxygen dissociation curve may be critical for oxygen uptake and supply, but are hard to predict. A Pubmed search lists 14 publications on oxygen affinity in COVID-19. While some investigations show no changes, three large studies found an increased affinity that was related to a good prognosis. Exact causes remain unknown. The cause of the associated anemia in COVID-19 is under discussion. Erythrocytes with structural alterations of membrane and cytoskeleton have been observed, and virus binding to Band 3 and also to ACE2 receptors in erythroblasts has been proposed. COVID-19 presentation is moderate in many subjects suffering from sickle cell disease. A possible explanation is that COVID-19 counteracts the unfavorable large right shift of the oxygen dissociation curve in these patients. Under discussion for therapy are mainly affinity-increasing drugs.
Collapse
Affiliation(s)
- Dieter Böning
- Institute of Physiology, Charité Medical University of Berlin, Berlin, Germany
| | - Wolfgang M. Kuebler
- Institute of Physiology, Charité Medical University of Berlin, Berlin, Germany
| | - Dominik Vogel
- Klinik für Interdisziplinäre Intensivmedizin, Vivantes Humboldt-Klinikum, Berlin, Germany
| | - Wilhelm Bloch
- Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne, Cologne, Germany
| |
Collapse
|
52
|
Zinatizadeh MR, Zarandi PK, Ghiasi M, Kooshki H, Mohammadi M, Amani J, Rezaei N. Immunosenescence and inflamm-ageing in COVID-19. Ageing Res Rev 2023; 84:101818. [PMID: 36516928 PMCID: PMC9741765 DOI: 10.1016/j.arr.2022.101818] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 11/04/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
The destructive effects of coronavirus disease 2019 (COVID-19) on the elderly and people with cardiovascular disease have been proven. New findings shed light on the role of aging pathways on life span and health age. New therapies that focus on aging-related pathways may positively impact the treatment of this acute respiratory infection. Using new therapies that boost the level of the immune system can support the elderly with co-morbidities against the acute form of COVID-19. This article discusses the effect of the aging immune system against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the pathways affecting this severity of infection.
Collapse
Affiliation(s)
- Mohammad Reza Zinatizadeh
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran,Cancer Biology Signaling Pathway Interest Group (CBSPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Peyman Kheirandish Zarandi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran,Cancer Biology Signaling Pathway Interest Group (CBSPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohsen Ghiasi
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hamid Kooshki
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mozafar Mohammadi
- Applied Biotechnology Research Centre, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Jafar Amani
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
53
|
Gabrielson K, Myers S, Yi J, Gabrielson E, Jimenez IA. Comparison of Cardiovascular Pathology In Animal Models of SARS-CoV-2 Infection: Recommendations Regarding Standardization of Research Methods. Comp Med 2023; 73:58-71. [PMID: 36731878 PMCID: PMC9948900 DOI: 10.30802/aalas-cm-22-000095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/04/2022] [Accepted: 01/06/2023] [Indexed: 02/04/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged as the viral pathogen that led to the global COVID-19 pandemic that began in late 2019. Because SARS-CoV-2 primarily causes a respiratory disease, much research conducted to date has focused on the respiratory system. However, SARS-CoV-2 infection also affects other organ systems, including the cardiovascular system. In this critical analysis of published data, we evaluate the evidence of cardiovascular pathology in human patients and animals. Overall, we find that the presence or absence of cardiovascular pathology is reported infrequently in both human autopsy studies and animal models of SARS-CoV-2 infection. Moreover, in those studies that have reported cardiovascular pathology, we identified issues in their design and execution that reduce confidence in the conclusions regarding SARS-CoV-2 infection as a cause of significant cardiovascular pathology. Throughout this overview, we expand on these limitations and provide recommendations to ensure a high level of scientific rigor and reproducibility.
Collapse
Affiliation(s)
- Kathleen Gabrielson
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Stephanie Myers
- School of Veterinary Medicine, Texas Tech University, Amarillo, Texas; and
| | - Jena Yi
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Edward Gabrielson
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Isabel A Jimenez
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
54
|
Thomas TA, Qiu A, Kim CY, Gordy DE, Miller A, Tredicine M, Dzieciatkowska M, Zotti FD, Hod EA, Dâ Alessandro A, Zimring JC, Spitalnik SL, Hudson KE. Reticulocytes in donor RBC units enhance RBC alloimmunization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.25.525560. [PMID: 36747702 PMCID: PMC9900826 DOI: 10.1101/2023.01.25.525560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Although red blood cell (RBC) transfusions save lives, some patients develop clinically-significant alloantibodies against donor blood group antigens, which then have adverse effects in multiple clinical settings. Few effective measures exist to prevent RBC alloimmunization and/or eliminate alloantibodies in sensitized patients. Donor-related factors may influence alloimmunization; thus, there is an unmet clinical need to identify which RBC units are immunogenic. Repeat volunteer blood donors and donors on iron supplements have elevated reticulocyte counts compared to healthy non-donors. Early reticulocytes retain mitochondria and other components, which may act as danger signals in immune responses. Herein, we tested whether reticulocytes in donor RBC units could enhance RBC alloimmunization. Using a murine model, we demonstrate that transfusing donor RBC units with increased reticulocyte frequencies dose-dependently increase RBC alloimmunization rates and alloantibody levels. Transfusing reticulocyte-rich RBC units was associated with increased RBC clearance from the circulation and a robust proinflammatory cytokine response. As compared to previously reported post-transfusion RBC consumption patterns, erythrophagocytosis from reticulocyte-rich units was increasingly performed by splenic B cells. These data suggest that reticulocytes in a donated RBC unit impact the quality of blood transfused, are targeted to a distinct compartment, and may be an underappreciated risk factor for RBC alloimmunization.
Collapse
|
55
|
Zhang Y, Gu X, Zhou Y, Si N, Gao W, Sun B, Sun J, Li T, Wang L, Wei X, Guo S, Cui X, Bian B, Wang H, Wang L, Zhao H. An integrative analysis of Qingfei Paidu Decoction for its anti-HCoV-229E mechanism in cold and damp environment based on the pharmacokinetics, metabolomics and molecular docking technology. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 108:154527. [PMID: 36332393 PMCID: PMC9605917 DOI: 10.1016/j.phymed.2022.154527] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/08/2022] [Accepted: 10/25/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND The novel coronavirus pneumonia (COVID-19) has spread rapidly around the world. As a member against the epidemic, Qingfei Paidu Decoction (QFPDD) has been approved for the treatment of COVID-19 in China. However, its antiviral mechanism was still largely unclear. PURPOSE An integrated strategy was used to explore the antiviral mechanisms of QFPDD in cold and damp environment, including pharmacokinetic (PK), network pharmacology, metabolomics and protein verification. METHODS Firstly, the pharmacokinetic study of the prototype absorbed ingredients were analyzed by UHPLC-QqQ-MS. Secondly, the metabolomics analysis of the endogenous constituents was carried out. Based on the aforementioned results, an integrated network was constructed to identify the curative components, crucial endogenous differential metabolites and related pathways. Finally, the validation tests were implemented by molecular docking and western blotting (WB). RESULTS According to the pharmacokinetic behaviors analysis of 31 components in vivo, the flavonoids presented more longer residence time and higher exposure compared with the other compounds. The efficacy and antiviral mechanism of QFPDD were verified by the poly-pharmacology, metabolomics, molecular docking and WB. For the occurrence of metabolic disorder, the change of amino acid transporters should not be neglected. Afterward, 8 curative compounds, 6 key genes and corresponding metabolic pathways were filtered by compound-reaction-enzyme-gene network. The molecular docking verified that the active ingredients bound to the relevant targets well. CONCLUSION In the present study, an in vivo comprehensive pharmacokinetic behaviors of QFPDD was analyzed for the first time. The results illustrated that QFPDD could exhibit immune regulation, anti-infection, anti-inflammation and metabolic disorder to perform a corresponding therapeutic effect. Moreover, our findings highlighted the roles of amino acid transporters in the coronavirus infection situation.
Collapse
Affiliation(s)
- Yan Zhang
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100007, China.
| | - Xinru Gu
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100007, China
| | - Yanyan Zhou
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100007, China
| | - Nan Si
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100007, China
| | - Wenya Gao
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100007, China
| | - Bo Sun
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100007, China
| | - Jing Sun
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100007, China
| | - Tao Li
- China Academy of Chinese Medical Sciences, Medical Experimental Center, Beijing 100007, China
| | - Linna Wang
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100007, China
| | - Xiaolu Wei
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100007, China
| | - Shanshan Guo
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100007, China
| | - Xiaolan Cui
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100007, China
| | - Baolin Bian
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100007, China
| | - Hongjie Wang
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100007, China.
| | - Liang Wang
- China Resources Sanjiu Medical and Pharmaceutical Co., Ltd., Shenzhen 518110, China.
| | - Haiyu Zhao
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100007, China.
| |
Collapse
|
56
|
Brandl A, Egner C, Reisser U, Lingenfelder C, Schleip R. Influence of high-energy laser therapy to the patellar tendon on its ligamentous microcirculation: An experimental intervention study. PLoS One 2023; 18:e0275883. [PMID: 36972259 PMCID: PMC10042339 DOI: 10.1371/journal.pone.0275883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 03/07/2023] [Indexed: 03/29/2023] Open
Abstract
Laser therapeutic applications, such as the use of high energy lasers (HILT), are widely used in physical therapy, but basic studies on the mechanisms of action of HILT on tendinous/ligamentous tissue are largely lacking. The aim of this study was to investigate microcirculatory changes of the patellar tendon by HILT. 21 healthy volunteers were included in the present investigation. Before and after HILT, as well as 10 minutes later, the microcirculation was measured by noninvasive laser Doppler and white light spectroscopy (O2C device). Tissue temperature was recorded at the measurement time points using thermography. Blood flow increased significantly by 86.38 arbitrary units (AU; p < 0.001) after the intervention and by 25.76 AU (p < 0.001) at follow-up. Oxygen saturation increased by 20.14% (p < 0.001) and 13.48%, respectively (p < 0.001), whereas relative hemoglobin decreased by 6.67 AU and 7.90 AU, respectively. Tendon temperature increased by 9.45° and 1.94° Celsius, respectively. Acceleration of blood flow by improving the flow properties of erythrocytes and platelets may have caused the results. HILT could be a therapeutic perspective for tendon pathologies with impaired microcirculation, although further studies are needed to validate the experimental results.
Collapse
Affiliation(s)
- Andreas Brandl
- Faculty for Psychology and Human Movement Science, Department of Sports Medicine, Institute for Human Movement Science, University of Hamburg, Hamburg, Germany
- DIPLOMA Hochschule, Bad Sooden-Allendorf, Germany
| | | | | | | | - Robert Schleip
- DIPLOMA Hochschule, Bad Sooden-Allendorf, Germany
- Department of Sport and Health Sciences, Conservative and Rehabilitative Orthopedics, Technical University of Munich, Munich, Germany
| |
Collapse
|
57
|
Mullen E, Bergin S, Healy G, Quinn J, Glavey S, Murphy PT. Red blood cells from COVID-19 patients suffer from increased oxidative stress and may have increased lactate influx. Blood Res 2022; 57:294-296. [PMID: 36579485 PMCID: PMC9812725 DOI: 10.5045/br.2022.2022084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/16/2022] [Accepted: 07/18/2022] [Indexed: 12/30/2022] Open
Affiliation(s)
- Edel Mullen
- Department of Haematology, Beaumont Hospital, Dublin, Ireland
| | - Stephen Bergin
- Department of Haematology, Beaumont Hospital, Dublin, Ireland
| | - Geraldine Healy
- Department of Haematology, Beaumont Hospital, Dublin, Ireland
| | - John Quinn
- Department of Haematology, Beaumont Hospital, Dublin, Ireland
| | - Siobhan Glavey
- Department of Haematology, Beaumont Hospital, Dublin, Ireland
| | - Philip Thomas Murphy
- Department of Haematology, Beaumont Hospital, Dublin, Ireland,Correspondence to: Philip Thomas Murphy Department of Haematology, Beaumont Hopsital, Beaumont Road Dublin 9, Ireland, E-mail:
| |
Collapse
|
58
|
Haj AK, Hasan H, Raife TJ. Heritability of Protein and Metabolite Biomarkers Associated with COVID-19 Severity: A Metabolomics and Proteomics Analysis. Biomolecules 2022; 13:46. [PMID: 36671431 PMCID: PMC9855380 DOI: 10.3390/biom13010046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/20/2022] [Accepted: 12/24/2022] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVES Prior studies have characterized protein and metabolite changes associated with SARS-CoV-2 infection; we hypothesized that these biomarkers may be part of heritable metabolic pathways in erythrocytes. METHODS Using a twin study of erythrocyte protein and metabolite levels, we describe the heritability of, and correlations among, previously identified biomarkers that correlate with COVID-19 severity. We used gene ontology and pathway enrichment analysis tools to identify pathways and biological processes enriched among these biomarkers. RESULTS Many COVID-19 biomarkers are highly heritable in erythrocytes. Among heritable metabolites downregulated in COVID-19, metabolites involved in amino acid metabolism and biosynthesis are enriched. Specific amino acid metabolism pathways (valine, leucine, and isoleucine biosynthesis; glycine, serine, and threonine metabolism; and arginine biosynthesis) are heritable in erythrocytes. CONCLUSIONS Metabolic pathways downregulated in COVID-19, particularly amino acid biosynthesis and metabolism pathways, are heritable in erythrocytes. This finding suggests that a component of the variation in COVID-19 severity may be the result of phenotypic variation in heritable metabolic pathways; future studies will be necessary to determine whether individual variation in amino acid metabolism pathways correlates with heritable outcomes of COVID-19.
Collapse
Affiliation(s)
| | | | - Thomas J. Raife
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, 3170 UW Medical Foundation Centennial Building (MFCB), Madison, WI 53705-2281, USA
| |
Collapse
|
59
|
Cui X, Chang Y, Yang C, Cong Z, Wang B, Leng Y. Development and Trends in Artificial Intelligence in Critical Care Medicine: A Bibliometric Analysis of Related Research over the Period of 2010-2021. J Pers Med 2022; 13:jpm13010050. [PMID: 36675711 PMCID: PMC9860734 DOI: 10.3390/jpm13010050] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/22/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The intensive care unit is a center for massive data collection, making it the best field to embrace big data and artificial intelligence. OBJECTIVE This study aimed to provide a literature overview on the development of artificial intelligence in critical care medicine (CCM) and tried to give valuable information about further precision medicine. METHODS Relevant studies published between January 2010 and June 2021 were manually retrieved from the Science Citation Index Expanded database in Web of Science (Clarivate), using keywords. RESULTS Research related to artificial intelligence in CCM has been increasing over the years. The USA published the most articles and had the top 10 active affiliations. The top ten active journals are bioinformatics journals and are in JCR Q1. Prediction, diagnosis, and treatment strategy exploration of sepsis, pneumonia, and acute kidney injury were the most focused topics. Electronic health records (EHRs) were the most widely used data and the "-omics" data should be integrated further. CONCLUSIONS Artificial intelligence in CCM has developed over the past decade. With the introduction of constantly growing data volume and novel data types, more investigation on artificial intelligence ethics and model correctness and extrapolation should be performed for generalization.
