1
|
Adhikari A, Maddumage J, Eriksson EM, Annesley SJ, Lawson VA, Bryant VL, Gras S. Beyond acute infection: mechanisms underlying post-acute sequelae of COVID-19 (PASC). Med J Aust 2024; 221 Suppl 9:S40-S48. [PMID: 39489518 DOI: 10.5694/mja2.52456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 08/10/2024] [Indexed: 11/05/2024]
Abstract
Immune dysregulation is a key aspect of post-acute sequelae of coronavirus disease 2019 (PASC), also known as long COVID, with sustained activation of immune cells, T cell exhaustion, skewed B cell profiles, and disrupted immune communication thereby resulting in autoimmune-related complications. The gut is emerging as a critical link between microbiota, metabolism and overall dysfunction, potentially sharing similarities with other chronic fatigue conditions and PASC. Immunothrombosis and neurological signalling dysfunction emphasise the complex interplay between the immune system, blood clotting, and the central nervous system in the context of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Clear research gaps in the design of PASC studies, especially in the context of longitudinal research, stand out as significant areas of concern.
Collapse
Affiliation(s)
- Anurag Adhikari
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC
- Kathmandu Research Institute for Biological Sciences, Lalitpur, Nepal
| | - Janesha Maddumage
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC
| | - Emily M Eriksson
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC
| | | | - Victoria A Lawson
- Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC
| | - Vanessa L Bryant
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC
- Royal Melbourne Hospital, Melbourne, VIC
- University of Melbourne, Melbourne, VIC
| | - Stephanie Gras
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC
- Monash University, Melbourne, VIC
| |
Collapse
|
2
|
Al-Sammarraie MR, Al-Sammarraie MR, Azaiez F, Al-Rubae ZMM, Litaiem H, Taay YM. mRNA vaccination reduces the thrombotic possibility in COVID-19: Inflammation risk estimates. Int Immunopharmacol 2024; 140:112776. [PMID: 39079343 DOI: 10.1016/j.intimp.2024.112776] [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: 04/04/2024] [Revised: 06/08/2024] [Accepted: 07/23/2024] [Indexed: 09/01/2024]
Abstract
Thrombosis is a common clinical feature associated with morbidity and mortality in coronavirus disease-2019 (COVID-19) patients. Cytokine storm in COVID-19 increases patients' systemic inflammation, which can cause multiple health consequences. In this work, we aimed to indicate the effect of Pfizer-BioNTech vaccination on the modulation of monocyte chemoattractant protein-3 (MCP-3), matrix metalloproteinase 1 (MMP-1), and tumor necrosis factor-alpha (TNF-α) levels, and other systemic inflammatory biomarkers that associates with COVID-19 severity in patients who suffers from thrombosis consequences. For this purpose, ninety people were collected from Ibn Al-Nafees Hospital and divided into three groups each of which contained 30 people, 15 of them were venous thromboembolism (VTE) positive and the other were VTE negative. The three groups were non-vaccinated COVID-19, vaccinated COVID-19, and control. The levels of MCP-3 and TNF-α were significantly (p < 0.05) increased in vaccinated and non-vaccinated COVID-19 patients regardless of their thrombosis condition, while MMP-1 level was non-significantly (p > 0.05) higher in vaccinated patients compared to control. MCP-3 and TNF-α were correlated positively with D-dimer (r = 0.544 and r = 0.513, respectively) in non-vaccinated patients, while MMP-1 and TNF-α were correlated positively with D-dimer (r = 0.624 and r = 0.575, respectively) in vaccinated patients. The odds ratio of MCP-3 (2.252), MMP-1 (1.062), and TNF-α (1.360) were reduced in vaccinated patients (2.093, 1.022, and 1.301 for MCP-3, MMP-1, and TNF-α respectively). Thus, MCP-3 plays a vital role in COVID-19 pathophysiology, and vaccination can reduce the risk of developing VTE in COVID-19 patients, and improve the inflammatory condition of patients.
