1
|
Roytenberg R, Yue H, DeHart A, Kim E, Bai F, Kim Y, Denning K, Kwei A, Zhang Q, Liu J, Zheng XL, Li W. Thymidine phosphorylase mediates SARS-CoV-2 spike protein enhanced thrombosis in K18-hACE2 TG mice. Thromb Res 2024; 244:109195. [PMID: 39442286 DOI: 10.1016/j.thromres.2024.109195] [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: 07/06/2024] [Revised: 09/09/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION Thymidine phosphorylase (TYMP), which facilitates platelet activation and thrombosis, is significantly increased in COVID-19 patients. We hypothesize that TYMP mediates SARS-CoV-2 spike protein (SP)-induced thrombosis. MATERIALS AND METHODS Plasmids encoding wildtype SP or empty vector (p3.1) were transfected into COS-7 cells, and cell lysates were prepared as a reservoir for SP or p3.1 (control), respectively. K18-hACE2TG and K18-hACE2TG/Tymp-/- mice were treated with a single dose of SP or p3.1 by intraperitoneal injection and then subjected to thrombosis studies three days later. The role of SP on inflammatory signaling activation was assessed in BEAS-2B cells. RESULTS SARS-CoV-2 SP increased the expression of TYMP, resulting in the activation of STAT3 and NF-κB in BEAS-2B cells. A siRNA-mediated knockdown of TYMP attenuated SP-enhanced activation of STAT3. SP significantly promoted arterial thrombosis in K18-hACE2TG mice. SP-accelerated thrombosis was attenuated by inhibition or genetic ablation of TYMP. SP treatment did not influence ADP- or collagen-induced platelet aggregation but significantly increased platelet adhesion to fibrinogen. SP treatment also significantly shortened activated partial thromboplastin time, which was reversed and even prolonged by TYMP deficiency. Additionally, SP binds to platelet factor 4 (PF4) and TYMP. TYMP does not bind PF4 but enhances the formation of the SP/PF4 complex, which may augment the procoagulant and prothrombotic effect of PF4. CONCLUSIONS We conclude that SP is prothrombotic and upregulates TYMP expression, and TYMP inhibition or knockout mitigates SP-enhanced thrombosis. These findings suggest that inhibition of TYMP may be a novel therapeutic strategy for COVID-19-associated thrombosis.
Collapse
Affiliation(s)
- Renat Roytenberg
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - Hong Yue
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - Autumn DeHart
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - Eugene Kim
- Department of Chemistry, College of Sciences, Marshall University, Huntington, WV 25755, USA
| | - Fang Bai
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - Yongick Kim
- Department of Chemistry, College of Sciences, Marshall University, Huntington, WV 25755, USA
| | - Krista Denning
- Department of Pathology, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - Alec Kwei
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - Quan Zhang
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jiang Liu
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - X Long Zheng
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Wei Li
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA.
| |
Collapse
|
2
|
Ryu JK, Yan Z, Montano M, Sozmen EG, Dixit K, Suryawanshi RK, Matsui Y, Helmy E, Kaushal P, Makanani SK, Deerinck TJ, Meyer-Franke A, Rios Coronado PE, Trevino TN, Shin MG, Tognatta R, Liu Y, Schuck R, Le L, Miyajima H, Mendiola AS, Arun N, Guo B, Taha TY, Agrawal A, MacDonald E, Aries O, Yan A, Weaver O, Petersen MA, Meza Acevedo R, Alzamora MDPS, Thomas R, Traglia M, Kouznetsova VL, Tsigelny IF, Pico AR, Red-Horse K, Ellisman MH, Krogan NJ, Bouhaddou M, Ott M, Greene WC, Akassoglou K. Fibrin drives thromboinflammation and neuropathology in COVID-19. Nature 2024; 633:905-913. [PMID: 39198643 PMCID: PMC11424477 DOI: 10.1038/s41586-024-07873-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 07/24/2024] [Indexed: 09/01/2024]
Abstract
Life-threatening thrombotic events and neurological symptoms are prevalent in COVID-19 and are persistent in patients with long COVID experiencing post-acute sequelae of SARS-CoV-2 infection1-4. Despite the clinical evidence1,5-7, the underlying mechanisms of coagulopathy in COVID-19 and its consequences in inflammation and neuropathology remain poorly understood and treatment options are insufficient. Fibrinogen, the central structural component of blood clots, is abundantly deposited in the lungs and brains of patients with COVID-19, correlates with disease severity and is a predictive biomarker for post-COVID-19 cognitive deficits1,5,8-10. Here we show that fibrin binds to the SARS-CoV-2 spike protein, forming proinflammatory blood clots that drive systemic thromboinflammation and neuropathology in COVID-19. Fibrin, acting through its inflammatory domain, is required for oxidative stress and macrophage activation in the lungs, whereas it suppresses natural killer cells, after SARS-CoV-2 infection. Fibrin promotes neuroinflammation and neuronal loss after infection, as well as innate immune activation in the brain and lungs independently of active infection. A monoclonal antibody targeting the inflammatory fibrin domain provides protection from microglial activation and neuronal injury, as well as from thromboinflammation in the lung after infection. Thus, fibrin drives inflammation and neuropathology in SARS-CoV-2 infection, and fibrin-targeting immunotherapy may represent a therapeutic intervention for patients with acute COVID-19 and long COVID.