Collapse
Affiliation(s)
- Xiao Cui
- Department of Critical Care Medicine, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Yundi Chang
- Department of Critical Care Medicine, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Cui Yang
- Department of Critical Care Medicine, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Zhukai Cong
- Department of Anesthesiology, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Baocheng Wang
- National Science Library, Chinese Academy of Sciences, 33 Beisihuan Xilu, Haidian District, Beijing 100090, China
- School of Economics and Management, University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
- Correspondence: (B.W.); (Y.L.)
| | - Yuxin Leng
- Department of Critical Care Medicine, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
- Correspondence: (B.W.); (Y.L.)
| |
Collapse
|
60
|
Recktenwald SM, Simionato G, Lopes MGM, Gamboni F, Dzieciatkowska M, Meybohm P, Zacharowski K, von Knethen A, Wagner C, Kaestner L, D'Alessandro A, Quint S. Cross-talk between red blood cells and plasma influences blood flow and omics phenotypes in severe COVID-19. eLife 2022; 11:e81316. [PMID: 36537079 PMCID: PMC9767455 DOI: 10.7554/elife.81316] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 11/27/2022] [Indexed: 12/24/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) and can affect multiple organs, among which is the circulatory system. Inflammation and mortality risk markers were previously detected in COVID-19 plasma and red blood cells (RBCs) metabolic and proteomic profiles. Additionally, biophysical properties, such as deformability, were found to be changed during the infection. Based on such data, we aim to better characterize RBC functions in COVID-19. We evaluate the flow properties of RBCs in severe COVID-19 patients admitted to the intensive care unit by using microfluidic techniques and automated methods, including artificial neural networks, for an unbiased RBC analysis. We find strong flow and RBC shape impairment in COVID-19 samples and demonstrate that such changes are reversible upon suspension of COVID-19 RBCs in healthy plasma. Vice versa, healthy RBCs resemble COVID-19 RBCs when suspended in COVID-19 plasma. Proteomics and metabolomics analyses allow us to detect the effect of plasma exchanges on both plasma and RBCs and demonstrate a new role of RBCs in maintaining plasma equilibria at the expense of their flow properties. Our findings provide a framework for further investigations of clinical relevance for therapies against COVID-19 and possibly other infectious diseases.
Collapse
Affiliation(s)
- Steffen M Recktenwald
- Dynamics of Fluids, Department of Experimental Physics, Saarland UniversitySaarbrückenGermany
| | - Greta Simionato
- Dynamics of Fluids, Department of Experimental Physics, Saarland UniversitySaarbrückenGermany
- Institute for Clinical and Experimental Surgery, Campus University Hospital, Saarland UniversityHomburgGermany
| | - Marcelle GM Lopes
- Dynamics of Fluids, Department of Experimental Physics, Saarland UniversitySaarbrückenGermany
- Cysmic GmbHSaarbrückenGermany
| | - Fabia Gamboni
- Department of Biochemistry and Molecular Genetics, University of Colorado DenverAuroraUnited States
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado DenverAuroraUnited States
| | - Patrick Meybohm
- Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital WuerzburgWuerzburgGermany
| | - Kai Zacharowski
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital FrankfurtFrankfurtGermany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMPFrankfurtGermany
| | - Andreas von Knethen
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital FrankfurtFrankfurtGermany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMPFrankfurtGermany
| | - Christian Wagner
- Dynamics of Fluids, Department of Experimental Physics, Saarland UniversitySaarbrückenGermany
- Department of Physics and Materials Science, University of LuxembourgLuxembourg CityLuxembourg
| | - Lars Kaestner
- Dynamics of Fluids, Department of Experimental Physics, Saarland UniversitySaarbrückenGermany
- Theoretical Medicine and Biosciences, Campus University Hospital, Saarland UniversityHomburgGermany
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado DenverAuroraUnited States
| | - Stephan Quint
- Dynamics of Fluids, Department of Experimental Physics, Saarland UniversitySaarbrückenGermany
- Cysmic GmbHSaarbrückenGermany
| |
Collapse
|
61
|
He J, Liu S, Tan Q, Liu Z, Fu J, Li T, Wei C, Liu X, Mei Z, Cheng J, Wang K, Fu J. Antiviral Potential of Small Molecules Cordycepin, Thymoquinone, and N6, N6-Dimethyladenosine Targeting SARS-CoV-2 Entry Protein ADAM17. Molecules 2022; 27:molecules27249044. [PMID: 36558177 PMCID: PMC9781528 DOI: 10.3390/molecules27249044] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/06/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
COVID-19 is an acute respiratory disease caused by SARS-CoV-2 that has spawned a worldwide pandemic. ADAM17 is a sheddase associated with the modulation of the receptor ACE2 of SARS-CoV-2. Studies have revealed that malignant phenotypes of several cancer types are closely relevant to highly expressed ADAM17. However, ADAM17 regulation in SARS-CoV-2 invasion and its role on small molecules are unclear. Here, we evaluated the ADAM17 inhibitory effects of cordycepin (CD), thymoquinone (TQ), and N6, N6-dimethyladenosine (m62A), on cancer cells and predicted the anti-COVID-19 potential of the three compounds and their underlying signaling pathways by network pharmacology. It was found that CD, TQ, and m62A repressed the ADAM17 expression upon different cancer cells remarkably. Moreover, CD inhibited GFP-positive syncytia formation significantly, suggesting its potential against SARS-CoV-2. Pharmacological analysis by constructing CD-, TQ-, and m62A-based drug-target COVID-19 networks further indicated that ADAM17 is a potential target for anti-COVID-19 therapy with these compounds, and the mechanism might be relevant to viral infection and transmembrane receptors-mediated signal transduction. These findings imply that ADAM17 is of potentially medical significance for cancer patients infected with SARS-CoV-2, which provides potential new targets and insights for developing innovative drugs against COVID-19.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Kai Wang
- Correspondence: (J.C.); (K.W.); (J.F.)
| | | |
Collapse
|
62
|
Khojah HMJ, Ahmed SA, Al-Thagfan SS, Alahmadi YM, Abdou YA. The Impact of Serum Levels of Vitamin D3 and Its Metabolites on the Prognosis and Disease Severity of COVID-19. Nutrients 2022; 14:nu14245329. [PMID: 36558489 PMCID: PMC9784025 DOI: 10.3390/nu14245329] [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: 11/21/2022] [Revised: 12/10/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Vitamin D is among the increasingly consumed dietary supplements during the COVID-19 pandemic. It plays a regulatory role in the immune system and moderates the renin-angiotensin system, which is implicated in infection pathogenesis. However, the investigation of serum levels of vitamin D3 forms and their relative ratios in COVID-19 patients is worth investigation to understand the impacts of disease severity. Hence, we investigated the serum levels of vitamin D3 (cholecalciferol) and its metabolites (calcifediol and calcitriol), in addition to their relative ratios and correlations with angiotensin-converting enzyme 2 (ACE2), interleukin-6 (Il-6), and neutrophil-lymphocyte ratio (NLR) in COVID-19 patients compared with healthy controls. Oropharyngeal specimens were collected from the study subjects for polymerase chain reaction testing for COVID-19. Whole blood samples were obtained for blood count and NLR testing, and sera were used for the analysis of the levels of the vitamin and its metabolites, ACE2, and IL-6. We enrolled 103 patients and 50 controls. ACE2, Il-6, and NLR were significantly higher in the patients group (72.37 ± 18.67 vs. 32.36 ± 11.27 U/L, 95.84 ± 25.23 vs. 2.76 ± 0.62 pg/mL, and 1.61 ± 0.30 vs. 1.07 ± 0.16, respectively). Cholecalciferol, calcifediol, and calcitriol were significantly lower in patients (18.50 ± 5.36 vs. 29.13 ± 4.94 ng/mL, 14.60 ± 3.30 vs. 23.10 ± 3.02 ng/mL, and 42.90 ± 8.44 vs. 65.15 ± 7.11 pg/mL, respectively). However, their relative ratios were normal in both groups. Levels of the vitamin and metabolites were strongly positively, strongly negatively, and moderately negatively correlated with ACE2, Il-6, and NLR, respectively. COVID-19 infection severity is associated with a significant decrease in vitamin D3 and its metabolites in a parallel pattern, and with a significant increase in ACE2, Il-6, and NLR. Higher levels of vitamin D and its metabolites are potentially protective against severe infection.
Collapse
Affiliation(s)
- Hani M. J. Khojah
- Department of Clinical and Hospital Pharmacy, College of Pharmacy, Taibah University, Al Madinah Al Munawarah 30001, Saudi Arabia
| | - Sameh A. Ahmed
- Department of Pharmacognosy and Pharmaceutical Chemistry, College of Pharmacy, Taibah University, Al Madinah Al Munawarah 30001, Saudi Arabia
- Correspondence: ; Tel.: +966-54-3110057
| | - Sultan S. Al-Thagfan
- Department of Clinical and Hospital Pharmacy, College of Pharmacy, Taibah University, Al Madinah Al Munawarah 30001, Saudi Arabia
| | - Yaser M. Alahmadi
- Department of Clinical and Hospital Pharmacy, College of Pharmacy, Taibah University, Al Madinah Al Munawarah 30001, Saudi Arabia
| | | |
Collapse
|
63
|
Bizjak DA, John L, Matits L, Uhl A, Schulz SVW, Schellenberg J, Peifer J, Bloch W, Weiß M, Grüner B, Bracht H, Steinacker JM, Grau M. SARS-CoV-2 Altered Hemorheological and Hematological Parameters during One-Month Observation Period in Critically Ill COVID-19 Patients. Int J Mol Sci 2022; 23:15332. [PMID: 36499657 PMCID: PMC9735540 DOI: 10.3390/ijms232315332] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 11/24/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Hematological and hemorheological parameters are known to be altered in COVID-19; however, the value of combined monitoring in order to deduce disease severity is only scarcely examined. A total of 44 acute SARS-CoV-2-infected patients (aCOV) and 44 age-matched healthy controls (Con) were included. Blood of aCOV was sampled at admission (T0), and at day 2 (T2), day 5 (T5), day 10 (T10), and day 30 (T30) while blood of Con was only sampled once. Inter- and intra-group differences were calculated for hematological and hemorheological parameters. Except for mean cellular volume and mean cellular hemoglobin, all blood cell parameters were significantly different between aCOV and Con. During the acute disease state (T0-T5), hematological and hemorheological parameters were highly altered in aCOV; in particular, anemic conditions and increased immune cell response/inflammation, oxidative/nitrosative stress, decreased deformability, as well as increased aggregation, were observed. During treatment and convalescence until T30, almost all abnormal values of aCOV improved towards Con values. During the acute state of the COVID-19 disease, the hematological, as well as the hemorheological system, show fast and potentially pathological changes that might contribute to the progression of the disease, but changes appear to be largely reversible after four weeks. Measuring RBC deformability and aggregation, as well as oxidative stress induction, may be helpful in monitoring critically ill COVID-19 patients.
Collapse
Affiliation(s)
| | - Lucas John
- Division of Sports and Rehabilitation Medicine, University Hospital Ulm, 89075 Ulm, Germany
| | - Lynn Matits
- Division of Sports and Rehabilitation Medicine, University Hospital Ulm, 89075 Ulm, Germany
- Clinical & Biological Psychology, Institute of Psychology and Education, Ulm University, 89081 Ulm, Germany
| | - Alisa Uhl
- Division of Sports and Rehabilitation Medicine, University Hospital Ulm, 89075 Ulm, Germany
| | | | - Jana Schellenberg
- Division of Sports and Rehabilitation Medicine, University Hospital Ulm, 89075 Ulm, Germany
| | - Johannes Peifer
- Central Emergency Services, University Hospital Ulm, 89081 Ulm, Germany
| | - Wilhelm Bloch
- Institute of Cardiovascular Research and Sports Medicine, Molecular and Cellular Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany
| | - Manfred Weiß
- Clinic for Anaesthesiology and Intensive Care Medicine, University Hospital Medical School, 89081 Ulm, Germany
| | - Beate Grüner
- Department of Internal Medicine III, Division of Infectious Diseases, University Hospital Ulm, 89081 Ulm, Germany
| | - Hendrik Bracht
- Central Emergency Services, University Hospital Ulm, 89081 Ulm, Germany
| | | | - Marijke Grau
- Institute of Cardiovascular Research and Sports Medicine, Molecular and Cellular Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany
| |
Collapse
|
64
|
Papadopoulos C, Spourita E, Tentes I, Steiropoulos P, Anagnostopoulos K. Red Blood Cell Malfunction in COVID-19: Molecular Mechanisms and Therapeutic Targets. Viral Immunol 2022; 35:649-652. [PMID: 36342758 DOI: 10.1089/vim.2021.0212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The world has been facing a pandemic for the past 2 years. COVID-19 still leads to millions of deaths worldwide, while deteriorating the global economy. The need for therapeutic targets, thus, remains. Interestingly, red blood cells, apart from gas exchange, also serve as modulators of innate and adaptive immunity. This function is accommodated mainly by surface molecules (proteins, lipids, and carbohydrates) and increased antioxidant capacity. However, under the circumstances of a disease state, red blood cells can become proinflammatory cells. Recent evidence has shown that, in the context of COVID-19, erythrocytes present protein oxidation, decreased antioxidant capacity, increased glycolysis, altered membrane lipidome, increased binding of Cytosine-Guanine (CpG) DNA and complement proteins, and low CD47 levels. These changes lead to an erythrocyte-dependent inflammation, which possibly participates in the hyperinflammation status of COVID-19. The current knowledge for the dysfunction of red blood cells during COVID-19 implies that the BAND3 protein and toll-like receptor 9 are potential therapeutic targets for COVID-19.
Collapse
Affiliation(s)
- Charalampos Papadopoulos
- Laboratory of Biochemistry, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| | - Eleftheria Spourita
- Laboratory of Biochemistry, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| | - Ioannis Tentes
- Laboratory of Biochemistry, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| | - Paschalis Steiropoulos
- Department of Respiratory Medicine, Medical School, Democritus University of Thrace, University General Hospital Dragana, Alexandroupolis, Greece
| | | |
Collapse
|
65
|
Moore A, Busch MP, Dziewulska K, Francis RO, Hod EA, Zimring JC, D’Alessandro A, Page GP. Genome-wide metabolite quantitative trait loci analysis (mQTL) in red blood cells from volunteer blood donors. J Biol Chem 2022; 298:102706. [PMID: 36395887 PMCID: PMC9763692 DOI: 10.1016/j.jbc.2022.102706] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
The red blood cell (RBC)-Omics study, part of the larger NHLBI-funded Recipient Epidemiology and Donor Evaluation Study (REDS-III), aims to understand the genetic contribution to blood donor RBC characteristics. Previous work identified donor demographic, behavioral, genetic, and metabolic underpinnings to blood donation, storage, and (to a lesser extent) transfusion outcomes, but none have yet linked the genetic and metabolic bodies of work. We performed a genome-wide association (GWA) analysis using RBC-Omics study participants with generated untargeted metabolomics data to identify metabolite quantitative trait loci in RBCs. We performed GWA analyses of 382 metabolites in 243 individuals imputed using the 1000 Genomes Project phase 3 all-ancestry reference panel. Analyses were conducted using ProbABEL and adjusted for sex, age, donation center, number of whole blood donations in the past 2 years, and first 10 principal components of ancestry. Our results identified 423 independent genetic loci associated with 132 metabolites (p < 5×10-8). Potentially novel locus-metabolite associations were identified for the region encoding heme transporter FLVCR1 and choline and for lysophosphatidylcholine acetyltransferase LPCAT3 and lysophosphatidylserine 16.0, 18.0, 18.1, and 18.2; these associations are supported by published rare disease and mouse studies. We also confirmed previous metabolite GWA results for associations, including N(6)-methyl-L-lysine and protein PYROXD2 and various carnitines and transporter SLC22A16. Association between pyruvate levels and G6PD polymorphisms was validated in an independent cohort and novel murine models of G6PD deficiency (African and Mediterranean variants). We demonstrate that it is possible to perform metabolomics-scale GWA analyses with a modest, trans-ancestry sample size.