Collapse
Affiliation(s)
- Marwah Raad Al-Sammarraie
- Department of Chemistry, College of Science for Women, University of Baghdad, Baghdad, Iraq; Laboratory of Inorganic Chemistry, Faculty of Sciences, University of Sfax, Tunisia.
| | | | - Fatma Azaiez
- Laboratory Clinical Virology Pasteur Institute and Department of Toxicology, Faculty of Pharmacy Monastir, Tunisia
| | - Zeinab M M Al-Rubae
- Department of Chemistry, College of Education for Pure Science, University of Baghdad, Baghdad, Iraq
| | - Hejer Litaiem
- Laboratory of Inorganic Chemistry, Faculty of Sciences, University of Sfax, Tunisia
| | - Yasser M Taay
- Department of Chemistry, College of Science, University of Baghdad, Baghdad, Iraq.
| |
Collapse
|
3
|
Zhang H, Feng Q, Ma Q, Li L, Xing Y. Serum lncRNA RAMP2-AS1 Served as a Biomarker of Deep Vein Thrombosis Occurrence and Development in Elderly. Indian J Hematol Blood Transfus 2024; 40:660-667. [PMID: 39469176 PMCID: PMC11512952 DOI: 10.1007/s12288-024-01782-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2024] [Indexed: 10/30/2024] Open
Abstract
The incidence of deep vein thrombosis (DVT) is increasing with aging, which needs a screening and monitoring biomarker. This study focused on the significance of aging- and angiogenesis-related lncRNA RAMP2-AS1 (RAMP2-AS1) aiming to identify a promising biomarker for the incidence of DVT. Serum samples were collected from 63 healthy individuals and 98 patients with DVT. The serum RAMP2-AS1 level was analyzed by PCR and its significance in DVT detection and development prediction was evaluated by ROC and multivariate Cox regression analysis. The regulatory effect of RAMP2-AS1 on endothelial progenitor cells (EPCs) was evaluated by CCK8 and transwell assays. RAMP2-AS1 was significantly downregulated in patients with DVT, which could discriminate patients with DVT from healthy individuals with relatively high sensitivity and specificity. The downregulation of RAMP2-AS1 could predict poor outcomes and was associated with activities of daily living (ADL), neutrophil to lymphocyte ratio (NLR), platelet to lymphocyte ratio (PLR), lymphocyte to monocyte ratio (LMR), and monocyte to high-density lipoprotein cholesterol ratio (MHR) of patients with DVT. RAMP2-AS1 was identified as an independent prognostic factor of DVT by Cox regression analysis. In EPCs, overexpressing RAMP2-AS1 significantly suppressed cell proliferation, migration, and invasion. Downregulated serum RAMP2-AS1 could predict the incidence and progression of DVT. RAMP2-AS1 inhibited EPCs growth and motility, which provides a target for thrombolytic therapy. RAMP2-AS1 level could be included in the risk assessment model of DVT.
Collapse
Affiliation(s)
- Hailong Zhang
- Department of Vascular Surgery, Beidahuang Industry Group General Hospital, Harbin, 150088 China
| | - Qichen Feng
- Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing, 100191 China
| | - Qingfeng Ma
- Laboratory Department, The People’s Hospital of Feicheng, Taian, 271600 China
| | - Li Li
- Laboratory Department, The People’s Hospital of Feicheng, Taian, 271600 China
| | - Youzhong Xing
- Department of Blood Transfusion, Jinan Central Hospital, No.105 Jiefang Road, Jinan, 250013 China
| |
Collapse
|
4
|
Palazzuoli A, Mingiano C, Manetti N, Leolini C, Fossi A. Pacing lead thrombus in patient with recent COVID-19 infection and subsequent vaccination: a case report. Eur Heart J Case Rep 2024; 8:ytae447. [PMID: 39258023 PMCID: PMC11384885 DOI: 10.1093/ehjcr/ytae447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/19/2024] [Accepted: 08/12/2024] [Indexed: 09/12/2024]
Abstract
Background The association between acute coronavirus disease-19 (COVID-19) infection and a hypercoagulable state has been exhaustively described throughout the pandemic. The presence of external devices, such as intracardiac leads, could predispose to higher thrombotic risk in this setting. We present a clinical case of intracardiac thrombosis on right ventricle device that occurred after COVID-19 infection and subsequent vaccination. Case summary A 56-year-old man, suffering from usual interstitial pneumonia-pattern fibrosis, was admitted to our hospital because of worsening of his clinical status. About 10 days earlier, he had got vaccinated for COVID-19. Three months earlier, the patient had been reported to have severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) infection. A chest computed tomography scan showed thrombus partially occluding the left pulmonary artery. A transthoracic echocardiography and later a transoesophageal echocardiogram showed a mass adhered to the lead in the right ventricle, compatible with thrombosis, confirmed on a cardiac computed tomography scan. Blood tests showed no major changes except for a slight increase in D-dimer and fibrinogen. Therefore, the subject was treated with anticoagulants. Discussion COVID-19 infection results in a hypercoagulable state with risk of developing thrombus diffusely, including intracardiac thrombosis. The presence of external devices, such as the intracardiac leads, may increase thrombotic risk since the presence of an external device in the bloodstream could trigger coagulation cascade. This case report highlights the need for special care in this patient setting, using specific imaging techniques for early and rapid diagnosis to optimize therapy.