Collapse
Affiliation(s)
- Jae Kyu Ryu
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Zhaoqi Yan
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Mauricio Montano
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
| | - Elif G Sozmen
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Karuna Dixit
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | | | - Yusuke Matsui
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
| | - Ekram Helmy
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
| | - Prashant Kaushal
- Department of Microbiology, Immunology and Molecular Genetics (MIMG), University of California Los Angeles, Los Angeles, CA, USA
- Institute for Quantitative and Computational Biosciences (QCBio), University of California Los Angeles, Los Angeles, CA, USA
| | - Sara K Makanani
- Department of Microbiology, Immunology and Molecular Genetics (MIMG), University of California Los Angeles, Los Angeles, CA, USA
- Institute for Quantitative and Computational Biosciences (QCBio), University of California Los Angeles, Los Angeles, CA, USA
| | - Thomas J Deerinck
- National Center for Microscopy and Imaging Research, Center for Research on Biological Systems, University of California San Diego, La Jolla, CA, USA
| | | | | | - Troy N Trevino
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Min-Gyoung Shin
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
| | - Reshmi Tognatta
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Yixin Liu
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Renaud Schuck
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Lucas Le
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Hisao Miyajima
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Andrew S Mendiola
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Nikhita Arun
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Brandon Guo
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Taha Y Taha
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
| | - Ayushi Agrawal
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
| | - Eilidh MacDonald
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Oliver Aries
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Aaron Yan
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Olivia Weaver
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Mark A Petersen
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Rosa Meza Acevedo
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Maria Del Pilar S Alzamora
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Reuben Thomas
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
| | - Michela Traglia
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
| | - Valentina L Kouznetsova
- San Diego Supercomputer Center, University of California San Diego, La Jolla, CA, USA
- CureScience Institute, San Diego, CA, USA
| | - Igor F Tsigelny
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- San Diego Supercomputer Center, University of California San Diego, La Jolla, CA, USA
- CureScience Institute, San Diego, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Alexander R Pico
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
| | - Kristy Red-Horse
- Department of Biology, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Mark H Ellisman
- National Center for Microscopy and Imaging Research, Center for Research on Biological Systems, University of California San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Nevan J Krogan
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA
- Quantitative Biosciences Institute (QBI), University of California San Francisco, San Francisco, CA, USA
- COVID-19 Research Group (QCRG), University of California San Francisco, San Francisco, CA, USA
| | - Mehdi Bouhaddou
- Department of Microbiology, Immunology and Molecular Genetics (MIMG), University of California Los Angeles, Los Angeles, CA, USA
- Institute for Quantitative and Computational Biosciences (QCBio), University of California Los Angeles, Los Angeles, CA, USA
| | - Melanie Ott
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
- COVID-19 Research Group (QCRG), University of California San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Warner C Greene
- Gladstone Institute of Virology, San Francisco, CA, USA.
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA.
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA.
| | - Katerina Akassoglou
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA.
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA.
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
3
|
Harte JV, Coleman-Vaughan C, Crowley MP, Mykytiv V. It's in the blood: a review of the hematological system in SARS-CoV-2-associated COVID-19. Crit Rev Clin Lab Sci 2023; 60:595-624. [PMID: 37439130 DOI: 10.1080/10408363.2023.2232010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/27/2023] [Indexed: 07/14/2023]
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to an unprecedented global healthcare crisis. While SARS-CoV-2-associated COVID-19 affects primarily the respiratory system, patients with COVID-19 frequently develop extrapulmonary manifestations. Notably, changes in the hematological system, including lymphocytopenia, neutrophilia and significant abnormalities of hemostatic markers, were observed early in the pandemic. Hematological manifestations have since been recognized as important parameters in the pathophysiology of SARS-CoV-2 and in the management of patients with COVID-19. In this narrative review, we summarize the state-of-the-art regarding the hematological and hemostatic abnormalities observed in patients with SARS-CoV-2-associated COVID-19, as well as the current understanding of the hematological system in the pathophysiology of acute and chronic SARS-CoV-2-associated COVID-19.
Collapse
Affiliation(s)
- James V Harte
- Department of Haematology, Cork University Hospital, Wilton, Cork, Ireland
- School of Biochemistry & Cell Biology, University College Cork, Cork, Ireland
| | | | - Maeve P Crowley
- Department of Haematology, Cork University Hospital, Wilton, Cork, Ireland
- Irish Network for Venous Thromboembolism Research (INViTE), Ireland
| | - Vitaliy Mykytiv
- Department of Haematology, Cork University Hospital, Wilton, Cork, Ireland
| |
Collapse
|
4
|
Jegorović B, Nikolić A, Milinković N, Ignjatović S, Šipetić-Grujičić S. The utility of serum amyloid A and other acute-phase reactants determination in ambulatory care COVID-19 patients. J Med Biochem 2023; 42:492-504. [PMID: 37790210 PMCID: PMC10542288 DOI: 10.5937/jomb0-42799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/13/2023] [Indexed: 10/05/2023] Open
Abstract
Background The unpredictable course of Coronavirus Disease 19 (COVID-19) is making good severity assessment tools crucial. This study aimed to assess the usefulness of serum amyloid A (SAA) and other acute-phase reactants (APRs) in ambulatory care COVID-19 patients and identified relationships between these markers and disease outcomes. Methods From August to November 2020, patients seen in the outpatient department of the Clinic for Infectious and Tropical Diseases (Belgrade, Serbia) with confirmed COVID-19 were included. Patients were classified into mild, moderate, and severe disease groups based on World Health Organization criteria. SAA, C-reactive protein (CRP), interleukin-6 (IL-6), procalcitonin (PCT), ferritin, fibrinogen, D-dimer, albumin, and transferrin were measured. The median values of all APRs were compared between COVID-19 severity groups, hospitalized and non-hospitalized patients, and survivors and non-survivors. The Receiver operator characteristic (ROC) curve analysis was used for the classification characteristics assessment of individual APRs for the severity of illness, hospitalization, and survival.