Collapse
Affiliation(s)
- Amy Moore
- Division of Biostatistics and Epidemiology, RTI International, Atlanta, Georgia, USA
| | | | - Karolina Dziewulska
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Richard O. Francis
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, USA
| | - Eldad A. Hod
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, USA
| | - James C. Zimring
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Angelo D’Alessandro
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, USA,Department of Biochemistry and Molecular Genetics, University of Colorado Denver – Anschutz Medical Campus, Aurora, CO, USA,For correspondence: Grier P. Page; Angelo D’Alessandro
| | - Grier P. Page
- Division of Biostatistics and Epidemiology, RTI International, Atlanta, Georgia, USA,For correspondence: Grier P. Page; Angelo D’Alessandro
| |
Collapse
|
66
|
Ciccosanti F, Antonioli M, Sacchi A, Notari S, Farina A, Beccacece A, Fusto M, Vergori A, D'Offizi G, Taglietti F, Antinori A, Nicastri E, Marchioni L, Palmieri F, Ippolito G, Piacentini M, Agrati C, Fimia GM. Proteomic analysis identifies a signature of disease severity in the plasma of COVID-19 pneumonia patients associated to neutrophil, platelet and complement activation. Clin Proteomics 2022; 19:38. [PMID: 36348270 PMCID: PMC9641302 DOI: 10.1186/s12014-022-09377-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 10/26/2022] [Indexed: 11/10/2022] Open
Abstract
Most patients infected with SARS-CoV-2 display mild symptoms with good prognosis, while 20% of patients suffer from severe viral pneumonia and up to 5% may require intensive care unit (ICU) admission due to severe acute respiratory syndrome, which could be accompanied by multiorgan failure.Plasma proteomics provide valuable and unbiased information about disease progression and therapeutic candidates. Recent proteomic studies have identified molecular changes in plasma of COVID-19 patients that implied significant dysregulation of several aspects of the inflammatory response accompanied by a general metabolic suppression. However, which of these plasma alterations are associated with disease severity remains only partly characterized.A known limitation of proteomic studies of plasma samples is the large difference in the macromolecule abundance, with concentration spanning at least 10 orders of magnitude. To improve the coverage of plasma contents, we performed a deep proteomic analysis of plasma from 10 COVID-19 patients with severe/fatal pneumonia compared to 10 COVID-19 patients with pneumonia who did not require ICU admission (non-ICU). To this aim, plasma samples were first depleted of the most abundant proteins, trypsin digested and peptides subjected to a high pH reversed-phase peptide fractionation before LC-MS analysis.These results highlighted an increase of proteins involved in neutrophil and platelet activity and acute phase response, which is significantly higher in severe/fatal COVID-19 patients when compared to non-ICU ones. Importantly, these changes are associated with a selective induction of complement cascade factors in severe/fatal COVID-19 patients. Data are available via ProteomeXchange with identifier PXD036491. Among these alterations, we confirmed by ELISA that higher levels of the neutrophil granule proteins DEFA3 and LCN2 are present in COVID-19 patients requiring ICU admission when compared to non-ICU and healthy donors.Altogether, our study provided an in-depth view of plasma proteome changes that occur in COVID-19 patients in relation to disease severity, which can be helpful to identify therapeutic strategies to improve the disease outcome.
Collapse
Affiliation(s)
- Fabiola Ciccosanti
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Manuela Antonioli
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Alessandra Sacchi
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Stefania Notari
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Anna Farina
- Infectious Disease-Clinical Department, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Alessia Beccacece
- Infectious Disease-Clinical Department, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Marisa Fusto
- Infectious Disease-Clinical Department, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Alessandra Vergori
- Infectious Disease-Clinical Department, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Gianpiero D'Offizi
- Infectious Disease-Clinical Department, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Fabrizio Taglietti
- Infectious Disease-Clinical Department, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Andrea Antinori
- Infectious Disease-Clinical Department, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Emanuele Nicastri
- Infectious Disease-Clinical Department, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Luisa Marchioni
- Infectious Disease-Clinical Department, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Fabrizio Palmieri
- Infectious Disease-Clinical Department, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Giuseppe Ippolito
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
- General Directorate for Research and Health Innovation, Italian Ministry of Health, Rome, Italy
| | - Mauro Piacentini
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Chiara Agrati
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy.
- Department of Hematology/Oncology and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS, Rome, Italy.
| | - Gian Maria Fimia
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy.
- Department of Molecular Medicine, University of Rome "Sapienza", Rome, Italy.
| |
Collapse
|
67
|
Abstract
PURPOSE OF THE REVIEW To discuss recent advances supporting the role of red blood cells (RBCs) in the host immune response. RECENT FINDINGS Over the last century, research has demonstrated that red blood cells exhibit functions beyond oxygen transport, including immune function. Recent work indicates that the nucleic acid sensing receptor, toll-like receptor 9 (TLR9), is expressed on the RBC surface and implicated in innate immune activation and red cell clearance during inflammatory states. In addition to this DNA-sensing role of RBCs, there is growing evidence that RBCs may influence immune function by inducing vascular dysfunction. RBC proteomics and metabolomics have provided additional insight into RBC immune function, with several studies indicating changes to RBC membrane structure and metabolism in response to severe acute respiratory syndrome coronavirus 2 infection. These structural RBC changes may even provide insight into the pathophysiology of the 'long-coronavirus disease 2019' phenomenon. Finally, evidence suggests that RBCs may influence host immune responses via complement regulation. Taken together, these recent findings indicate RBCs possess immune function. Further studies will be required to elucidate better how RBC immune function contributes to the heterogeneous host response during inflammatory states. SUMMARY The appreciation for nongas exchanging, red blood cell immune functions is rapidly growing. A better understanding of these RBC functions may provide insight into the heterogeneity observed in the host immune response to infection and inflammation.
Collapse
Affiliation(s)
- Jane Dobkin
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nilam S. Mangalmurti
- Division of Pulmonary, Allergy and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
68
|
Guntur VP, Nemkov T, de Boer E, Mohning MP, Baraghoshi D, Cendali FI, San-Millán I, Petrache I, D’Alessandro A. Signatures of Mitochondrial Dysfunction and Impaired Fatty Acid Metabolism in Plasma of Patients with Post-Acute Sequelae of COVID-19 (PASC). Metabolites 2022; 12:1026. [PMID: 36355108 PMCID: PMC9699059 DOI: 10.3390/metabo12111026] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 07/30/2023] Open
Abstract
Exercise intolerance is a major manifestation of post-acute sequelae of severe acute respiratory syndrome coronavirus infection (PASC, or "long-COVID"). Exercise intolerance in PASC is associated with higher arterial blood lactate accumulation and lower fatty acid oxidation rates during graded exercise tests to volitional exertion, suggesting altered metabolism and mitochondrial dysfunction. It remains unclear whether the profound disturbances in metabolism that have been identified in plasma from patients suffering from acute coronavirus disease 2019 (COVID-19) are also present in PASC. To bridge this gap, individuals with a history of previous acute COVID-19 infection that did not require hospitalization were enrolled at National Jewish Health (Denver, CO, USA) and were grouped into those that developed PASC (n = 29) and those that fully recovered (n = 16). Plasma samples from the two groups were analyzed via mass spectrometry-based untargeted metabolomics and compared against plasma metabolic profiles of healthy control individuals (n = 30). Observational demographic and clinical data were retrospectively abstracted from the medical record. Compared to plasma of healthy controls or individuals who recovered from COVID-19, PASC plasma exhibited significantly higher free- and carnitine-conjugated mono-, poly-, and highly unsaturated fatty acids, accompanied by markedly lower levels of mono-, di- and tricarboxylates (pyruvate, lactate, citrate, succinate, and malate), polyamines (spermine) and taurine. Plasma from individuals who fully recovered from COVID-19 exhibited an intermediary metabolic phenotype, with milder disturbances in fatty acid metabolism and higher levels of spermine and taurine. Of note, depletion of tryptophan-a hallmark of disease severity in COVID-19-is not normalized in PASC patients, despite normalization of kynurenine levels-a tryptophan metabolite that predicts mortality in hospitalized COVID-19 patients. In conclusion, PASC plasma metabolites are indicative of altered fatty acid metabolism and dysfunctional mitochondria-dependent lipid catabolism. These metabolic profiles obtained at rest are consistent with previously reported mitochondrial dysfunction during exercise, and may pave the way for therapeutic intervention focused on restoring mitochondrial fat-burning capacity.
Collapse
Affiliation(s)
- Vamsi P. Guntur
- Division of Pulmonary and Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, CO 80206, USA
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Travis Nemkov
- Department of Biochemical and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Esther de Boer
- Division of Pulmonary and Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, CO 80206, USA
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Michael P. Mohning
- Division of Pulmonary and Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, CO 80206, USA
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - David Baraghoshi
- Department of Biostatistics, National Jewish Health, Denver, CO 80206, USA
| | - Francesca I. Cendali
- Department of Biochemical and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Inigo San-Millán
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Human Physiology and Nutrition, University of Colorado, Colorado Springs, CO 80918, USA
| | - Irina Petrache
- Division of Pulmonary and Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, CO 80206, USA
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Angelo D’Alessandro
- Department of Biochemical and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
69
|
O’Connor T, Javidi B. COVID-19 screening with digital holographic microscopy using intra-patient probability functions of spatio-temporal bio-optical attributes. BIOMEDICAL OPTICS EXPRESS 2022; 13:5377-5389. [PMID: 36425632 PMCID: PMC9664885 DOI: 10.1364/boe.466005] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/23/2022] [Accepted: 08/28/2022] [Indexed: 06/16/2023]
Abstract
We present an automated method for COVID-19 screening using the intra-patient population distributions of bio-optical attributes extracted from digital holographic microscopy reconstructed red blood cells. Whereas previous approaches have aimed to identify infection by classifying individual cells, here, we propose an approach to incorporate the attribute distribution information from the population of a given human subjects' cells into our classification scheme and directly classify subjects at the patient level. To capture the intra-patient distribution information in a generalized way, we propose an approach based on the Bag-of-Features (BoF) methodology to transform histograms of bio-optical attribute distributions into feature vectors for classification via a linear support vector machine. We compare our approach with simpler classifiers directly using summary statistics such as mean, standard deviation, skewness, and kurtosis of the distributions. We also compare to a k-nearest neighbor classifier using the Kolmogorov-Smirnov distance as a distance metric between the attribute distributions of each subject. We lastly compare our approach to previously published methods for classification of individual red blood cells. In each case, the methodology proposed in this paper provides the highest patient classification performance, correctly classifying 22 out of 24 individuals and achieving 91.67% classification accuracy with 90.00% sensitivity and 92.86% specificity. The incorporation of distribution information for classification additionally led to the identification of a singular temporal-based bio-optical attribute capable of highly accurate patient classification. To the best of our knowledge, this is the first report of a machine learning approach using the intra-patient probability distribution information of bio-optical attributes obtained from digital holographic microscopy for disease screening.
Collapse
Affiliation(s)
- Timothy O’Connor
- Biomedical Engineering Department, University of Connecticut, Storrs, CT 06269, USA
| | - Bahram Javidi
- Electrical and Computer Engineering Department, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
70
|
Weber-Fishkin S, Seidner HS, Gunter G, Frame MD. Erythrocyte aggregation in sudden flow arrest is linked to hyperthermia, hypoxemia, and band 3 availability. J Thromb Haemost 2022; 20:2284-2292. [PMID: 35841276 DOI: 10.1111/jth.15821] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 07/11/2022] [Accepted: 07/13/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Erythrocyte aggregation is a phenomenon that is commonly found in several pathological disease states: stroke, myocardial infarction, thermal burn injury, and COVID-19. Erythrocyte aggregation is characterized by rouleaux, closely packed stacks of cells, forming three-dimensional structures. Healthy blood flow monodisperses the red blood cells (RBCs) throughout the vasculature; however, in select pathological conditions, involving hyperthermia and hypoxemia, rouleaux formation remains and results in occlusion of microvessels with decreased perfusion. OBJECTIVES Our objective is to address the kinetics of rouleaux formation with sudden cessation of flow in variable temperature and oxygen conditions. METHODS RBCs used in this in vitro system were obtained from healthy human donors. Using a vertical stop-flow system aligned with a microscope, images were acquired and analyzed for increased variation in grayscale to indicate increased aggregation. The onset of aggregation after sudden cessation of flow was determined at proscribed temperatures (37-49°C) and oxygen (0%, 10%), and in the presence and absence of 4, 4'-Diisothiocyano-2,2'-stilbenedisulfonic acid (DIDS). Both autologous and homologous plasma were tested. RESULTS RBCs in autologous plasma aggregate faster and with a higher magnitude with both hyperthermia and hypoxemia. Preventing deoxyhemoglobin from binding to band 3 with DIDS (dissociates the cytoskeleton from the membrane) fully blocks aggregation. Further, RBC aggregation magnitude is greater in autologous plasma. CONCLUSIONS We show that the C-terminal domain of band 3 plays a pivotal role in RBC aggregation. Further, aggregation is enhanced by hyperthermia and hypoxemia.
Collapse
Affiliation(s)
- Samantha Weber-Fishkin
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| | - Harrison S Seidner
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| | - Geoffrey Gunter
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| | - Mary D Frame
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
71
|
Pontolillo M, Ucciferri C, Borrelli P, Di Nicola M, Vecchiet J, Falasca K. Molnupiravir as an Early Treatment for COVID-19: A Real Life Study. Pathogens 2022; 11:1121. [PMID: 36297178 PMCID: PMC9610792 DOI: 10.3390/pathogens11101121] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/22/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVES Below we report our experience in the use of molnupiravir, the first antiviral drug against SARS-CoV-2 available to us, in the treatment of patients with COVID-19. MATERIALS AND METHODS We enrolled patients diagnosed with COVID-19 and comorbidities who were candidates for antiviral drug therapy. All patients received molnupiravir (800 mg twice daily). Blood chemistry checks were carried out at T0 and after 7/10 days after starting therapy (T1). RESULTS There were enrolled within the cohort 100 patients. There was 100.0% compliance with the antiviral treatment. No patient required hospitalization due to worsening of respiratory function or the appearance of serious side effects. The median downtime of viral load was ten days (IQR 8.0-13.0), regardless of the type of vaccination received. The patients who had a shorter distance from vaccination more frequently presented vomiting/diarrhea. During baseline and T1 we found significant differences in the median serum concentrations of the main parameters, in particular of platelets, RDW CV, neutrophils and lymphocytes, the eGFR, liver enzymes, as well as of the main inflammatory markers, CRP and Ferritin. CONCLUSION Participants treated with molnupiravir, albeit in risk categories, demonstrated early clinical improvement, no need for hospitalization, and a low rate of adverse events.
Collapse
Affiliation(s)
- Michela Pontolillo
- Clinic of Infectious Diseases, Department of Medicine and Science of Aging, University “G. d’Annunzio” Chieti, 66100 Pescara, Italy
| | - Claudio Ucciferri
- Clinic of Infectious Diseases, Department of Medicine and Science of Aging, University “G. d’Annunzio” Chieti, 66100 Pescara, Italy
| | - Paola Borrelli
- Laboratory of Biostatistics, Department of Medical, Oral and Biotechnological Sciences, University “G. d’Annunzio” Chieti, 66100 Pescara, Italy
| | - Marta Di Nicola
- Laboratory of Biostatistics, Department of Medical, Oral and Biotechnological Sciences, University “G. d’Annunzio” Chieti, 66100 Pescara, Italy
| | - Jacopo Vecchiet
- Clinic of Infectious Diseases, Department of Medicine and Science of Aging, University “G. d’Annunzio” Chieti, 66100 Pescara, Italy
| | - Katia Falasca
- Clinic of Infectious Diseases, Department of Medicine and Science of Aging, University “G. d’Annunzio” Chieti, 66100 Pescara, Italy
| |
Collapse
|
72
|
Haunhorst S, Bloch W, Wagner H, Ellert C, Krüger K, Vilser DC, Finke K, Reuken P, Pletz MW, Stallmach A, Puta C. Long COVID: a narrative review of the clinical aftermaths of COVID-19 with a focus on the putative pathophysiology and aspects of physical activity. OXFORD OPEN IMMUNOLOGY 2022; 3:iqac006. [PMID: 36846561 PMCID: PMC9494493 DOI: 10.1093/oxfimm/iqac006] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/30/2022] [Accepted: 09/13/2022] [Indexed: 11/14/2022] Open
Abstract
The pandemic coronavirus disease 2019 (COVID-19) can cause multi-systemic symptoms that can persist beyond the acute symptomatic phase. The post-acute sequelae of COVID-19 (PASC), also referred to as long COVID, describe the persistence of symptoms and/or long-term complications beyond 4 weeks from the onset of the acute symptoms and are estimated to affect at least 20% of the individuals infected with SARS-CoV-2 regardless of their acute disease severity. The multi-faceted clinical picture of long COVID encompasses a plethora of undulating clinical manifestations impacting various body systems such as fatigue, headache, attention disorder, hair loss and exercise intolerance. The physiological response to exercise testing is characterized by a reduced aerobic capacity, cardiocirculatory limitations, dysfunctional breathing patterns and an impaired ability to extract and use oxygen. Still, to this day, the causative pathophysiological mechanisms of long COVID remain to be elucidated, with long-term organ damage, immune system dysregulation and endotheliopathy being among the hypotheses discussed. Likewise, there is still a paucity of treatment options and evidence-based strategies for the management of the symptoms. In sum, this review explores different aspects of long COVID and maps the literature on what is known about its clinical manifestations, potential pathophysiological mechanisms, and treatment options.