Collapse
Affiliation(s)
- Alberto Palazzuoli
- Cardiovascular Diseases Unit, Cardio Thoracic and Vascular Department, Le Scotte Hospital, University of Siena, 53100 Siena, Italy
| | - Christian Mingiano
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
| | - Niccolò Manetti
- Cardiovascular Diseases Unit, Cardio Thoracic and Vascular Department, Le Scotte Hospital, University of Siena, 53100 Siena, Italy
| | - Chiara Leolini
- Cardiovascular Diseases Unit, Cardio Thoracic and Vascular Department, Le Scotte Hospital, University of Siena, 53100 Siena, Italy
| | - Antonella Fossi
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena, Siena, Italy
| |
Collapse
|
5
|
Silvestri C, Stasi C, Profili F, Bartolacci S, Sessa E, Tacconi D, Villari L, Carrozzi L, Dotta F, Bargagli E, Donnini S, Masotti L, Rasero L, Lavorini F, Pistelli F, Chimera D, Sorano A, Pacifici M, Milli C, Voller F, Group SPRINTS. Retrospective Study on the Features and Outcomes of a Tuscany COVID-19 Hospitalized Patients Cohort: Preliminary Results. J Clin Med 2024; 13:4626. [PMID: 39200770 PMCID: PMC11354555 DOI: 10.3390/jcm13164626] [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: 06/27/2024] [Revised: 07/26/2024] [Accepted: 08/05/2024] [Indexed: 09/02/2024] Open
Abstract
Background: A few months after the COVID-19 pandemic onset, knowledge of SARS-CoV-2 infection and outcomes and treatments blew up. This paper aimed to evaluate the features of a Tuscany COVID-19 hospitalized cohort and to identify risk factors for COVID-19 severity. Methods: This retrospective observational COVID-19 cohort study (1 March 2020-1 March 2021) was conducted on patients ≥ 18 years old, admitted to Tuscany Hospital, and subjected to follow-up within 12 months after discharge. Patients were enrolled at Pisana, Senese and Careggi University Hospitals, and South East, North West, and Center Local Hospitals. Results: 2888 patients (M = 58.5%, mean age = 66.2 years) were enrolled, of whom 14.3% (N = 413) were admitted to an intensive care unit. Smokers were 25%, and overweight and obese 65%. The most used drugs were corticosteroids, antacids, antibiotics, and antithrombotics, all antiviral drugs, with slight differences between 2020 and 2021. A strong association was found between outcomes of evolution towards critical COVID-19 (non-invasive mechanical ventilation (NIV) and/or admission to intensive care) and smoking (RR = 4.91), ex-smoking (RR = 3.48), overweight (RR = 1.30), obese subjects (RR = 1.62), comorbidities (aRR = 1.38). The alteration of liver enzymes (aspartate aminotransferase, alanine aminotransferase, or gamma-glutamyl transpeptidase) was associated with NIV (aOR = 2.28). Conclusions: Our cohort, characterized by patients with a mean age of 66.2 years, showed 65% of patients were overweight and obese. Smoking/ex-smoking, overweight/obesity, and other comorbidities were associated with COVID-19 adverse outcomes. The findings also demonstrated that alterations in liver enzymes were associated with worse outcomes.