Collapse
Affiliation(s)
- Boris Jegorović
- University Clinical Center of Serbia, Clinic for Infectious and Tropical Diseases "Prof. Dr. Kosta Todorović", Belgrade
| | - Aleksandra Nikolić
- University of Belgrade, Faculty of Medicine, Institute for Epidemiology, Belgrade
| | - Neda Milinković
- University of Belgrade, Faculty of Pharmacy, Department of Medical Biochemistry, Belgrade
| | - Svetlana Ignjatović
- University of Belgrade, Faculty of Pharmacy, Department of Medical Biochemistry, Belgrade
| | | |
Collapse
|
5
|
Hahn J, Bressler J, Domingo-Relloso A, Chen MH, McCartney DL, Teumer A, van Dongen J, Kleber ME, Aïssi D, Swenson BR, Yao J, Zhao W, Huang J, Xia Y, Brown MR, Costeira R, de Geus EJC, Delgado GE, Dobson DA, Elliott P, Grabe HJ, Guo X, Harris SE, Huffman JE, Kardia SLR, Liu Y, Lorkowski S, Marioni RE, Nauck M, Ratliff SM, Sabater-Lleal M, Spector TD, Suchon P, Taylor KD, Thibord F, Trégouët DA, Wiggins KL, Willemsen G, Bell JT, Boomsma DI, Cole SA, Cox SR, Dehghan A, Greinacher A, Haack K, März W, Morange PE, Rotter JI, Sotoodehnia N, Tellez-Plaza M, Navas-Acien A, Smith JA, Johnson AD, Fornage M, Smith NL, Wolberg AS, Morrison AC, de Vries PS. DNA methylation analysis is used to identify novel genetic loci associated with circulating fibrinogen levels in blood. J Thromb Haemost 2023; 21:1135-1147. [PMID: 36716967 PMCID: PMC11556295 DOI: 10.1016/j.jtha.2023.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 11/04/2022] [Accepted: 01/17/2023] [Indexed: 01/30/2023]
Abstract
BACKGROUND Fibrinogen plays an essential role in blood coagulation and inflammation. Circulating fibrinogen levels may be determined based on interindividual differences in DNA methylation at cytosine-phosphate-guanine (CpG) sites and vice versa. OBJECTIVES To perform an EWAS to examine an association between blood DNA methylation levels and circulating fibrinogen levels to better understand its biological and pathophysiological actions. METHODS We performed an epigenome-wide association study of circulating fibrinogen levels in 18 037 White, Black, American Indian, and Hispanic participants, representing 14 studies from the Cohorts for Heart and Aging Research in Genomic Epidemiology consortium. Circulating leukocyte DNA methylation was measured using the Illumina 450K array in 12 904 participants and using the EPIC array in 5133 participants. In each study, an epigenome-wide association study of fibrinogen was performed using linear mixed models adjusted for potential confounders. Study-specific results were combined using array-specific meta-analysis, followed by cross-replication of epigenome-wide significant associations. We compared models with and without CRP adjustment to examine the role of inflammation. RESULTS We identified 208 and 87 significant CpG sites associated with fibrinogen levels from the 450K (p < 1.03 × 10-7) and EPIC arrays (p < 5.78 × 10-8), respectively. There were 78 associations from the 450K array that replicated in the EPIC array and 26 vice versa. After accounting for overlapping sites, there were 83 replicated CpG sites located in 61 loci, of which only 4 have been previously reported for fibrinogen. The examples of genes located near these CpG sites were SOCS3 and AIM2, which are involved in inflammatory pathways. The associations of all 83 replicated CpG sites were attenuated after CRP adjustment, although many remained significant. CONCLUSION We identified 83 CpG sites associated with circulating fibrinogen levels. These associations are partially driven by inflammatory pathways shared by both fibrinogen and CRP.
Collapse
Affiliation(s)
- Julie Hahn
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA.