Collapse
Affiliation(s)
- Simon Haunhorst
- Department of Sports Medicine and Health Promotion, Friedrich-Schiller-University Jena, Jena 07749, Germany
- Department of Movement Science, University of Münster, Münster 48149, Germany
| | - Wilhelm Bloch
- Department for Molecular and Cellular Sports Medicine, Institute for Cardiovascular Research and Sports Medicine, German Sport University Cologne, Cologne 50933, Germany
| | - Heiko Wagner
- Department of Movement Science, University of Münster, Münster 48149, Germany
| | - Claudia Ellert
- Department for Vascular Surgery, Lahn-Dill Clinics Wetzlar, Wetzlar 35578, Germany
| | - Karsten Krüger
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University Giessen, Giessen 35394, Germany
| | - Daniel C Vilser
- Hospital for Pediatrics and Adolescent Medicine, Jena University Hospital, Jena 07747, Germany
| | - Kathrin Finke
- Department of Neurology, Jena University Hospital, Jena 07747, Germany
| | - Philipp Reuken
- Clinic for Internal Medicine IV (Gastroenterology, Hepatology and Infectious Diseases), Jena University Hospital, Jena 07747, Germany
| | - Mathias W Pletz
- Institute for Infectious Diseases and Infection Control, Jena University Hospital, Jena 07747, Germany
| | - Andreas Stallmach
- Clinic for Internal Medicine IV (Gastroenterology, Hepatology and Infectious Diseases), Jena University Hospital, Jena 07747, Germany
| | - Christian Puta
- Department of Sports Medicine and Health Promotion, Friedrich-Schiller-University Jena, Jena 07749, Germany
- Center for Interdisciplinary Prevention of Diseases related to Professional Activities, Jena 07749, Germany
| |
Collapse
|
73
|
Himbert S, Rheinstädter MC. Structural and mechanical properties of the red blood cell's cytoplasmic membrane seen through the lens of biophysics. Front Physiol 2022; 13:953257. [PMID: 36171967 PMCID: PMC9510598 DOI: 10.3389/fphys.2022.953257] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/15/2022] [Indexed: 11/27/2022] Open
Abstract
Red blood cells (RBCs) are the most abundant cell type in the human body and critical suppliers of oxygen. The cells are characterized by a simple structure with no internal organelles. Their two-layered outer shell is composed of a cytoplasmic membrane (RBC cm ) tethered to a spectrin cytoskeleton allowing the cell to be both flexible yet resistant against shear stress. These mechanical properties are intrinsically linked to the molecular composition and organization of their shell. The cytoplasmic membrane is expected to dominate the elastic behavior on small, nanometer length scales, which are most relevant for cellular processes that take place between the fibrils of the cytoskeleton. Several pathologies have been linked to structural and compositional changes within the RBC cm and the cell's mechanical properties. We review current findings in terms of RBC lipidomics, lipid organization and elastic properties with a focus on biophysical techniques, such as X-ray and neutron scattering, and Molecular Dynamics simulations, and their biological relevance. In our current understanding, the RBC cm 's structure is patchy, with nanometer sized liquid ordered and disordered lipid, and peptide domains. At the same time, it is surprisingly soft, with bending rigidities κ of 2-4 kBT. This is in strong contrast to the current belief that a high concentration of cholesterol results in stiff membranes. This extreme softness is likely the result of an interaction between polyunsaturated lipids and cholesterol, which may also occur in other biological membranes. There is strong evidence in the literature that there is no length scale dependence of κ of whole RBCs.
Collapse
Affiliation(s)
- Sebastian Himbert
- Department of Physics and Astronomy, McMaster University, Hamilton, ON, Canada
- Origins Institute, McMaster University, Hamilton, ON, Canada
| | - Maikel C. Rheinstädter
- Department of Physics and Astronomy, McMaster University, Hamilton, ON, Canada
- Origins Institute, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
74
|
D’Alessandro A. Editorial: Rising stars in red blood cell physiology: 2022. Front Physiol 2022; 13:1020144. [PMID: 36160846 PMCID: PMC9501848 DOI: 10.3389/fphys.2022.1020144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 08/17/2022] [Indexed: 11/17/2022] Open
|
75
|
Al-kuraishy HM, Al-Gareeb AI, Kaushik A, Kujawska M, Batiha GES. Hemolytic anemia in COVID-19. Ann Hematol 2022; 101:1887-1895. [PMID: 35802164 PMCID: PMC9263052 DOI: 10.1007/s00277-022-04907-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/25/2022] [Indexed: 12/15/2022]
Abstract
COVID-19 is a global pandemic triggered by the severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2). The SARS-CoV-2 entry point involves the interaction with angiotensin-converting enzyme 2 (ACE2) receptor, CD147, and erythrocyte Band3 protein. Hemolytic anemia has been linked to COVID-19 through induction of autoimmune hemolytic anemia (AIHA) caused by the formation of autoantibodies (auto-Abs) or directly through CD147 or erythrocyte Band3 protein-mediated erythrocyte injury. Here, we aim to provide a comprehensive view of the potential mechanisms contributing to hemolytic anemia during the SARS-CoV-2 infection. Taken together, data discussed here highlight that SARS-CoV-2 infection may lead to hemolytic anemia directly through cytopathic injury or indirectly through induction of auto-Abs. Thus, as SARS-CoV-2-induced hemolytic anemia is increasingly associated with COVID-19, early detection and management of this condition may prevent the poor prognostic outcomes in COVID-19 patients. Moreover, since hemolytic exacerbations may occur upon medicines for COVID-19 treatment and anti-SARS-CoV-2 vaccination, continued monitoring for complications is also required. Given that, intelligent nanosystems offer tools for broad-spectrum testing and early diagnosis of the infection, even at point-of-care sites.
Collapse
Affiliation(s)
- Hayder M. Al-kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, M.B.Ch.B, FRCP, Baghdad, Iraq
| | - Ali I. Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, M.B.Ch.B, FRCP, Baghdad, Iraq
| | - Ajeet Kaushik
- NanoBioTech Laboratory, Department of Environmental Engineering, Florida Polytechnic University, Lakeland, FL 33805-8531 USA
| | - Małgorzata Kujawska
- Department of Toxicology, Faculty of Pharmacy, Poznan University of Medical Sciences, Dojazd 30, 60-631 Poznań, Poland
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Al Beheira, Egypt
| |
Collapse
|
76
|
Subramanian SK, Sripad VD, Dharmalingam A, Guhan VN, Kalidoss VK, Gautam N, Shankaralingappa A, Rajendran R, Mohiuddin SG. Effect of 4-Week Heartfulness Meditation on Stress Scores, Sleep Quality, and Oxidative and Inflammatory Biochemical Parameters in COVID-19 Patients after Completion of Standard Treatment - A Randomized Controlled Trial. Int J Yoga 2022; 15:195-204. [PMID: 36949840 PMCID: PMC10026342 DOI: 10.4103/ijoy.ijoy_95_22] [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: 05/27/2022] [Revised: 08/10/2022] [Accepted: 08/16/2022] [Indexed: 01/19/2023] Open
Abstract
Context COVID-19-affected patients showed increased stress, impaired sleep quality, altered complete blood count, and increased inflammatory and oxidative parameters. Yoga is an add-on nonpharmacological treatment that is established to normalize the abovementioned parameters. Heartfulness meditation is a form of Raja yoga. Aims The present study aimed to study the effects of 4 weeks of heartfulness meditation on the abovementioned parameters in COVID-19 patients following treatment completion. Settings and Design The present study was a randomized controlled trial carried out in the Department of Physiology, AIIMS, Mangalagiri, Andhra Pradesh. Subjects and Methods Out of 50 COVID-19 treatment-completed patients recruited for the study, 25 were randomly assigned to the study group who received 4-week app-based heartfulness meditation. Other 25 patients were assigned to the control group who received app-based relaxation for 4 weeks. Perceived stress score, Pittsburgh Sleep Quality Index questionnaire, baseline cardiovascular parameters, complete blood count, serum cortisol, inflammatory parameters, oxidative stress parameters, and antioxidant parameters were assessed before and after 4 weeks of intervention in both the groups. The outcome assessor was blinded in the present study. Statistical Analysis Used The mean difference between the two groups was tested using the Student's t-test or Mann-Whitney U-test based on data distribution. Effect of intervention was analyzed using paired Student's t-test for dependent samples test or Wilcoxon signed-rank test based on data distribution. Results The groups were comparable before intervention for all the variables. After 4 weeks of intervention, we observed a significant decrease in stress, circulating cortisol, inflammatory markers, and oxidative stress biomarker in both the groups. Further, we observed improved sleep quality and antioxidant biomarkers in both the groups. These beneficial alterations following intervention were high in the study group compared to the control group. Conclusions Our results suggest that app-based heartfulness meditation/relaxation can be used as a nonpharmacological adjuvant to hasten the recovery process in patients who have completed the COVID-19 treatment protocol. Beneficial effects in subjects practicing heartfulness meditation were more than that observed in subjects practicing relaxation.
Collapse
Affiliation(s)
- Senthil Kumar Subramanian
- Department of Physiology, All India Institute of Medical Sciences, Mangalagiri, Andhra Pradesh, India
| | - Vidya Desai Sripad
- Department of Biochemistry, All India Institute of Medical Sciences, Mangalagiri, Andhra Pradesh, India
| | - Amudharaj Dharmalingam
- Department of Physiology, All India Institute of Medical Sciences, Mangalagiri, Andhra Pradesh, India
| | - V. Naga Guhan
- Department of Biochemistry, All India Institute of Medical Sciences, Mangalagiri, Andhra Pradesh, India
| | - Vinoth Kumar Kalidoss
- Department of Community and Family Medicine, All India Institute of Medical Sciences, Mangalagiri, Andhra Pradesh, India
| | - Nichenametla Gautam
- Department of Biochemistry, All India Institute of Medical Sciences, Mangalagiri, Andhra Pradesh, India
- Department of Biochemistry, Trichy SRM Medical College Hospital and Research Centre, Tiruchirappalli, Tamil Nadu, India
| | | | - Rajathi Rajendran
- Department of Physiology, All India Institute of Medical Sciences, Mangalagiri, Andhra Pradesh, India
| | - Syed Ghouse Mohiuddin
- Department of Physiology, All India Institute of Medical Sciences, Mangalagiri, Andhra Pradesh, India
| |
Collapse
|
77
|
Yang M. Redox stress in COVID-19: Implications for hematologic disorders. Best Pract Res Clin Haematol 2022; 35:101373. [PMID: 36494143 PMCID: PMC9374492 DOI: 10.1016/j.beha.2022.101373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/01/2022] [Accepted: 08/07/2022] [Indexed: 01/08/2023]
Abstract
COVID-19 is the respiratory illness caused by the beta coronavirus SARS-CoV-2. COVID-19 is complicated by an increased risk for adverse thrombotic events that promote organ failure and death. While the mechanism of action for SARS-CoV-2 is still being understood, how SARS-CoV-2 infection impacts the redox environment in hematologic conditions is unclear. In this review, the redox mechanisms contributing to SARS-CoV-2 infection, coagulopathy and inflammation are briefly discussed. Specifically, sources of oxidant generation by hematopoietic and non-hematopoietic cells are identified with special emphasis on leukocytes, platelets, red cells, and endothelial cells. Furthermore, reactive cysteines in SARS-CoV-2 are also discussed with respect to oxidative cysteine modification and current therapeutic implications. Lastly, sickle cell disease will be discussed as a hematologic disorder with a pre-existing prothrombotic redox condition that complicates treatment strategies for COVID-19. An understanding of the redox mechanism may identify potential targets for COVID-19-mediated thrombosis in hematologic disorders.
Collapse
Affiliation(s)
- Moua Yang
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02115, United States.
| |
Collapse
|
78
|
Wang T, Cao Y, Zhang H, Wang Z, Man CH, Yang Y, Chen L, Xu S, Yan X, Zheng Q, Wang Y. COVID-19 metabolism: Mechanisms and therapeutic targets. MedComm (Beijing) 2022; 3:e157. [PMID: 35958432 PMCID: PMC9363584 DOI: 10.1002/mco2.157] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/26/2022] [Accepted: 06/29/2022] [Indexed: 01/18/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) dysregulates antiviral signaling, immune response, and cell metabolism in human body. Viral genome and proteins hijack host metabolic network to support viral biogenesis and propagation. However, the regulatory mechanism of SARS-CoV-2-induced metabolic dysfunction has not been elucidated until recently. Multiomic studies of coronavirus disease 2019 (COVID-19) revealed an intensive interaction between host metabolic regulators and viral proteins. SARS-CoV-2 deregulated cellular metabolism in blood, intestine, liver, pancreas, fat, and immune cells. Host metabolism supported almost every stage of viral lifecycle. Strikingly, viral proteins were found to interact with metabolic enzymes in different cellular compartments. Biochemical and genetic assays also identified key regulatory nodes and metabolic dependencies of viral replication. Of note, cholesterol metabolism, lipid metabolism, and glucose metabolism are broadly involved in viral lifecycle. Here, we summarized the current understanding of the hallmarks of COVID-19 metabolism. SARS-CoV-2 infection remodels host cell metabolism, which in turn modulates viral biogenesis and replication. Remodeling of host metabolism creates metabolic vulnerability of SARS-CoV-2 replication, which could be explored to uncover new therapeutic targets. The efficacy of metabolic inhibitors against COVID-19 is under investigation in several clinical trials. Ultimately, the knowledge of SARS-CoV-2-induced metabolic reprogramming would accelerate drug repurposing or screening to combat the COVID-19 pandemic.
Collapse
Affiliation(s)
- Tianshi Wang
- Shanghai Key Laboratory for Tumor Microenvironment and InflammationDepartment of Biochemistry and Molecular Cell BiologyShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ying Cao
- State Key Laboratory of Oncogenes and Related GenesShanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Haiyan Zhang
- Bai Jia Obstetrics and Gynecology HospitalShanghaiChina
| | - Zihao Wang
- Fudan University Shanghai Cancer CenterKey Laboratory of Breast Cancer in ShanghaiShanghai Key Laboratory of Radiation OncologyCancer Instituteand The Shanghai Key Laboratory of Medical EpigeneticsInstitutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
- The International Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyShanghaiChina
| | - Cheuk Him Man
- Division of HematologyDepartment of MedicineUniversity of Hong KongPokfulamHong Kong, China
| | - Yunfan Yang
- Department of Cell BiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanChina
| | - Lingchao Chen
- Department of NeurosurgeryHuashan HospitalShanghai Medical CollegeFudan UniversityNational Center for Neurological DisordersShanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationNeurosurgical Institute of Fudan UniversityShanghai Clinical Medical Center of NeurosurgeryShanghaiChina
| | - Shuangnian Xu
- Department of HematologySouthwest HospitalArmy Medical UniversityChongqingChina
| | - Xiaojing Yan
- Department of HematologyThe First Affiliated Hospital of China Medical UniversityShenyangChina
| | - Quan Zheng
- Center for Single‐Cell OmicsSchool of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yi‐Ping Wang
- Fudan University Shanghai Cancer CenterKey Laboratory of Breast Cancer in ShanghaiShanghai Key Laboratory of Radiation OncologyCancer Instituteand The Shanghai Key Laboratory of Medical EpigeneticsInstitutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
- The International Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyShanghaiChina
| |
Collapse
|
79
|
Kell DB, Pretorius E. The potential role of ischaemia-reperfusion injury in chronic, relapsing diseases such as rheumatoid arthritis, Long COVID, and ME/CFS: evidence, mechanisms, and therapeutic implications. Biochem J 2022; 479:1653-1708. [PMID: 36043493 PMCID: PMC9484810 DOI: 10.1042/bcj20220154] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 02/07/2023]
Abstract
Ischaemia-reperfusion (I-R) injury, initiated via bursts of reactive oxygen species produced during the reoxygenation phase following hypoxia, is well known in a variety of acute circumstances. We argue here that I-R injury also underpins elements of the pathology of a variety of chronic, inflammatory diseases, including rheumatoid arthritis, ME/CFS and, our chief focus and most proximally, Long COVID. Ischaemia may be initiated via fibrin amyloid microclot blockage of capillaries, for instance as exercise is started; reperfusion is a necessary corollary when it finishes. We rehearse the mechanistic evidence for these occurrences here, in terms of their manifestation as oxidative stress, hyperinflammation, mast cell activation, the production of marker metabolites and related activities. Such microclot-based phenomena can explain both the breathlessness/fatigue and the post-exertional malaise that may be observed in these conditions, as well as many other observables. The recognition of these processes implies, mechanistically, that therapeutic benefit is potentially to be had from antioxidants, from anti-inflammatories, from iron chelators, and via suitable, safe fibrinolytics, and/or anti-clotting agents. We review the considerable existing evidence that is consistent with this, and with the biochemical mechanisms involved.
Collapse
Affiliation(s)
- Douglas B. Kell
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, U.K
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Kemitorvet 200, 2800 Kgs Lyngby, Denmark
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland 7602, South Africa
| | - Etheresia Pretorius
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, U.K
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland 7602, South Africa
| |
Collapse
|
80
|
Alkahtani AM, Alraey Y, Zaman GS, Al‐Shehri H, Alghamdi IS, Chandramoorthy HC, Al-Hakami AM, Alamri AM, Alshehri HA. Haematological Traits in Symptomatic and Asymptomatic COVID-19 Positive Patients for Predicting Severity and Hospitalization. J Blood Med 2022; 13:447-459. [PMID: 36062061 PMCID: PMC9432386 DOI: 10.2147/jbm.s365218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 06/29/2022] [Indexed: 11/23/2022] Open
Abstract
Objective The present investigation aims on the clinical attributes and haematological parameters between symptomatic (COVID-19 ICU) and asymptomatic (COVID-19 homes isolation) patients as predisposing sign for COVID-19 related mortality. Materials and Methods A retrospective cohort research was conducted of admitted patients to ICU, who were suffering from severe COVID-19 in Aseer Central Hospital, Abha, Kingdom of Saudi Arabia (KSA) from July 2020 until September 2020. The study included individuals with COVID -19 and ICU admission as symptomatic group and others who are COVID-19 positives with quarantine as asymptomatic group. Epidemiological, clinical and haematological laboratory data were retrospectively collected, analysed with control subjects. Results Of the 38 ICU patients studied, the most common symptoms were fever and respiratory distress (100%), cough (86.8%). Majority were of Saudi origin (78.9%). Eighteen (47.4%) COVID-19 ICU patients showed leukocytosis, 6 (15.8%) had severe thrombocytopenia (with most having thrombocytopenia), 18 (47.4%) were anaemic. A significant correlation was observed between the WBC, RBC, Hb, platelets, neutrophil and lymphocyte count between ICU inmates compared with quarantine (p < 0.001) and RBC, Hb, neutrophil and lymphocyte count with control groups (p < 0.001). Conclusion From the observations it is evident that, the blood tests have potential clinical value in predicting COVID-19 progression. Further, patient characteristics including age, leukocyte count, RBC, platelets and differential leukocyte counts may be significant predictors for monitoring the progression of the critical illness observed in SARS-COV-2 patients. Also, treatment procedures can be re-defined further to reduce COVID-19 mortalities in more critically ill COVID-19 individuals.
Collapse
Affiliation(s)
- Abdullah M Alkahtani
- Department of Microbiology and Clinical Parasitology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Yasser Alraey
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Gaffar Sarwar Zaman
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Hajri Al‐Shehri
- Central Laboratory, Assir Central Hospital, Ministry of Health, Abha, Saudi Arabia
| | | | - Harish C Chandramoorthy
- Department of Microbiology and Clinical Parasitology, College of Medicine, King Khalid University, Abha, Saudi Arabia
- Center for Stem Cell Research, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Ahmed M Al-Hakami
- Department of Microbiology and Clinical Parasitology, College of Medicine, King Khalid University, Abha, Saudi Arabia
- Center for Stem Cell Research, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Ahmad M Alamri
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
- Cancer Research Unit, King Khalid University, Abha, Saudi Arabia
| | - Hassan Ali Alshehri
- Central Laboratory, Assir Central Hospital, Ministry of Health, Abha, Saudi Arabia
| |
Collapse
|
81
|
Upadhyai P, Shenoy PU, Banjan B, Albeshr MF, Mahboob S, Manzoor I, Das R. Exome-Wide Association Study Reveals Host Genetic Variants Likely Associated with the Severity of COVID-19 in Patients of European Ancestry. Life (Basel) 2022; 12:1300. [PMID: 36143338 PMCID: PMC9504138 DOI: 10.3390/life12091300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 12/03/2022] Open
Abstract
Host genetic variability plays a pivotal role in modulating COVID-19 clinical outcomes. Despite the functional relevance of protein-coding regions, rare variants located here are less likely to completely explain the considerable numbers of acutely affected COVID-19 patients worldwide. Using an exome-wide association approach, with individuals of European descent, we sought to identify common coding variants linked with variation in COVID-19 severity. Herein, cohort 1 compared non-hospitalized (controls) and hospitalized (cases) individuals, and in cohort 2, hospitalized subjects requiring respiratory support (cases) were compared to those not requiring it (controls). 229 and 111 variants differed significantly between cases and controls in cohorts 1 and 2, respectively. This included FBXO34, CNTN2, and TMCC2 previously linked with COVID-19 severity using association studies. Overall, we report SNPs in 26 known and 12 novel candidate genes with strong molecular evidence implicating them in the pathophysiology of life-threatening COVID-19 and post-recovery sequelae. Of these few notable known genes include, HLA-DQB1, AHSG, ALOX5AP, MUC5AC, SMPD1, SPG7, SPEG,GAS6, and SERPINA12. These results enhance our understanding of the pathomechanisms underlying the COVID-19 clinical spectrum and may be exploited to prioritize biomarkers for predicting disease severity, as well as to improve treatment strategies in individuals of European ancestry.
Collapse
Affiliation(s)
- Priyanka Upadhyai
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - Pooja U. Shenoy
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India
| | - Bhavya Banjan
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Mohammed F. Albeshr
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Shahid Mahboob
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Irfan Manzoor
- Department of Biology, The College of Arts and Sciences, Indiana University, Bloomington, IN 47405, USA
| | - Ranajit Das
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India
| |
Collapse
|
82
|
Konozy EHE, Osman MEFM, Ghartey-Kwansah G, Abushama HM. The striking mimics between COVID-19 and malaria: A review. Front Immunol 2022; 13:957913. [PMID: 36081516 PMCID: PMC9445119 DOI: 10.3389/fimmu.2022.957913] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/04/2022] [Indexed: 11/25/2022] Open
Abstract
Objectives COVID-19 is a transmissible illness triggered by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Since its onset in late 2019 in Wuhan city of China, it continues to spread universally, leading to an ongoing pandemic that shattered all efforts to restrain it. On the other hand, in Africa, the COVID-19 infection may be influenced by malaria coinfection. Hence, in this review article, we aimed to give a comprehensive account of the similarities between COVID-19 and malaria in terms of symptoms, clinical, immunological, and molecular perspectives. Methodology In this article, we reviewed over 50 research papers to highlight the multilayered similarities between COVID-19 and malaria infections that might influence the ontology of COVID-19. Results Despite the poor health and fragile medical system of many sub-Saharan African countries, they persisted with a statistically significantly low number of COVID-19 cases. This was attributed to many factors such as the young population age, the warm weather, the lack of proper diagnosis, previous infection with malaria, the use of antimalarial drugs, etc. Additionally, population genetics appears to play a significant role in shaping the COVID-19 dynamics. This is evident as recent genomic screening analyses of the angiotensin-converting enzyme 2 (ACE2) and malaria-associated-variants identified 6 candidate genes that might play a role in malaria and COVID-19 incidence and severity. Moreover, the clinical and pathological resemblances between the two diseases have made considerable confusion in the diagnosis and thereafter curb the disease in Africa. Therefore, possible similarities between the diseases in regards to the clinical, pathological, immunological, and genetical ascription were discussed. Conclusion Understanding the dynamics of COVID-19 infection in Sub-Saharan Africa and how it is shaped by another endemic disease like malaria can provide insights into how to tailor a successful diagnostic, intervention, and control plans that lower both disease morbidity and mortality.
Collapse
Affiliation(s)
| | | | - George Ghartey-Kwansah
- Department of Biomedical Sciences, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | | |
Collapse
|
83
|
Ousaka D, Nishibori M. Is hemolysis a novel therapeutic target in COVID-19? Front Immunol 2022; 13:956671. [PMID: 36059481 PMCID: PMC9438449 DOI: 10.3389/fimmu.2022.956671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/26/2022] [Indexed: 12/15/2022] Open
Affiliation(s)
- Daiki Ousaka
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Masahiro Nishibori
- Translational Research and Drug Development, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
- *Correspondence: Masahiro Nishibori,
| |
Collapse
|
84
|
Valle A, Rodriguez J, Camiña F, Rodriguez-Segade M, Ortola JB, Rodriguez-Segade S. The oxyhaemoglobin dissociation curve is generally left-shifted in COVID-19 patients at admission to hospital, and this is associated with lower mortality. Br J Haematol 2022; 199:332-338. [PMID: 35971642 PMCID: PMC9538192 DOI: 10.1111/bjh.18431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/12/2022] [Accepted: 08/14/2022] [Indexed: 01/08/2023]
Abstract
Lung damage caused by SARS‐Cov‐2 virus results in marked arterial hypoxia, accompanied in many cases by hypocapnia. The literature is inconclusive as to whether these conditions induce alteration of the affinity of haemoglobin for oxygen. We studied the oxyhaemoglobin dissociation curves (ODCs) of 517 patients hospitalized with coronavirus disease 2019 (COVID‐19) for whom arterial blood gas analysis (BGA) was performed upon hospitalization (i.e., before treatment). With respect to a conventional normal p50 (pO2 at 50% saturation of haemoglobin) of 27 mmHg, 76% had a lower standardized p50 (p50s) and 85% a lower in vivo p50 (p50i). In a 33‐patient subgroup with follow‐up BGAs after 3, 6, 9, 12, 15 and 18 days' treatment, p50s and p50i exhibited statistically significant differences between baseline values and values recorded at all these time points. The 30‐day Kaplan–Meier survival curves of COVID‐19 patients stratified by p50i level show a higher probability of survival among patients who at admission had p50 values below 27 mmHg (p = 0.012). Whether the observed alteration of the affinity of haemoglobin for oxygen in COVID‐19 patients is a direct or indirect effect of the virus on haemoglobin is unknown.
Collapse
Affiliation(s)
- Andrea Valle
- Clinical Biochemistry Laboratory of University Hospital Complex, Santiago de Compostela. Travesía de la Choupana s/n, Santiago de Compostela, Spain
| | - Javier Rodriguez
- Clinical Biochemistry Laboratory of University Hospital Complex, Santiago de Compostela. Travesía de la Choupana s/n, Santiago de Compostela, Spain.,Department of Biochemistry and Molecular Biology, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Félix Camiña
- Department of Biochemistry and Molecular Biology, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Miguel Rodriguez-Segade
- Structural Mechanics Group. ETS Caminos Canales y Puertos. Universidade da Coruña. Campus de Elviña. 15071-, A Coruña, Spain
| | - Juan B Ortola
- Clinical Biochemistry Laboratory of University Hospital Complex, Santiago de Compostela. Travesía de la Choupana s/n, Santiago de Compostela, Spain
| | - Santiago Rodriguez-Segade
- Clinical Biochemistry Laboratory of University Hospital Complex, Santiago de Compostela. Travesía de la Choupana s/n, Santiago de Compostela, Spain.,Department of Biochemistry and Molecular Biology, University of Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
85
|
Allahyani MA, Aljuaid AA, Almehmadi MM, Alghamdi AA, Halawani IF, Aldairi AF, Alharbi AM, Albshri MH, Mutwalli AA, Alhazmi AS. Detection of erythroid progenitors and erythrocytopathies in patients with severe COVID-19 disease. Saudi Med J 2022; 43:899-906. [PMID: 35964959 PMCID: PMC9749667 DOI: 10.15537/smj.2022.43.8.20220311] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/15/2022] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVES To assess the effect of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection on erythropoiesis and red blood cells (RBC) surface markers by evaluating erythroid progenitor cells (CD [cluster of differentiation]71+/CD235a+) and RBC surface markers (CD235a and CD36), together with various hematological parameters. METHODS This case-control study includes 47 participants recruited in the study: 30 patients with coronavirus disease 2019 (COVID-19) and 17 healthy individuals. The COVID-19 patients were recruited from the intensive care unit (ICU) of various hospitals in Makkah, Saudi Arabia. Blood samples were collected during July and September 2021. Red blood cells indices were measured using a CBC analyzer. The expression of CD235a, CD71, and CD36 was obtained using flow cytometry technique. The unpaired t-test was conducted to evaluate the differences in these markers in COVID-19 patients and healthy individuals. RESULTS The data showed that more than half of the COVID-19 patients were anemic (64%). Expansion of erythroid progenitors (CD71+/CD235a+) was detected in the COVID-19 patients. Analysis of the expression of RBC surface markers, such as CD235a and CD36, showed that SARS-CoV-2 was associated with significantly higher expression of these markers in COVID-19 patients. CONCLUSION Severe acute respiratory syndrome coronavirus-2 promoted the expansion of erythroid progenitors in the peripheral blood of COVID-19 patients. In addition, the expression of RBC surface markers was higher in COVID-19 patients. The expansion of erythroid progenitors and alteration of RBC surface markers can contribute to erythrocytopathies observed in severe COVID-19 patients and can therefore be used as prognostic factors.
Collapse
Affiliation(s)
- Mamdouh A. Allahyani
- From the Department of Clinical Laboratory Sciences (Allahyani, Aljuaid, Almehmadi, Alghamdi, Halawani, Alharbi, Alhazmi), College of Applied Medical Sciences, Taif University, Taif; from Department of Laboratory Medicine (Aldairi), Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah; and from the Department of Molecular Biology (Albshri, Mutwalli), the Regional Laboratory, Ministry of Health, Makkah, Saudi Arabia.