| | - Jan Bressler
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Arce Domingo-Relloso
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, New York, USA; Department of Chronic Diseases Epidemiology, National Center for Epidemiology, Carlos III Health Institutes, Madrid, Spain; Department of Statistics and Operations Research, University of Valencia, Burjassot, Spain
| | - Ming-Huei Chen
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, Framingham, Massachusetts, USA
| | - Daniel L McCartney
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Alexander Teumer
- Department SHIP/Clinical-Epidemiological Research, Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany; Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Bialystok, Poland
| | - Jenny van Dongen
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Marcus E Kleber
- Vth Department of Medicine, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; SYNLAB MVZ Humangenetik Mannheim, Mannheim, Germany
| | - Dylan Aïssi
- Univ. Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR 1219, Molecular Epidemiology of Vascular and Brain Disorders, Bordeaux, France
| | - Brenton R Swenson
- Cardiovascular Health Research Unit, School of Public Health, University of Washington, Seattle, Washington, USA
| | - Jie Yao
- Pediatrics, Genomic Outcomes, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA; Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Jian Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
| | - Yujing Xia
- Department of Twin Research and Genetic Epidemiology, St Thomas Hospital Campus, King's College London, London, United Kingdom
| | - Michael R Brown
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Ricardo Costeira
- Department of Twin Research and Genetic Epidemiology, St Thomas Hospital Campus, King's College London, London, United Kingdom
| | - Eco J C de Geus
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Graciela E Delgado
- Vth Department of Medicine, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Dre'Von A Dobson
- Pathology and Laboratory Medicine and UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, United Kingdom; UK Dementia Research Institute, Imperial College London, London, United Kingdom; British Heart Foundation Centre for Research Excellence, Imperial College London, London, United Kingdom
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany; German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Greifswald, Germany
| | - Xiuqing Guo
- Pediatrics, Genomic Outcomes, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Sarah E Harris
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, United Kingdom
| | - Jennifer E Huffman
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, Massachusetts, USA
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Yongmei Liu
- Medicine, Cardiology, Duke Molecular Physiology Institute, Durham, North Carolina, USA
| | - Stefan Lorkowski
- Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany; Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Jena, Germany
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Matthias Nauck
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany; Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Scott M Ratliff
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Maria Sabater-Lleal
- Genomics of Complex Disease Unit, Sant Pau Biomedical Research Institute (IIB Sant Pau), Barcelona, Spain; Department of Medicine, Cardiovascular Medicine Unit, Karolinska Institutet, Stockholm, Sweden
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, St Thomas Hospital Campus, King's College London, London, United Kingdom
| | - Pierre Suchon
- Center for CardioVascular and Nutrition research (C2VN), INSERM 1263, INRAE 1260, Hematology Laboratory, La Timone University Hospital of Marseille, Aix-Marseille University, Marseille, France
| | - Kent D Taylor
- Pediatrics, Genomic Outcomes, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Florian Thibord
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, USA
| | - David-Alexandre Trégouët
- Univ. Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR 1219, Molecular Epidemiology of Vascular and Brain Disorders, Bordeaux, France
| | - Kerri L Wiggins
- Department of Medicine, Division of General Internal Medicine, University of Washington, Seattle, Washington, USA
| | - Gonneke Willemsen
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Jordana T Bell
- Department of Twin Research and Genetic Epidemiology, St Thomas Hospital Campus, King's College London, London, United Kingdom
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Shelley A Cole
- Population Health Program, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Simon R Cox
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, United Kingdom
| | - Abbas Dehghan
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
| | - Andreas Greinacher
- Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Karin Haack
- Population Health Program, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Winfried März
- Vth Department of Medicine, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; SYNLAB Academy, SYNLAB Holding Deutschland GmbH, Mannheim, Germany; Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Pierre-Emmanuel Morange
- Cardiovascular and Nutrition Reserach Center (C2VN), INSERM, INRAE, Aix-Marseille University, Marseille, France
| | - Jerome I Rotter
- Pediatrics, Genomic Outcomes, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Nona Sotoodehnia
- Department of Medicine, Division of Cardiology, University of Washington, Seattle, Washington, USA
| | - Maria Tellez-Plaza
- Department of Chronic Diseases Epidemiology, National Center for Epidemiology, Carlos III Health Institutes, Madrid, Spain
| | - Ana Navas-Acien
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, New York, USA
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA; Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Andrew D Johnson
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, Framingham, Massachusetts, USA
| | - Myriam Fornage
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA; Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Nicholas L Smith
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, Washington, USA; Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, Washington, USA; Seattle Epidemiologic Research and Information Center, Department of Veterans Affairs Office of Research and Development, Seattle, Washington, USA
| | - Alisa S Wolberg
- Pathology and Laboratory Medicine and UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Paul S de Vries
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA.
| |
Collapse
|
6
|
Schmidt W, Pawlak-Buś K, Jóźwiak B, Leszczyński P. Identification of Clinical Response Predictors of Tocilizumab Treatment in Patients with Severe COVID-19 Based on Single-Center Experience. J Clin Med 2023; 12:jcm12062429. [PMID: 36983429 PMCID: PMC10051490 DOI: 10.3390/jcm12062429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Hyperinflammation in COVID-19 plays a crucial role in pathogenesis and severity; thus, many immunomodulatory agents are applied in its treatment. We aimed to identify good clinical response predictors of tocilizumab (TCZ) treatment in severe COVID-19, among clinical, laboratory, and radiological variables. We conducted a prospective, observational study with 120 patients with severe COVID-19 not improving despite dexamethasone (DEX) treatment. We used parametric and non-parametric statistics, univariate logistic regression, receiver operating characteristic (ROC) curves, and nonlinear factors tertile analysis. In total, 86 (71.7%) patients achieved the primary outcome of a good clinical response to TCZ. We identified forty-nine predictive factors with potential utility in patient selection and treatment monitoring. The strongest included time from symptom onset between 9 and 12 days, less than 70% of estimated radiological lung involvement, and lower activity of lactate dehydrogenase. Additional predictors were associated with respiratory function, vitamin D concentration, comorbidities, and inflammatory/organ damage biomarkers. Adverse events analysis proved the safety of such a regimen. Our study confirmed that using TCZ early in the hyperinflammatory phase, before severe respiratory failure development, is most beneficial. Considering the described predictive factors, employing simple and widely available laboratory, radiological, and clinical tools can optimize patient selection for immunomodulatory treatment with TCZ.