- Address correspondence and reprint request to: Dr. Mamdouh Allahyani, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Kingdom of Saudi Arabia. E-mail: ORIC ID: https://orcid.org/0000-0001-7929-7748
| | - Abdulelah A. Aljuaid
- From the Department of Clinical Laboratory Sciences (Allahyani, Aljuaid, Almehmadi, Alghamdi, Halawani, Alharbi, Alhazmi), College of Applied Medical Sciences, Taif University, Taif; from Department of Laboratory Medicine (Aldairi), Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah; and from the Department of Molecular Biology (Albshri, Mutwalli), the Regional Laboratory, Ministry of Health, Makkah, Saudi Arabia.
| | - Mazen M. Almehmadi
- From the Department of Clinical Laboratory Sciences (Allahyani, Aljuaid, Almehmadi, Alghamdi, Halawani, Alharbi, Alhazmi), College of Applied Medical Sciences, Taif University, Taif; from Department of Laboratory Medicine (Aldairi), Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah; and from the Department of Molecular Biology (Albshri, Mutwalli), the Regional Laboratory, Ministry of Health, Makkah, Saudi Arabia.
| | - Ahmad A. Alghamdi
- From the Department of Clinical Laboratory Sciences (Allahyani, Aljuaid, Almehmadi, Alghamdi, Halawani, Alharbi, Alhazmi), College of Applied Medical Sciences, Taif University, Taif; from Department of Laboratory Medicine (Aldairi), Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah; and from the Department of Molecular Biology (Albshri, Mutwalli), the Regional Laboratory, Ministry of Health, Makkah, Saudi Arabia.
| | - Ibrahim F. Halawani
- From the Department of Clinical Laboratory Sciences (Allahyani, Aljuaid, Almehmadi, Alghamdi, Halawani, Alharbi, Alhazmi), College of Applied Medical Sciences, Taif University, Taif; from Department of Laboratory Medicine (Aldairi), Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah; and from the Department of Molecular Biology (Albshri, Mutwalli), the Regional Laboratory, Ministry of Health, Makkah, Saudi Arabia.
| | - Abdullah F. Aldairi
- From the Department of Clinical Laboratory Sciences (Allahyani, Aljuaid, Almehmadi, Alghamdi, Halawani, Alharbi, Alhazmi), College of Applied Medical Sciences, Taif University, Taif; from Department of Laboratory Medicine (Aldairi), Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah; and from the Department of Molecular Biology (Albshri, Mutwalli), the Regional Laboratory, Ministry of Health, Makkah, Saudi Arabia.
| | - Ahmad M. Alharbi
- From the Department of Clinical Laboratory Sciences (Allahyani, Aljuaid, Almehmadi, Alghamdi, Halawani, Alharbi, Alhazmi), College of Applied Medical Sciences, Taif University, Taif; from Department of Laboratory Medicine (Aldairi), Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah; and from the Department of Molecular Biology (Albshri, Mutwalli), the Regional Laboratory, Ministry of Health, Makkah, Saudi Arabia.
| | - Mohammad H. Albshri
- From the Department of Clinical Laboratory Sciences (Allahyani, Aljuaid, Almehmadi, Alghamdi, Halawani, Alharbi, Alhazmi), College of Applied Medical Sciences, Taif University, Taif; from Department of Laboratory Medicine (Aldairi), Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah; and from the Department of Molecular Biology (Albshri, Mutwalli), the Regional Laboratory, Ministry of Health, Makkah, Saudi Arabia.
| | - Abdulqader A. Mutwalli
- From the Department of Clinical Laboratory Sciences (Allahyani, Aljuaid, Almehmadi, Alghamdi, Halawani, Alharbi, Alhazmi), College of Applied Medical Sciences, Taif University, Taif; from Department of Laboratory Medicine (Aldairi), Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah; and from the Department of Molecular Biology (Albshri, Mutwalli), the Regional Laboratory, Ministry of Health, Makkah, Saudi Arabia.
| | - Ayman S. Alhazmi
- From the Department of Clinical Laboratory Sciences (Allahyani, Aljuaid, Almehmadi, Alghamdi, Halawani, Alharbi, Alhazmi), College of Applied Medical Sciences, Taif University, Taif; from Department of Laboratory Medicine (Aldairi), Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah; and from the Department of Molecular Biology (Albshri, Mutwalli), the Regional Laboratory, Ministry of Health, Makkah, Saudi Arabia.
| |
Collapse
|
86
|
Moriconi C, Dzieciatkowska M, Roy M, D'Alessandro A, Roingeard P, Lee JY, Gibb DR, Tredicine M, McGill MA, Qiu A, La Carpia F, Francis RO, Hod EA, Thomas T, Picard M, Akpan IJ, Luckey CJ, Zimring JC, Spitalnik SL, Hudson KE. Retention of functional mitochondria in mature red blood cells from patients with sickle cell disease. Br J Haematol 2022; 198:574-586. [PMID: 35670632 PMCID: PMC9329257 DOI: 10.1111/bjh.18287] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/26/2022] [Accepted: 05/17/2022] [Indexed: 01/07/2023]
Abstract
Sickle cell disease (SCD) is an inherited blood disorder characterized by sickled red blood cells (RBCs), which are more sensitive to haemolysis and can contribute to disease pathophysiology. Although treatment of SCD can include RBC transfusion, patients with SCD have high rates of alloimmunization. We hypothesized that RBCs from patients with SCD have functionally active mitochondria and can elicit a type 1 interferon response. We evaluated blood samples from more than 100 patients with SCD and found elevated frequencies of mitochondria in reticulocytes and mature RBCs, as compared to healthy blood donors. The presence of mitochondria in mature RBCs was confirmed by flow cytometry, electron microscopy, and proteomic analysis. The mitochondria in mature RBCs were metabolically competent, as determined by enzymatic activities and elevated levels of mitochondria-derived metabolites. Metabolically-active mitochondria in RBCs may increase oxidative stress, which could facilitate and/or exacerbate SCD complications. Coculture of mitochondria-positive RBCs with neutrophils induced production of type 1 interferons, which are known to increase RBC alloimmunization rates. These data demonstrate that mitochondria retained in mature RBCs are functional and can elicit immune responses, suggesting that inappropriate retention of mitochondria in RBCs may play an underappreciated role in SCD complications and be an RBC alloimmunization risk factor.
Collapse
Affiliation(s)
- Chiara Moriconi
- Laboratory of Transfusion Biology, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York City, New York, USA
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, Colorado, USA
| | - Micaela Roy
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, Colorado, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, Colorado, USA
| | - Philippe Roingeard
- INSERM U1259 and Electron Microscopy Facility, Université de Tours and CHRU de Tours, Tours, France
| | - June Young Lee
- Department of Pathology and Laboratory Medicine, Division of Transfusion Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - David R Gibb
- Department of Pathology and Laboratory Medicine, Division of Transfusion Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Maria Tredicine
- Department of Translational Medicine and Surgery, Section of General Pathology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marlon A McGill
- Department of Psychiatry, Division of Behavioral Medicine, Columbia University Irving Medical Center, New York City, New York, USA
| | - Annie Qiu
- Laboratory of Transfusion Biology, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York City, New York, USA
| | - Francesca La Carpia
- Laboratory of Transfusion Biology, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York City, New York, USA
| | - Richard O Francis
- Laboratory of Transfusion Biology, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York City, New York, USA
| | - Eldad A Hod
- Laboratory of Transfusion Biology, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York City, New York, USA
| | - Tiffany Thomas
- Laboratory of Transfusion Biology, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York City, New York, USA
| | - Martin Picard
- Department of Psychiatry, Division of Behavioral Medicine, Columbia University Irving Medical Center, New York City, New York, USA
| | - Imo J Akpan
- Division of Hematology/Oncology, Department of Medicine, Columbia University Irving Medical Center, New York City, New York, USA
| | - Chance John Luckey
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - James C Zimring
- University of Virginia School of Medicine, Charlottesville, Virginia, USA.,Carter Immunology Center, University of Virginia, Charlottesville, Virginia, USA
| | - Steven L Spitalnik
- Laboratory of Transfusion Biology, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York City, New York, USA
| | - Krystalyn E Hudson
- Laboratory of Transfusion Biology, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York City, New York, USA
| |
Collapse
|
87
|
Himbert S, D’Alessandro A, Qadri SM, Majcher MJ, Hoare T, Sheffield WP, Nagao M, Nagle JF, Rheinstädter MC. The bending rigidity of the red blood cell cytoplasmic membrane. PLoS One 2022; 17:e0269619. [PMID: 35913930 PMCID: PMC9342732 DOI: 10.1371/journal.pone.0269619] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 05/24/2022] [Indexed: 11/19/2022] Open
Abstract
An important mechanical property of cells is the membrane bending modulus, κ. In the case of red blood cells (RBCs) there is a composite membrane consisting of a cytoplasmic membrane and an underlying spectrin network. Literature values of κ are puzzling, as they are reported over a wide range, from 5 kBT to 230 kBT. To disentangle the contribution of the cytoplasmic membrane from the spectrin network, we investigated the bending of red blood cell cytoplasmic membranes (RBCcm) in the absence of spectrin and adenosine triphosphate (ATP). We used a combination of X-ray diffuse scattering (XDS), neutron spin-echo (NSE) spectrometry and Molecular Dynamics (MD) simulations. Our results indicate values of κ of order 4 kBT to 6 kBT, relatively small compared to literature values for most single component lipid bilayers. We suggest two ways this relative softness might confer biological advantage.
Collapse
Affiliation(s)
- Sebastian Himbert
- Department of Physics and Astronomy, McMaster University, Hamilton, ON, Canada
- Origins Institute, McMaster University, Hamilton, ON, Canada
| | - Angelo D’Alessandro
- Department of Pathology and Cell Biology, Columbia University Vagelos College of Physicians and Surgeons and New York-Presbyterian Hospital, New York, New York, United States of America
- University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Syed M. Qadri
- Faculty of Health Sciences, Ontario Tech University, Oshawa, ON, Canada
| | - Michael J. Majcher
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada
| | - Todd Hoare
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada
| | - William P. Sheffield
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
- Centre for Innovation, Canadian Blood Services, Hamilton, ON, Canada
| | - Michihiro Nagao
- Center for Neutron Research, National Institute of Standards and Technology, Gaithersburg, MD, United States of America
- Department of Materials Science and Engineering, University of Maryland, College Park, MD, United States of America
- Department of Physics and Astronomy, University of Delaware, Newark, DE, United States of America
| | - John F. Nagle
- Department of Physics, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Maikel C. Rheinstädter
- Department of Physics and Astronomy, McMaster University, Hamilton, ON, Canada
- Origins Institute, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
88
|
Wang Z, Li S, Huang B. Alveolar macrophages: Achilles' heel of SARS-CoV-2 infection. Signal Transduct Target Ther 2022; 7:242. [PMID: 35853858 PMCID: PMC9295089 DOI: 10.1038/s41392-022-01106-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/11/2022] [Accepted: 07/04/2022] [Indexed: 11/23/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has caused more than 6.3 million deaths to date. Despite great efforts to curb the spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), vaccines and neutralizing antibodies are in the gloom due to persistent viral mutations and antiviral compounds face challenges of specificity and safety. In addition, vaccines are unable to treat already-infected individuals, and antiviral drugs cannot be used prophylactically. Therefore, exploration of unconventional strategies to curb the current pandemic is highly urgent. Alveolar macrophages (AMs) residing on the surface of alveoli are the first immune cells that dispose of alveoli-invading viruses. Our findings demonstrate that M1 AMs have an acidic endosomal pH, thus favoring SARS-CoV-2 to leave endosomes and release into the cytosol where the virus initiates replication; in contrast, M2 AMs have an increased endosomal pH, which dampens the viral escape and facilitates delivery of the virus for lysosomal degradation. In this review, we propose that AMs are the Achilles’ heel of SARS-CoV-2 infection and that modulation of the endosomal pH of AMs has the potential to eliminate invaded SARS-CoV-2; the same strategy might also be suitable for other lethal respiratory viruses.
Collapse
Affiliation(s)
- Zhenfeng Wang
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, 100005, Beijing, China
| | - Shunshun Li
- Department of Immunology, Basic Medicine College, China Medical University, 110122, Shenyang, Liaoning, China
| | - Bo Huang
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, 100005, Beijing, China. .,Department of Biochemistry & Molecular Biology, Tongji Medical College, Huazhong University of Science & Technology, 430030, Wuhan, China.
| |
Collapse
|
89
|
Nemkov T, Skinner S, Diaw M, Diop S, Samb A, Connes P, D’Alessandro A. Plasma Levels of Acyl-Carnitines and Carboxylic Acids Correlate With Cardiovascular and Kidney Function in Subjects With Sickle Cell Trait. Front Physiol 2022; 13:916197. [PMID: 35910560 PMCID: PMC9326174 DOI: 10.3389/fphys.2022.916197] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Subjects with sickle cell trait (SCT) carry one copy of mutated β-globin gene at position E6V at the origin of the production of sickle hemoglobin (HbS). Indeed, individuals with SCT have both normal hemoglobin and HbS, in contrast to patients with sickle cell disease who inherited of two copies of the mutated gene. Although SCT is generally benign/asymptomatic, carriers may develop certain adverse outcomes such as renal complications, venous thromboembolism, exercise-induced rhabdomyolysis … However, little is known about whether similar metabolic pathways are affected in individuals with SCT and whether these metabolic derangements, if present, correlate to clinically relevant parameters. In this study, we performed metabolomics analysis of plasma from individuals with sickle cell trait (n = 34) compared to healthy controls (n = 30). Results indicated a significant increase in basal circulating levels of hemolysis markers, mono- (pyruvate, lactate), di- and tri-carboxylates (including all Krebs cycle intermediates), suggestive of systems-wide mitochondrial dysfunction in individuals with SCT. Elevated levels of kynurenines and indoles were observed in SCT samples, along with increases in the levels of oxidative stress markers (advanced glycation and protein-oxidation end-products, malondialdehyde, oxylipins, eicosanoids). Increases in circulating levels of acyl-carnitines and fatty acids were observed, consistent with increased membrane lipid damage in individuals with sickle cell trait. Finally, correlation analyses to clinical co-variates showed that alterations in the aforementioned pathways strongly correlated with clinical measurements of blood viscosity, renal (glomerular filtration rate, microalbuminuria, uremia) and cardiovascular function (carotid-femoral pulse wave velocity, blood pressure).
Collapse
Affiliation(s)
- Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, United States
| | - Sarah Skinner
- Inter-university Laboratory of Biology of Motor Function EA7424, Vascular Biology and the Red Blood Cell Team, Claude Bernard University Lyon 1, Lyon, France
| | - Mor Diaw
- Laboratory of Physiology and Functional Exploration, FMPO, UCAD, Dakar, Senegal
- IRL3189 Environnement, Santé, Sociétés CNRS/UCAD Dakar/ UGB Saint-Louis/ USTT Bamako/ CNRST Ouagadougou, Dakar, Senegal
| | - Saliou Diop
- Laboratory of Hemato-immunology, FMPO, UCAD, Dakar, Senegal
| | - Abdoulaye Samb
- Laboratory of Physiology and Functional Exploration, FMPO, UCAD, Dakar, Senegal
- IRL3189 Environnement, Santé, Sociétés CNRS/UCAD Dakar/ UGB Saint-Louis/ USTT Bamako/ CNRST Ouagadougou, Dakar, Senegal
| | - Philippe Connes
- Inter-university Laboratory of Biology of Motor Function EA7424, Vascular Biology and the Red Blood Cell Team, Claude Bernard University Lyon 1, Lyon, France
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, United States
| |
Collapse
|
90
|
Martínez-Gómez LE, Ibarra-González I, Fernández-Lainez C, Tusie T, Moreno-Macías H, Martinez-Armenta C, Jimenez-Gutierrez GE, Vázquez-Cárdenas P, Vidal-Vázquez P, Ramírez-Hinojosa JP, Rodríguez-Zulueta AP, Vargas-Alarcón G, Rojas-Velasco G, Sánchez-Muñoz F, Posadas-Sanchez R, Martínez-Ruiz FDJ, Zayago-Angeles DM, Moreno ML, Barajas-Galicia E, Lopez-Cisneros G, Gonzalez-Fernández NC, Ortega-Peña S, Herrera-López B, Olea-Torres J, Juárez-Arias M, Rosas-Vásquez M, Cabrera-Nieto SA, Magaña JJ, Camacho-Rea MDC, Suarez-Ahedo C, Coronado-Zarco I, Valdespino-Vázquez MY, Martínez-Nava GA, Pineda C, Vela-Amieva M, López-Reyes A, Mex-Gen-COVID Initiative Group. Metabolic Reprogramming in SARS-CoV-2 Infection Impacts the Outcome of COVID-19 Patients. Front Immunol 2022; 13:936106. [PMID: 36341434 PMCID: PMC9634751 DOI: 10.3389/fimmu.2022.936106] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/10/2022] [Indexed: 11/25/2023] Open
Abstract
Severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) infection triggers inflammatory clinical stages that affect the outcome of patients with coronavirus disease 2019 (COVID-19). Disease severity may be associated with a metabolic imbalance related to amino acids, lipids, and energy-generating pathways. The aim of this study was to characterize the profile of amino acids and acylcarnitines in COVID-19 patients. A multicenter, cross-sectional study was carried out. A total of 453 individuals were classified by disease severity. Levels of 11 amino acids, 31 acylcarnitines, and succinylacetone in serum samples were analyzed by electrospray ionization-triple quadrupole tandem mass spectrometry. Different clusters were observed in partial least squares discriminant analysis, with phenylalanine, alanine, citrulline, proline, and succinylacetone providing the major contribution to the variability in each cluster (variable importance in the projection >1.5). In logistic models adjusted by age, sex, type 2 diabetes mellitus, hypertension, and nutritional status, phenylalanine was associated with critical outcomes (odds ratio=5.3 (95% CI 3.16-9.2) in the severe vs. critical model, with an area under the curve of 0.84 (95% CI 0.77-0.90). In conclusion the metabolic imbalance in COVID-19 patients might affect disease progression. This work shows an association of phenylalanine with critical outcomes in COVID-19 patients, highlighting phenylalanine as a potential metabolic biomarker of disease severity.
Collapse
Affiliation(s)
- Laura E. Martínez-Gómez
- Laboratorio de Gerociencias, Laboratorio Facilitador, Laboratorio de Medicina Genómica, Dirección General, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico
| | - Isabel Ibarra-González
- Unidad de Genética de la Nutrición, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Cynthia Fernández-Lainez
- Laboratorio de Errores Innatos del Metabolismo y Tamiz, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México, Mexico
| | - Teresa Tusie
- Unidad de Biología Molecular y Medicina Genómica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Instituto de Investigaciones Biomédicas UNAM, Ciudad de México, Mexico
| | - Hortensia Moreno-Macías
- Unidad de Biología Molecular y Medicina Genómica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Instituto de Investigaciones Biomédicas UNAM, Ciudad de México, Mexico
- Departamento de Economía. División de Ciencias Sociales y Humanidades, Universidad Autónoma Metropolitana, Iztapalapa, Ciudad de México, Mexico
| | - Carlos Martinez-Armenta
- Posgrado en Biología Experimental, Dirección de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana Iztapalapa, Ciudad de México, Mexico
| | - Guadalupe Elizabeth Jimenez-Gutierrez
- Laboratorio de Gerociencias, Laboratorio Facilitador, Laboratorio de Medicina Genómica, Dirección General, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico
| | - Paola Vázquez-Cárdenas
- Centro de Innovación Médica Aplicada, Subdirección de Epidemiología e Infectología, Hospital General Dr. Manuel Gea González, Secretaría de Salud, Ciudad de México, Mexico
| | - Patricia Vidal-Vázquez
- Centro de Innovación Médica Aplicada, Subdirección de Epidemiología e Infectología, Hospital General Dr. Manuel Gea González, Secretaría de Salud, Ciudad de México, Mexico
| | - Juan P. Ramírez-Hinojosa
- Centro de Innovación Médica Aplicada, Subdirección de Epidemiología e Infectología, Hospital General Dr. Manuel Gea González, Secretaría de Salud, Ciudad de México, Mexico
| | - Ana P. Rodríguez-Zulueta
- Centro de Innovación Médica Aplicada, Subdirección de Epidemiología e Infectología, Hospital General Dr. Manuel Gea González, Secretaría de Salud, Ciudad de México, Mexico
| | - Gilberto Vargas-Alarcón
- Departamentos de Biología Molecular, Inmunología, Endocrinologia y Unidad de Cuidados Intensivos, Instituto Nacional de Cardiología Ignacio Chavez, Secretaría de Salud, Ciudad de México, Mexico
| | - Gustavo Rojas-Velasco
- Departamentos de Biología Molecular, Inmunología, Endocrinologia y Unidad de Cuidados Intensivos, Instituto Nacional de Cardiología Ignacio Chavez, Secretaría de Salud, Ciudad de México, Mexico
| | - Fausto Sánchez-Muñoz
- Departamentos de Biología Molecular, Inmunología, Endocrinologia y Unidad de Cuidados Intensivos, Instituto Nacional de Cardiología Ignacio Chavez, Secretaría de Salud, Ciudad de México, Mexico
| | - Rosalinda Posadas-Sanchez
- Departamentos de Biología Molecular, Inmunología, Endocrinologia y Unidad de Cuidados Intensivos, Instituto Nacional de Cardiología Ignacio Chavez, Secretaría de Salud, Ciudad de México, Mexico
| | - Felipe de J. Martínez-Ruiz
- Nuevo Hospital General Delegación Regional Sur de la Ciudad de México, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado (ISSSTE), Ciudad de México, Mexico
| | - Dulce M. Zayago-Angeles
- Nuevo Hospital General Delegación Regional Sur de la Ciudad de México, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado (ISSSTE), Ciudad de México, Mexico
| | - Mariana L. Moreno
- Nuevo Hospital General Delegación Regional Sur de la Ciudad de México, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado (ISSSTE), Ciudad de México, Mexico
| | - Edith Barajas-Galicia
- Hospital Central Norte Petróleos Mexicanos (PEMEX), Estado de México, Mexico City, Mexico
| | - Gerardo Lopez-Cisneros
- Hospital Central Norte Petróleos Mexicanos (PEMEX), Estado de México, Mexico City, Mexico
| | | | - Silvestre Ortega-Peña
- Laboratorio de Gerociencias, Laboratorio Facilitador, Laboratorio de Medicina Genómica, Dirección General, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico
| | - Brígida Herrera-López
- Laboratorio de Gerociencias, Laboratorio Facilitador, Laboratorio de Medicina Genómica, Dirección General, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico
| | - Jessel Olea-Torres
- Laboratorio de Gerociencias, Laboratorio Facilitador, Laboratorio de Medicina Genómica, Dirección General, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico
| | - Manuel Juárez-Arias
- Unidad de Investigación y Desarrollo en Alimentos, Tecnológico Nacional de México/Instituto Tecnológico (IT) Veracruz, Veracruz, Mexico
| | - Maritza Rosas-Vásquez
- Unidad de Investigación y Desarrollo en Alimentos, Tecnológico Nacional de México/Instituto Tecnológico (IT) Veracruz, Veracruz, Mexico
| | - Sara Aileen Cabrera-Nieto
- Posgrado en Ciencias Médicas, Facultad de Ciencias de la Salud, Universidad Anáhuac, Ciudad de México, Mexico
| | - Jonathan J. Magaña
- Laboratorio de Gerociencias, Laboratorio Facilitador, Laboratorio de Medicina Genómica, Dirección General, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico
| | - María del Carmen Camacho-Rea
- Departamento de Nutrición Animal, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Secretaría de Salud, Ciudad de México, Mexico
| | - Carlos Suarez-Ahedo
- Laboratorio de Gerociencias, Laboratorio Facilitador, Laboratorio de Medicina Genómica, Dirección General, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico
| | - Irma Coronado-Zarco
- Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México, Mexico
| | | | - Gabriela Angélica Martínez-Nava
- Laboratorio de Gerociencias, Laboratorio Facilitador, Laboratorio de Medicina Genómica, Dirección General, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico
| | - Carlos Pineda
- Laboratorio de Gerociencias, Laboratorio Facilitador, Laboratorio de Medicina Genómica, Dirección General, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico
| | - Marcela Vela-Amieva
- Laboratorio de Errores Innatos del Metabolismo y Tamiz, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México, Mexico
| | - Alberto López-Reyes
- Laboratorio de Gerociencias, Laboratorio Facilitador, Laboratorio de Medicina Genómica, Dirección General, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico
| | | |
Collapse
|
91
|
Pharmacological Profile of Nigella sativa Seeds in Combating COVID-19 through In-Vitro and Molecular Docking Studies. Processes (Basel) 2022. [DOI: 10.3390/pr10071346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
COVID-19 infection is associated with elevated oxidative stress, systemic hyper-inflammatory responses, endothelial dysfunction, and red blood cell membrane deformability. Nigella sativa extract is widely used in alternative and complementary medicine systems in a large population, due to its highly therapeutic, economic, natural, and safe nature. The aim of this study was to evaluate the effect of N. sativa extract on oxidative stress, hemolysis, proteolysis, and glycation through in vitro studies, as well as to find out its anti-viral potential against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) using in silico studies. N. sativa seed extract (at 600 µg/mL) displayed 67.33% scavenging activity in the 2,2-diphenyl-1-picrylhydrazyl (DPPH) test, and 70.28% hydrogen peroxide reducing activity. N. sativa exhibited anti-proteolytic activity by decreasing heat-induced denaturation of bovine serum albumin (BSA) and egg albumin by 63.14% and 57.95%, respectively, and exhibited anti-proteinase potential of 66.28% at 600 μg/mL. In addition, heat-induced hemolysis and hypersalinity-induced hemolysis were inhibited by 57.86% and 61.7%, respectively, by the N. sativa seeds. N. sativa also inhibited browning intensity by 56.38%, and percent aggregation index by 51.38%, amyloid structure by 48.28%, and AGE-specific fluorescence by 52.18%, thereby protecting the native structure of BSA from glycation. The binding interactions between bioactive molecules of N. sativa seed with SARS-CoV-2 spike glycoprotein were proven by using in silico molecular docking tools. The functional amino acids involved in the interactions are Asp467, Thr108, Thr114, Ile468, Asn234, Gln155, Glu465, Arg466, Gly232, and Ile233, indicating the inhibiting property of N. sativa on SARS-CoV-2. Finally, we may infer that phytoconstituents of N. sativa seeds have the potential to protect against the spike protein of SARS-CoV-2. Studies on N. sativa seeds might act as a path to develop a potent alternative therapy against viral infections, especially COVID-19 infection, in the future. However, the limitations linked with the use of natural products are also needed to be considered in this regard.
Collapse
|
92
|
Aminianfar M, Soleiman-Meigooni S, Hamidi-Farahani R, Darvishi M, Hoseini-Shokouh SJ, Asgari A, Faraji-Hormozi S, Asli M. Efficacy of Red Blood Cell Exchange as Adjunctive Treatment for Hypoxemia and Survival Rate of Patients With Severe Coronavirus-2 Disease: An Open-Labeled Phase 2 Randomized Clinical Trial. Front Med (Lausanne) 2022; 9:899593. [PMID: 35872770 PMCID: PMC9304762 DOI: 10.3389/fmed.2022.899593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Background Severe acute respiratory syndrome (SARS) coronavirus-2 may infect red blood cells (RBCs) and impact oxygenation. We aimed to evaluate the efficacy of RBC exchange as an adjunctive treatment for hypoxemia and the survival rate of patients with severe coronavirus disease 2019 (COVID-19). Methods In a randomized clinical trial, we divided sixty patients with severe COVID-19 into two groups. The intervention group received the standard treatment of severe COVID-19 with RBC exchange three to four times in 2 days. The control group only received the standard treatment. Our primary outcomes were improving hypoxemia in 7 days, recovery or discharge, and death in 28 days. We conducted Chi-square test, independent samples t-test, and Fisher’s exact test to analyze the results. The ethical committee of Aja University of Medical Sciences approved the study (IR.AJAUMS.REC.1399.054), and the Iranian clinical trial registration organization registered it (IRCT20160316027081N2). Results Twenty-nine men and thirty-one women with a mean age of 67.5 years entered the study. The frequency of hypertension and diabetes mellitus was 86.7 and 68.3%, respectively. The most common symptoms of severe COVID-19 were dyspnea (91.6%), cough (75%), and fever (66.6%). Our results showed that hypoxemia improved in 21 of the 30 patients (70%) in the intervention group and 10 of the 30 patients (33.3%) in the control group (P < 0.004). The recovery and discharge rates were 19 of 30 patients (63.3%) in the intervention group and 2 of 30 patients (6.7%) in the control group (P < 0.001). Conclusion The RBC exchange improved the oxygenation and survival rate in patients with severe COVID-19.
Collapse
|
93
|
Hao M, Wang D, Xia Q, Kan S, Chang L, Liu H, Yang Z, Liu W. Pathogenic Mechanism and Multi-omics Analysis of Oral Manifestations in COVID-19. Front Immunol 2022; 13:879792. [PMID: 35860279 PMCID: PMC9290522 DOI: 10.3389/fimmu.2022.879792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 06/10/2022] [Indexed: 11/16/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a respiratory infectious disease that seriously threatens human life. The clinical manifestations of severe COVID-19 include acute respiratory distress syndrome and multiple organ failure. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causal agent of COVID-19, spreads through contaminated droplets. SARS-CoV-2 particles have been detected in the saliva of COVID-19 patients, implying that the virus can infect and damage the oral cavity. The oral manifestations of COVID-19 include xerostomia and gustatory dysfunction. Numerous studies showed that the four structural proteins of SARS-CoV-2 are its potential pathogenic factors, especially the S protein, which binds to human ACE2 receptors facilitating the entry of the virus into the host cells. Usually, upon entry into the host cell, a pathogen triggers the host’s immune response. However, a mount of multi-omics and immunological analyses revealed that COVID-19 is caused by immune dysregulation. A decrease in the number and phenotypes of immune cells, IFN-1 production and excessive release of certain cytokines have also been reported. In conclusion, this review summarizes the oral manifestations of COVID-19 and multi-omics analysis of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Ming Hao
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China
| | - Dongxu Wang
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Qianyun Xia
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Shaoning Kan
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China
| | - Lu Chang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China
| | - Huimin Liu
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China
| | - Zhijing Yang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China
| | - Weiwei Liu
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
- *Correspondence: Weiwei Liu,
| |
Collapse
|
94
|
Marchi G, Bozzini C, Bertolone L, Dima F, Busti F, Castagna A, Stranieri C, Fratta Pasini AM, Friso S, Lippi G, Girelli D, Vianello A. Red Blood Cell Morphologic Abnormalities in Patients Hospitalized for COVID-19. Front Physiol 2022; 13:932013. [PMID: 35860651 PMCID: PMC9289213 DOI: 10.3389/fphys.2022.932013] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/14/2022] [Indexed: 12/16/2022] Open
Abstract
Peripheral blood smear is a simple laboratory tool, which remains of invaluable help for diagnosing primary and secondary abnormalities of blood cells despite advances in automated and molecular techniques. Red blood cells (RBCs) abnormalities are known to occur in many viral infections, typically in the form of mild normo-microcytic anemia. While several hematological alterations at automated complete blood count (including neutrophilia, lymphopenia, and increased red cell distribution width—RDW) have been consistently associated with severity of COVID-19, there is scarce information on RBCs morphological abnormalities, mainly as case-reports or small series of patients, which are hardly comparable due to heterogeneity in sampling times and definition of illness severity. We report here a systematic evaluation of RBCs morphology at peripheral blood smear in COVID-19 patients within the first 72 h from hospital admission. One hundred and fifteen patients were included, with detailed collection of other clinical variables and follow-up. A certain degree of abnormalities in RBCs morphology was observed in 75 (65%) patients. Heterogenous alterations were noted, with spiculated cells being the more frequent morphology. The group with >10% RBCs abnormalities had more consistent lymphopenia and thrombocytopenia compared to those without abnormalities or <10% RBCs abnormalities (p < 0.018, and p < 0.021, respectively), thus underpinning a possible association with an overall more sustained immune-inflammatory “stress” hematopoiesis. Follow-up analysis showed a different mortality rate across groups, with the highest rate in those with more frequent RBCs morphological alterations compared to those with <10% or no abnormalities (41.9%, vs. 20.5%, vs. 12.5%, respectively, p = 0.012). Despite the inherent limitations of such simple association, our results point out towards further studies on erythropoiesis alterations in the pathophysiology of COVID-19.