Collapse
Affiliation(s)
- Wiktor Schmidt
- Department of Rheumatology, Systemic Connective Tissue Diseases and Immunotherapy of Rheumatic Diseases, J. Strus Municipal Hospital, 61-285 Poznan, Poland
- Department of Internal Medicine, Poznan University of Medical Sciences, 61-701 Poznan, Poland
- Doctoral School, Poznan University of Medical Sciences, 60-812 Poznan, Poland
| | - Katarzyna Pawlak-Buś
- Department of Rheumatology, Systemic Connective Tissue Diseases and Immunotherapy of Rheumatic Diseases, J. Strus Municipal Hospital, 61-285 Poznan, Poland
- Department of Internal Medicine, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Barbara Jóźwiak
- Department of Rheumatology, Systemic Connective Tissue Diseases and Immunotherapy of Rheumatic Diseases, J. Strus Municipal Hospital, 61-285 Poznan, Poland
| | - Piotr Leszczyński
- Department of Rheumatology, Systemic Connective Tissue Diseases and Immunotherapy of Rheumatic Diseases, J. Strus Municipal Hospital, 61-285 Poznan, Poland
- Department of Internal Medicine, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| |
Collapse
|
7
|
Zhang P, Liu Y, Li C, Stine LD, Wang PH, Turnbull MW, Wu H, Liu Q. Ectopic expression of SARS-CoV-2 S and ORF-9B proteins alters metabolic profiles and impairs contractile function in cardiomyocytes. Front Cell Dev Biol 2023; 11:1110271. [PMID: 36910162 PMCID: PMC9994814 DOI: 10.3389/fcell.2023.1110271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/30/2023] [Indexed: 02/25/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is associated with adverse impacts in the cardiovascular system, but the mechanisms driving this response remain unclear. In this study, we conducted "pseudoviral infection" of SARS-CoV-2 subunits to evaluate their toxic effects in cardiomyocytes (CMs), that were derived from human induced pluripotent stem cells (hiPSCs). We found that the ectopic expression of S and ORF-9B subunits significantly impaired the contractile function and altered the metabolic profiles in human cardiomyocytes. Further mechanistic study has shown that the mitochondrial oxidative phosphorylation (OXPHOS), membrane potential, and ATP production were significantly decreased two days after the overexpression of S and ORF-9B subunits, while S subunits induced higher level of reactive oxygen species (ROS). Two weeks after overexpression, glycolysis was elevated in the ORF-9B group. Based on the transcriptomic analysis, both S and ORF-9B subunits dysregulated signaling pathways associated with metabolism and cardiomyopathy, including upregulated genes involved in HIF-signaling and downregulated genes involved in cholesterol biosynthetic processes. The ORF-9B subunit also enhanced glycolysis in the CMs. Our results collectively provide an insight into the molecular mechanisms underlying SARS-CoV-2 subunits-induced metabolic alterations and cardiac dysfunctions in the hearts of COVID-19 patients.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Biological Sciences, Clemson University, Clemson, SC, United States
| | - Yu Liu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States
| | - Chunfeng Li
- Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, CA, United States
| | - Lindsay D. Stine
- Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Pei-Hui Wang
- Key Laboratory for Experimental Teratology of Ministry of Education and Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Matthew W. Turnbull
- Department of Biological Sciences, Clemson University, Clemson, SC, United States
| | - Haodi Wu
- Department of Medicine, Division of Cardiology, Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Qing Liu
- Department of Biological Sciences, Clemson University, Clemson, SC, United States
- Center for Human Genetics, Clemson University, Greenwood, SC, United States
| |
Collapse
|
8
|
Tauekelova AT, Kalila Z, Bakhtiyar A, Sautbayeva Z, Len P, Sailybayeva A, Khamitov S, Kadroldinova N, Barteneva NS, Bekbossynova MS. Association of Lung Fibrotic Changes and Cardiological Dysfunction with Comorbidities in Long COVID-19 Cohort. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:2567. [PMID: 36767932 PMCID: PMC9915134 DOI: 10.3390/ijerph20032567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/23/2022] [Accepted: 01/23/2023] [Indexed: 06/18/2023]
Abstract
Background. Long COVID-19 symptoms appeared in many COVID-19 survivors. However, the prevalence and symptoms associated with long COVID-19 and its comorbidities have not been established. Methods. In total, 312 patients with long COVID-19 from 21 primary care centers were included in the study. At the six-month follow-up, their lung function was assessed by computerized tomography (CT) and spirometry, whereas cardiac function was assessed by elec-trocardiogram (ECG), Holter ECG, echocardiography, 24 h blood pressure monitoring, and a six-minute walk test (6MWT). Results. Of the 312 persons investigated, significantly higher sys-tolic and diastolic blood pressure, left ventricular hypertrophy, and elevated NT-proBNP were revealed in participants with hypertension or type 2 diabetes. Left ventricular diastolic dysfunc-tion was more frequently present in patients with hypertension. The most common registered CT abnormalities were fibrotic changes (83, 36.6%) and mediastinal lymphadenopathy (23, 10.1%). Among the tested biochemical parameters, three associations were found in long COVID-19 patients with hypertension but not diabetes: increased hemoglobin, fibrinogen, and ferritin. Nine patients had persisting IgM antibodies to SARS-CoV-2. Conclusions. We demon-strated a strong association between signs of cardiac dysfunction and lung fibrotic changes with comorbidities in a cohort of long COVID-19 subjects.