Collapse
Affiliation(s)
- Giacomo Marchi
- Section of Internal Medicine, Department of Medicine, University of Verona, Verona, Italy
- EuroBloodNet Referral Center for Rare Hematological Disorders, University of Verona, Verona, Italy
| | - Claudia Bozzini
- Section of Internal Medicine, Department of Medicine, University of Verona, Verona, Italy
- EuroBloodNet Referral Center for Rare Hematological Disorders, University of Verona, Verona, Italy
| | - Lorenzo Bertolone
- Section of Internal Medicine, Department of Medicine, University of Verona, Verona, Italy
- EuroBloodNet Referral Center for Rare Hematological Disorders, University of Verona, Verona, Italy
| | - Francesco Dima
- Section of Clinical Biochemistry, University of Verona, Verona, Italy
| | - Fabiana Busti
- Section of Internal Medicine, Department of Medicine, University of Verona, Verona, Italy
- EuroBloodNet Referral Center for Rare Hematological Disorders, University of Verona, Verona, Italy
| | - Annalisa Castagna
- Section of Internal Medicine, Department of Medicine, University of Verona, Verona, Italy
- EuroBloodNet Referral Center for Rare Hematological Disorders, University of Verona, Verona, Italy
| | - Chiara Stranieri
- Section of Internal Medicine, Department of Medicine, University of Verona, Verona, Italy
- EuroBloodNet Referral Center for Rare Hematological Disorders, University of Verona, Verona, Italy
| | | | - Simonetta Friso
- Section of Internal Medicine, Department of Medicine, University of Verona, Verona, Italy
| | - Giuseppe Lippi
- Section of Clinical Biochemistry, University of Verona, Verona, Italy
| | - Domenico Girelli
- Section of Internal Medicine, Department of Medicine, University of Verona, Verona, Italy
- EuroBloodNet Referral Center for Rare Hematological Disorders, University of Verona, Verona, Italy
| | - Alice Vianello
- Section of Internal Medicine, Department of Medicine, University of Verona, Verona, Italy
- EuroBloodNet Referral Center for Rare Hematological Disorders, University of Verona, Verona, Italy
- *Correspondence: Alice Vianello, , orcid.org/0000-0002-2428-4760
| |
Collapse
|
95
|
Hafez W, Ziade MA, Arya A, Saleh H, Abdelrahman A. The significance of antiglobulin (Coombs) test reactivity in patients with COVID-19. Immunobiology 2022; 227:152240. [PMID: 35839729 PMCID: PMC9258419 DOI: 10.1016/j.imbio.2022.152240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 06/17/2022] [Accepted: 07/02/2022] [Indexed: 12/15/2022]
Abstract
Previous case reports have described patients with COVID-19-associated autoimmune hemolytic anemia (AIHA), and cold agglutinin disease (CAD) which is characterized by a positive direct antiglobulin (DAT) or "Coombs" test, yet the mechanism is not well understood. To investigate the significance of Coombs test reactivity among COVID-19 patients, we conducted a retrospective study on hospitalized COVID-19 patients treated at NMC Royal Hospital between 15 April and 30 May 2020. There were 27 (20%) patients in the Coombs-positive group and 108 (80%) in the Coombs-negative group. The cold agglutinin titer was examined in 22 patients due to symptoms suggestive of cold agglutinin disease, and all tested negative. We demonstrated a significant association with reactive Coombs test results in univariate analysis through clinical findings such as ICU admission rate, the severity of COVID-19, and several laboratory findings such as CRP, D-dimer, and hemoglobin levels lactate dehydrogenase, and RDW-CV. However, only hemoglobin levels and disease severity had a statistically significant association in multivariate analysis. A possible explanation of COVID-19-associated positive Coombs is cytokine storm-induced hyperinflammation, complement system activation, alterations of RBCs, binding of SARS-CoV-2 proteins to hemoglobin or its metabolites, and autoantibody production. Coombs-positive patients were tested for hemolysis using indirect bilirubin, consumed haptoglobin, and/or peripheral smear that ruled out any evidence of hemolysis. Understanding this etiology sheds new light on RBC involvement as a pathophysiological target for SARS-CoV-2 by interfering with their function; consequently, therapies capable of restoring RBC function, such as erythrocytapheresis, could be repurposed for the treatment of worsening severe and critical COVID-19.
Collapse
Affiliation(s)
- Wael Hafez
- NMC Royal Hospital, 16th Street, Khalifa City, Abu Dhabi, United Arab Emirates; Medical Research Division, Department of Internal Medicine, The National Research Center, Cairo, Egypt.
| | - Mohamad Azzam Ziade
- NMC Royal Hospital, 16th Street, Khalifa City, Abu Dhabi, United Arab Emirates
| | - Arun Arya
- NMC Royal Hospital, 16th Street, Khalifa City, Abu Dhabi, United Arab Emirates
| | - Husam Saleh
- NMC Royal Hospital, 16th Street, Khalifa City, Abu Dhabi, United Arab Emirates
| | - Ahmed Abdelrahman
- NMC Royal Hospital, 16th Street, Khalifa City, Abu Dhabi, United Arab Emirates; Internal Medicine Department, Zagazig School of Medicine, Zagazig, Egypt
| |
Collapse
|
96
|
Soma P, Bester J. Pathophysiological Changes in Erythrocytes Contributing to Complications of Inflammation and Coagulation in COVID-19. Front Physiol 2022; 13:899629. [PMID: 35784888 PMCID: PMC9240594 DOI: 10.3389/fphys.2022.899629] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/26/2022] [Indexed: 12/15/2022] Open
Abstract
Higher thrombotic burden in the acute phase of COVID-19 relies on a complex interplay between pro-inflammatory cytokine/chemokine release, increased endothelial dysfunction/damage, and potential sepsis-induced coagulopathy development in severe cases, all promoting coagulation activation. Plasma levels of cytokines and chemokines are known to be increased in COVID-19 however, are much higher in severe infections. Increased levels of IL-1β, IL-6, and IL-8 are known to play an important role in both acute and chronic inflammation, resulting in pathological clotting. However, little has been published on the effects of these interleukins on red blood cells (RBCs). Evidence shows that cytokines have a negative effect on the RBCs ultrastructure and introduce signs of eryptosis. Eryptosis can be described as a form of suicidal death of RBCs characterized by distinct findings of cell shrinkage, membrane blebbing, activation of proteases, and phosphatidylserine exposure at the outer membrane leaflet. Red blood cells from COVID-19 patients had increased levels of glycolytic intermediates, accompanied by oxidation and fragmentation of ankyrin, spectrin beta, and the N-terminal cytosolic domain of band 3 (AE1). Significantly altered lipid metabolism was also observed, in particular, short- and medium-chain saturated fatty acids, acyl-carnitines, and sphingolipids. Emerging research suggests that RBCs may contribute to a precision medicine approach to sepsis and have diagnostic value in monitoring complement dysregulation in COVID-19-sepsis and non-COVID sepsis as research indicates that complement activation products and viral antigens are present on RBCs in patients with COVID-19.
Collapse
Affiliation(s)
- Prashilla Soma
- Department of Anatomy, University of Pretoria, Pretoria, South Africa
- *Correspondence: Prashilla Soma,
| | - Janette Bester
- Department of Physiology, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
97
|
Böning D, Bloch W, Kuebler WM. Comment on Ceruti et al. Temporal Changes in the Oxyhemoglobin Dissociation Curve of Critically Ill COVID-19 Patients. J. Clin. Med. 2022, 11, 788. J Clin Med 2022; 11:3376. [PMID: 35743447 PMCID: PMC9225162 DOI: 10.3390/jcm11123376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/05/2022] [Accepted: 06/09/2022] [Indexed: 02/01/2023] Open
Abstract
Ceruti et al. describe in their article very low standard half saturation pressures (P50) in COVID-19 patients, calculated with the Dash et al. equations. By using the Hill equation and Severinghaus' coefficients we obtained normal values. The authors who do not present a pathophysiological cause for their results should explain this discrepancy. Independent of the absolute values a continuous moderate decrease of P50 in the surviving patients might be of clinical importance.
Collapse
Affiliation(s)
- Dieter Böning
- Institute of Physiology, Campus Mitte, Charité Medical University of Berlin, 10117 Berlin, Germany;
| | - Wilhelm Bloch
- Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne, 50933 Cologne, Germany;
| | - Wolfgang M. Kuebler
- Institute of Physiology, Campus Mitte, Charité Medical University of Berlin, 10117 Berlin, Germany;
| |
Collapse
|
98
|
Prudinnik DS, Sinauridze EI, Shakhidzhanov SS, Bovt EA, Protsenko DN, Rumyantsev AG, Ataullakhanov FI. Filterability of Erythrocytes in Patients with COVID-19. Biomolecules 2022; 12:biom12060782. [PMID: 35740907 PMCID: PMC9220947 DOI: 10.3390/biom12060782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/27/2022] [Accepted: 06/01/2022] [Indexed: 12/02/2022] Open
Abstract
For the first time, the influence of COVID-19 on blood microrheology was studied. For this, the method of filtering erythrocytes through filters with pores of 3.5 μm was used. Filterability was shown to significantly decrease with the increasing severity of the patient’s condition, as well as with a decrease in the ratio of hemoglobin oxygen saturation to the oxygen fraction in the inhaled air (SpO2/FiO2). The filterability of ≤ 0.65, or its fast decrease during treatment, were indicators of a poor prognosis. Filterability increased significantly with an increase in erythrocyte count, hematocrit and blood concentrations of hemoglobin, albumin, and total protein. The effect of these parameters on the erythrocyte filterability is directly opposite to their effect on blood macrorheology, where they all increase blood viscosity, worsening the erythrocyte deformability. The erythrocyte filterability decreased with increasing oxygen supply rate, especially in patients on mechanical ventilation, apparently not due to the oxygen supplied, but to the deterioration of the patients’ condition. Filterability significantly correlates with the C-reactive protein, which indicates that inflammation affects the blood microrheology in the capillaries. Thus, the filterability of erythrocytes is a good tool for studying the severity of the patient’s condition and his prognosis in COVID-19.
Collapse
Affiliation(s)
- Dmitry S. Prudinnik
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Ministry of Healthcare of Russian Federation, Samory Mashela Str., 1, GSP-7, 117198 Moscow, Russia; (D.S.P.); (S.S.S.); (E.A.B.); (A.G.R.)
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Srednyaya Kalitnikovskaya Str., 30, 109029 Moscow, Russia
| | - Elena I. Sinauridze
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Ministry of Healthcare of Russian Federation, Samory Mashela Str., 1, GSP-7, 117198 Moscow, Russia; (D.S.P.); (S.S.S.); (E.A.B.); (A.G.R.)
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Srednyaya Kalitnikovskaya Str., 30, 109029 Moscow, Russia
- Correspondence: (E.I.S.); (F.I.A.)
| | - Soslan S. Shakhidzhanov
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Ministry of Healthcare of Russian Federation, Samory Mashela Str., 1, GSP-7, 117198 Moscow, Russia; (D.S.P.); (S.S.S.); (E.A.B.); (A.G.R.)
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Srednyaya Kalitnikovskaya Str., 30, 109029 Moscow, Russia
| | - Elizaveta A. Bovt
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Ministry of Healthcare of Russian Federation, Samory Mashela Str., 1, GSP-7, 117198 Moscow, Russia; (D.S.P.); (S.S.S.); (E.A.B.); (A.G.R.)
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Srednyaya Kalitnikovskaya Str., 30, 109029 Moscow, Russia
| | - Denis N. Protsenko
- City Clinical Hospital No. 40 Moscow Health Department, Kasatkin Str., 7, 129301 Moscow, Russia;
- Department of Anaesthesia and Critical Care, Pirogov Russian National Research Medical University, Ostrovityanov Str., 1, 117997 Moscow, Russia
| | - Alexander G. Rumyantsev
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Ministry of Healthcare of Russian Federation, Samory Mashela Str., 1, GSP-7, 117198 Moscow, Russia; (D.S.P.); (S.S.S.); (E.A.B.); (A.G.R.)
| | - Fazoil I. Ataullakhanov
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Ministry of Healthcare of Russian Federation, Samory Mashela Str., 1, GSP-7, 117198 Moscow, Russia; (D.S.P.); (S.S.S.); (E.A.B.); (A.G.R.)
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Srednyaya Kalitnikovskaya Str., 30, 109029 Moscow, Russia
- Department of Biophysics, Physics Faculty, Lomonosov Moscow State University, Leninskie Gory, 1, Build. 2, GSP-1, 119991 Moscow, Russia
- Moscow Institute of Physics and Technology, National Research University, Institutskiy Per., 9, 141701 Dolgoprudny, Russia
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd., Philadelphia, PA 19104, USA
- Correspondence: (E.I.S.); (F.I.A.)
| |
Collapse
|
99
|
Liang M, Liu D, Nie Y, Liu Y, Qiao X. Exploiting styrene-maleic acid copolymer grafting chromatographic stationary phase materials for separation of membrane lipids. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.10.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
100
|
Telek E, Ujfalusi Z, Kemenesi G, Zana B, Jakab F, Hild G, Lukács A, Hild G. A Possible Way to Relate the Effects of SARS-CoV-2-Induced Changes in Transferrin to Severe COVID-19-Associated Diseases. Int J Mol Sci 2022; 23:6189. [PMID: 35682873 PMCID: PMC9181396 DOI: 10.3390/ijms23116189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 02/04/2023] Open
Abstract
SARS-CoV-2 infections are responsible for the COVID-19 pandemic. Transferrin has been found to explain the link between diseases associated with impaired iron transport and COVID-19 infection. The effect of SARS-CoV-2 on human whole blood was studied by differential scanning calorimetry. The analysis of the thermal transition curves showed that the melting temperature of the transferrin-related peak decreased in the presence of SARS-CoV-2. The ratio of the under-curve area of the two main peaks was greatly affected, while the total enthalpy of the heat denaturation remained nearly unchanged in the presence of the virus. These results indicate that SARS-CoV-2, through binding to transferrin, may influence its Fe3+ uptake by inducing thermodynamic changes. Therefore, transferrin may remain in an iron-free apo-conformational state, which depends on the SARS-CoV-2 concentration. SARS-CoV-2 can induce disturbance in erythropoiesis due to toxicity generated by free iron overload.
Collapse
Affiliation(s)
- Elek Telek
- Department of Biophysics, Medical School, University of Pécs, Szigeti Str. 12, H-7624 Pécs, Hungary; (E.T.); (Z.U.); (A.L.)
| | - Zoltán Ujfalusi
- Department of Biophysics, Medical School, University of Pécs, Szigeti Str. 12, H-7624 Pécs, Hungary; (E.T.); (Z.U.); (A.L.)
| | - Gábor Kemenesi
- Szentágothai Research Centre, Virological Research Group, University of Pécs, Ifjúság Str. 20, H-7624 Pécs, Hungary; (G.K.); (B.Z.); (F.J.)
- Faculty of Sciences, Institute of Biology, University of Pécs, Ifjúság Str. 6, H-7624 Pécs, Hungary
- National Laboratory of Virology, University of Pécs, Ifjúság Str. 20, H-7624 Pécs, Hungary
| | - Brigitta Zana
- Szentágothai Research Centre, Virological Research Group, University of Pécs, Ifjúság Str. 20, H-7624 Pécs, Hungary; (G.K.); (B.Z.); (F.J.)
- Faculty of Sciences, Institute of Biology, University of Pécs, Ifjúság Str. 6, H-7624 Pécs, Hungary
- National Laboratory of Virology, University of Pécs, Ifjúság Str. 20, H-7624 Pécs, Hungary
| | - Ferenc Jakab
- Szentágothai Research Centre, Virological Research Group, University of Pécs, Ifjúság Str. 20, H-7624 Pécs, Hungary; (G.K.); (B.Z.); (F.J.)
- Faculty of Sciences, Institute of Biology, University of Pécs, Ifjúság Str. 6, H-7624 Pécs, Hungary
- National Laboratory of Virology, University of Pécs, Ifjúság Str. 20, H-7624 Pécs, Hungary
| | - Gabriella Hild
- Languages for Biomedical Purposes and Communication, Medical School, University of Pécs, Szigeti Str. 12, H-7624 Pécs, Hungary;
| | - András Lukács
- Department of Biophysics, Medical School, University of Pécs, Szigeti Str. 12, H-7624 Pécs, Hungary; (E.T.); (Z.U.); (A.L.)
| | - Gábor Hild
- Department of Biophysics, Medical School, University of Pécs, Szigeti Str. 12, H-7624 Pécs, Hungary; (E.T.); (Z.U.); (A.L.)
- Department of Medical Imaging, Clinical Centre, University of Pécs, Ifjúság Str. 13, H-7624 Pécs, Hungary
| |
Collapse
|