Collapse
Affiliation(s)
| | - Zhanar Kalila
- National Research Center for Cardiac Surgery, Astana 010000, Kazakhstan
| | - Akerke Bakhtiyar
- National Research Center for Cardiac Surgery, Astana 010000, Kazakhstan
| | - Zarina Sautbayeva
- School of Sciences and Humanities, Nazarbayev University, Astana 010000, Kazakhstan
| | - Polina Len
- School of Sciences and Humanities, Nazarbayev University, Astana 010000, Kazakhstan
| | - Aliya Sailybayeva
- National Research Center for Cardiac Surgery, Astana 010000, Kazakhstan
| | - Sadyk Khamitov
- National Research Center for Cardiac Surgery, Astana 010000, Kazakhstan
| | | | - Natasha S. Barteneva
- School of Sciences and Humanities, Nazarbayev University, Astana 010000, Kazakhstan
| | | |
Collapse
|
9
|
Significance of Catecholamine Biosynthetic/Metabolic Pathway in SARS-CoV-2 Infection and COVID-19 Severity. Cells 2022; 12:cells12010012. [PMID: 36611805 PMCID: PMC9818320 DOI: 10.3390/cells12010012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
The SARS-CoV-2 infection was previously associated with the expression of the dopamine biosynthetic enzyme L-Dopa decarboxylase (DDC). Specifically, a negative correlation was detected between DDC mRNA and SARS-CoV-2 RNA levels in in vitro infected epithelial cells and the nasopharyngeal tissue of COVID-19 patients with mild/no symptoms. However, DDC, among other genes related to both DDC expression and SARS-CoV-2-infection (ACE2, dACE2, EPO), was upregulated in these patients, possibly attributed to an orchestrated host antiviral response. Herein, by comparing DDC expression in the nasopharyngeal swab samples of severe/critical to mild COVID-19 cases, we showed a 20 mean-fold reduction, highlighting the importance of the expression of this gene as a potential marker of COVID-19 severity. Moreover, we identified an association of SARS-CoV-2 infection with the expression of key catecholamine biosynthesis/metabolism-related genes, in whole blood samples from hospitalized patients and in cultured cells. Specifically, viral infection downregulated the biosynthetic part of the dopamine pathway (reduction in DDC expression up to 7.5 mean-fold), while enhanced the catabolizing part (increase in monoamine oxidases A and B expression up to 15 and 10 mean-fold, respectively) in vivo, irrespectively of the presence of comorbidities. In accordance, dopamine levels in the sera of severe cases were reduced (up to 3.8 mean-fold). Additionally, a moderate positive correlation between DDC and MAOA mRNA levels (r = 0.527, p < 00001) in the blood was identified upon SARS-CoV-2-infection. These observations were consistent to the gene expression data from SARS-CoV-2-infected Vero E6 and A549 epithelial cells. Furthermore, L-Dopa or dopamine treatment of infected cells attenuated the virus-derived cytopathic effect by 55% and 59%, respectively. The SARS-CoV-2 mediated suppression of dopamine biosynthesis in cell culture was, at least in part, attributed to hypoxia-like conditions triggered by viral infection. These findings suggest that L-Dopa/dopamine intake may have a preventive or therapeutic value for COVID-19 patients.
Collapse
|
10
|
Heissig B, Salama Y, Iakoubov R, Vehreschild JJ, Rios R, Nogueira T, Vehreschild MJGT, Stecher M, Mori H, Lanznaster J, Adachi E, Jakob C, Tabe Y, Ruethrich M, Borgmann S, Naito T, Wille K, Valenti S, Hower M, Hattori N, Rieg S, Nagaoka T, Jensen BE, Yotsuyanagi H, Hertenstein B, Ogawa H, Wyen C, Kominami E, Roemmele C, Takahashi S, Rupp J, Takahashi K, Hanses F, Hattori K. COVID-19 Severity and Thrombo-Inflammatory Response Linked to Ethnicity. Biomedicines 2022; 10:biomedicines10102549. [PMID: 36289811 PMCID: PMC9599040 DOI: 10.3390/biomedicines10102549] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/03/2022] [Accepted: 10/08/2022] [Indexed: 01/08/2023] Open
Abstract
Although there is strong evidence that SARS-CoV-2 infection is associated with adverse outcomes in certain ethnic groups, the association of disease severity and risk factors such as comorbidities and biomarkers with racial disparities remains undefined. This retrospective study between March 2020 and February 2021 explores COVID-19 risk factors as predictors for patients’ disease progression through country comparison. Disease severity predictors in Germany and Japan were cardiovascular-associated comorbidities, dementia, and age. We adjusted age, sex, body mass index, and history of cardiovascular disease comorbidity in the country cohorts using a propensity score matching (PSM) technique to reduce the influence of differences in sample size and the surprisingly young, lean Japanese cohort. Analysis of the 170 PSM pairs confirmed that 65.29% of German and 85.29% of Japanese patients were in the uncomplicated phase. More German than Japanese patients were admitted in the complicated and critical phase. Ethnic differences were identified in patients without cardiovascular comorbidities. Japanese patients in the uncomplicated phase presented a suppressed inflammatory response and coagulopathy with hypocoagulation. In contrast, German patients exhibited a hyperactive inflammatory response and coagulopathy with hypercoagulation. These differences were less pronounced in patients in the complicated phase or with cardiovascular diseases. Coagulation/fibrinolysis-associated biomarkers rather than inflammatory-related biomarkers predicted disease severity in patients with cardiovascular comorbidities: platelet counts were associated with severe illness in German patients. In contrast, high D-dimer and fibrinogen levels predicted disease severity in Japanese patients. Our comparative study indicates that ethnicity influences COVID-19-associated biomarker expression linked to the inflammatory and coagulation (thrombo-inflammatory) response. Future studies will be necessary to determine whether these differences contributed to the less severe disease progression observed in Japanese COVID-19 patients compared with those in Germany.
Collapse
Affiliation(s)
- Beate Heissig
- School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Yousef Salama
- An-Najah Center for Cancer and Stem Cell Research, Faculty of Medicine and Health Sciences, An-Najah National University, P.O. Box 7, Nablus 99900800, Palestine
| | - Roman Iakoubov
- Department of Internal Medicine II, University Hospital Rechts der Isar, School of Medicine, Technical University, 81675 Munich, Germany
| | | | - Ricardo Rios
- Institute of Computing, Federal University of Bahia, Salvador 40110060, Brazil
| | - Tatiane Nogueira
- Institute of Computing, Federal University of Bahia, Salvador 40110060, Brazil
| | - Maria J. G. T. Vehreschild
- Department of Internal Medicine, Infectious Diseases, University Hospital Frankfurt, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Melanie Stecher
- Department I for Internal Medicine, University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany
- German Center for Infection Research (DZIF), Partner-Site Bonn-Cologne, 50937 Cologne, Germany
| | - Hirotake Mori
- School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | | | - Eisuke Adachi
- IMSUT Hospital of The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Carolin Jakob
- Department I for Internal Medicine, University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany
- German Center for Infection Research (DZIF), Partner-Site Bonn-Cologne, 50937 Cologne, Germany
| | - Yoko Tabe
- School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | | | | | - Toshio Naito
- School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Kai Wille
- Johannes Wesling Klinikum Minden, Ruhr-Universitaet, 44801 Bochum, Germany
| | - Simon Valenti
- School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Martin Hower
- Klinikum Dortmund gGmbH, Hospital of University Witten/Herdecke, 44137 Dortmund, Germany
| | - Nobutaka Hattori
- School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | | | - Tetsutaro Nagaoka
- School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | | | - Hiroshi Yotsuyanagi
- IMSUT Hospital of The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | | | - Hideoki Ogawa
- School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | | | - Eiki Kominami
- School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Christoph Roemmele
- Internal Medicine III—Gastroenterology and Infectious Diseases, University Hospital of Augsburg, 86156 Augsburg, Germany
| | - Satoshi Takahashi
- IMSUT Hospital of The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University Hospital Schleswig-Holstein/Campus Luebeck, 23538 Luebeck, Germany
| | - Kazuhisa Takahashi
- School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Frank Hanses
- Emergency Department and Department for Infectious Diseases and Infection Control, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Koichi Hattori
- School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
- Correspondence:
| | | |
Collapse
|
11
|
Fibrinogen-to-Albumin Ratio and Blood Urea Nitrogen-to-Albumin Ratio in COVID-19 Patients: A Systematic Review and Meta-Analysis. Trop Med Infect Dis 2022; 7:tropicalmed7080150. [PMID: 36006242 PMCID: PMC9414552 DOI: 10.3390/tropicalmed7080150] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 01/08/2023] Open
Abstract
Fibrinogen-to-albumin ratio (FAR) and blood urea nitrogen-to-albumin ratio (BAR) are inflammatory biomarkers that have been associated with clinical outcomes of multiple diseases. The objective of this study is to evaluate the association of these biomarkers with the severity and mortality of COVID-19 patients. A systematic search was performed in five databases. Observational studies that reported the association between FAR and BAR values with the severity and mortality of COVID-19 patients were included. Random-effects models were used for meta-analyses, and effects were expressed as Odds Ratio (OR) and their 95% confidence intervals (CI). Publication bias was assessed using the Begg test, while the quality assessment was assessed using the Newcastle Ottawa Scale. A total of 21 studies (n = 7949) were included. High FAR values were associated with a higher risk of severity (OR: 2.41; 95% CI 1.41−4.12; p < 0.001) and mortality (OR: 2.05; 95% CI 1.66−2.54; p < 0.001). High BAR values were associated with higher risk of mortality (OR: 4.63; 95% CI 2.11−10.15; p < 0.001). However, no statistically significant association was found between BAR values and the risk of severity (OR: 1.16; 95% CI 0.83−1.63; p = 0.38). High FAR and BAR values were associated with poor clinical outcomes.
Collapse
|
12
|
Konstantinidou AE, Angelidou S, Havaki S, Paparizou K, Spanakis N, Chatzakis C, Sotiriadis A, Theodora M, Donoudis C, Daponte A, Skaltsounis P, Gorgoulis VG, Papaevangelou V, Kalantaridou S, Tsakris A. Stillbirth due to SARS-CoV-2 placentitis without evidence of intrauterine transmission to fetus: association with maternal risk factors. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2022; 59:813-822. [PMID: 35353936 PMCID: PMC9111139 DOI: 10.1002/uog.24906] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 05/23/2023]
Abstract
OBJECTIVES To describe the placental pathology, fetal autopsy findings and clinical characteristics of pregnancies that resulted in stillbirth owing to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) placentitis, and to identify potential risk factors. METHODS This was a prospective multicenter study of non-vaccinated pregnant women affected by coronavirus disease 2019 (COVID-19) in Greece from April 2020 to August 2021. A total of 165 placentas were examined histologically and six cases of stillbirth associated with SARS-CoV-2 placentitis were retrieved. Complete fetal autopsy was performed in three of these cases. Gross, histopathological, immunohistochemical, molecular and electron microscopy examinations were carried out in the stillbirth placentas and fetal organs. The histological findings of cases with SARS-CoV-2 placentitis were compared with those in 159 cases with maternal COVID-19 which resulted in a live birth. Regression analysis was used to identify predisposing risk factors for SARS-CoV-2 placentitis. RESULTS The placentas of all six stillborn cases showed severe and extensive histological changes typical of SARS-CoV-2 placentitis, characterized by a combination of marked intervillositis with a mixed inflammatory infiltrate and massive perivillous fibrinoid deposition with trophoblast damage, associated with intensely positive immunostaining for SARS-CoV-2 spike protein, the presence of virions on electron microscopy and positive reverse-transcription polymerase chain reaction test of placental tissues. The histological lesions obliterated over 75% of the maternal intervillous space, accounting for intrauterine fetal death. Similar histological lesions affecting less than 25% of the placenta were observed in seven liveborn neonates, while the remaining 152 placentas of COVID-19-affected pregnancies with a live birth did not show these findings. Complete fetal autopsy showed evidence of an asphyctic mode of death without evidence of viral transmission to the fetus. The mothers had mild clinical symptoms or were asymptomatic, and the interval between maternal COVID-19 diagnosis and fetal death ranged from 3 to 15 days. Statistically significant predisposing factors for SARS-CoV-2 placentitis included thrombophilia and prenatally diagnosed fetal growth restriction (FGR). Multiple sclerosis was seen in one case. CONCLUSIONS SARS-CoV-2 placentitis occurred uncommonly in COVID-19-affected pregnancies of non-vaccinated mothers and, when extensive, caused fetal demise, with no evidence of transplacental fetal infection. Thrombophilia and prenatally detected FGR emerged as independent predisposing factors for the potentially lethal SARS-CoV-2 placentitis. © 2022 International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- A. E. Konstantinidou
- First Department of Pathology, Perinatal Pathology Unit, School of MedicineNational and Kapodistrian University of AthensAthensGreece
- Laboratory of Pathology, Aretaieion University Hospital, School of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - S. Angelidou
- Department of PathologyHippokration HospitalThessalonikiGreece
| | - S. Havaki
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - K. Paparizou
- First Department of Pathology, Perinatal Pathology Unit, School of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - N. Spanakis
- Department of Microbiology, School of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - C. Chatzakis
- Second Department of Obstetrics and Gynecology, Medical SchoolAristotle University of ThessalonikiThessalonikiGreece
| | - A. Sotiriadis
- Second Department of Obstetrics and Gynecology, Medical SchoolAristotle University of ThessalonikiThessalonikiGreece
| | - M. Theodora
- First Department of Obstetrics and GynecologyAlexandra Hospital, School of Medicine, National and Kapodistrian University of AthensAthensGreece
| | - C. Donoudis
- Department of Obstetrics and Gynecology, Faculty of Medicine, School of Health SciencesUniversity of ThessalyLarissaGreece
| | - A. Daponte
- Department of Obstetrics and Gynecology, Faculty of Medicine, School of Health SciencesUniversity of ThessalyLarissaGreece
| | - P. Skaltsounis
- First Department of Pathology, Perinatal Pathology Unit, School of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - V. G. Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of MedicineNational and Kapodistrian University of AthensAthensGreece
- Faculty of Biology, Medicine and Health, Manchester Cancer Research Centre, Manchester Academic Health Sciences CentreThe University of ManchesterManchesterUK
- Center for New Biotechnologies and Precision Medicine, Medical SchoolNational and Kapodistrian University of AthensAthensGreece
- Biomedical Research Foundation of the Academy of AthensAthensGreece
- Ninewells Hospital and Medical SchoolUniversity of DundeeDundeeUK
| | - V. Papaevangelou
- Third Department of Pediatrics, National and Kapodistrian University of Athens, School of MedicineUniversity General Hospital AttikonAthensGreece
| | - S. Kalantaridou
- Department of Obstetrics and Gynecology, National and Kapodistrian University of AthensSchool of Medicine, University General Hospital AttikonAthensGreece
| | - A. Tsakris
- Department of Microbiology, School of MedicineNational and Kapodistrian University of AthensAthensGreece
| |
Collapse
|
13
|
Jamil SW, Ilyas M, Ahmad N, Bakri S, Hardjianti T, Parewangi ML, Seweng A. Coagulation Profile and Outcomes of COVID-19 Patients at Wahidin Sudirohusodo Hospital, Makassar, Indonesia. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.9109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background: Coronavirus disease 2019 (COVID-19) is a viral pneumonia infection that spreads rapidly globally (with a number of cases > 15,000,000 worldwide and mortality of ±4%) until it is designated a pandemic by the World Health Organization (WHO). One of the complications of COVID-19 is the incidence of coagulopathy and thromboembolism. The coronavirus, SARS-CoV-2, activates inflammatory and thrombotic processes. Coagulopathy and abnormal coagulation parameters are indicated among the most significant biomarkers of poor prognosis in COVID-19 patients. COVID-19-associated coagulopathy is characterized by a decreased platelet count and the presence of a cytokine storm indicating an extreme hypercoagulable state. This study aims to determine the coagulation profile of moderate-severe patients and outcomes in COVID-19 patients
Methods: The study was conducted in a hospital in Makassar: Infection Center RS. Wahidin Sudirohusodo. Medical Record Data for all inpatients who have been diagnosed with COVID-19 through the RT-PCR test taken from January 2021-August 2021.Statistical tests in the form of the Kolmogorov-Smirnov test to assess the Normality of the Data, Chi-Square test, and the calculation of the out ratio (OR) Mann-Whitney test, Independent T-Test. Multivariate analysis was carried out using a Multiple Logistic Regression-Backward Wald Method. The results of the statistical test were significant if the p-value <0.05.
Results:
The research subjects were 231 patients with confirmed COVID-19. The mean PT, D-Dimer, and Fibrinogen were higher in severe COVID-19 than moderate COVID-19 and had significant results. While PLT did not have significant results against moderate-severe COVID-19. The relationship between groups of coagulation marker variables was found to have a significant relationship with moderate to severe COVID-19. 4. All coagulation markers were significantly related to the outcome (p<0.05). The mean value of each variable was found to be greater in patients with outcomes who died
Conclusion:
There was an increase in all coagulation markers in moderate to severe COVID-19 except for PLT which was not significant. All coagulation markers are significantly related to outcome
Collapse
|