Collapse
Affiliation(s)
- Caterina Silvestri
- Epidemiology Unit, Regional Health Agency of Tuscany, 50141 Florence, Italy
| | - Cristina Stasi
- Epidemiology Unit, Regional Health Agency of Tuscany, 50141 Florence, Italy
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
| | - Francesco Profili
- Epidemiology Unit, Regional Health Agency of Tuscany, 50141 Florence, Italy
| | - Simone Bartolacci
- Epidemiology Unit, Regional Health Agency of Tuscany, 50141 Florence, Italy
| | - Emiliano Sessa
- Epidemiology Unit, Regional Health Agency of Tuscany, 50141 Florence, Italy
| | - Danilo Tacconi
- Infectious Diseases Unit, PO San Donato, 52100 Arezzo, Italy
| | - Liliana Villari
- Division of Pneumology, AUSL Toscana Nord-Ovest, Apuane Hospital, 54100 Massa, Italy;
| | - Laura Carrozzi
- Pneumology Unit, Pisa University Hospital, 56124 Pisa, Italy
| | - Francesco Dotta
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
- Diabetes and Metabolic Diseases Unit, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy
| | - Elena Bargagli
- Respiratory Diseases Unit, Department Medical Sciences, Surgery and Neurological Sciences, Siena University, 53100 Siena, Italy
| | - Sandra Donnini
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Luca Masotti
- Internal Medicine II and Stroke Unit, San Giuseppe Hospital, 20123 Empoli, Italy
| | - Laura Rasero
- Department of Health Sciences, Clinical Innovations and Research Unit, Careggi University Hospital, University of Florence, 50121 Florence, Italy
| | - Federico Lavorini
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (F.L.)
| | | | - Davide Chimera
- Pneumology Unit, Pisa University Hospital, 56124 Pisa, Italy
| | - Alessandra Sorano
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (F.L.)
| | - Martina Pacifici
- Epidemiology Unit, Regional Health Agency of Tuscany, 50141 Florence, Italy
| | - Caterina Milli
- Epidemiology Unit, Regional Health Agency of Tuscany, 50141 Florence, Italy
| | - Fabio Voller
- Epidemiology Unit, Regional Health Agency of Tuscany, 50141 Florence, Italy
| | | |
Collapse
|
6
|
Silva-Santos Y, Pagni RL, Gamon THM, de Azevedo MSP, Bielavsky M, Darido MLG, de Oliveira DBL, de Souza EE, Wrenger C, Durigon EL, Luvizotto MCR, Ackerman HC, Marinho CRF, Epiphanio S, Carvalho LJM. Lisinopril increases lung ACE2 levels and SARS-CoV-2 viral load and decreases inflammation but not disease severity in experimental COVID-19. Front Pharmacol 2024; 15:1414406. [PMID: 39070798 PMCID: PMC11282493 DOI: 10.3389/fphar.2024.1414406] [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: 04/08/2024] [Accepted: 06/04/2024] [Indexed: 07/30/2024] Open
Abstract
COVID-19 causes more severe and frequently fatal disease in patients with pre-existing comorbidities such as hypertension and heart disease. SARS-CoV-2 virus enters host cells through the angiotensin-converting enzyme 2 (ACE2), which is fundamental in maintaining arterial pressure through the renin-angiotensin system (RAS). Hypertensive patients commonly use medications such as angiotensin-converting enzyme inhibitors (ACEi), which can modulate the expression of ACE2 and, therefore, potentially impact the susceptibility and severity of SARS-CoV-2 infection. Here we assessed whether treatment of ACE2-humanized (K18-hACE2) mice with the ACEi Lisinopril affects lung ACE2 levels and the outcome of experimental COVID-19. K18-hACE2 mice were treated for 21 days with Lisinopril 10 mg/kg and were then infected with 105 PFU of SARS-CoV-2 (Wuhan strain). Body weight, clinical score, respiratory function, survival, lung ACE2 levels, viral load, lung histology, and cytokine (IL-6, IL-33, and TNF-α) levels were assessed. Mice treated with Lisinopril for 21 days showed increased levels of ACE2 in the lungs. Infection with SARS-CoV-2 led to massive decrease in lung ACE2 levels at 3 days post-infection (dpi) in treated and untreated animals, but Lisinopril-treated mice showed a fast recovery (5dpi) of ACE2 levels. Higher ACE2 levels in Lisinopril-treated mice led to remarkably higher lung viral loads at 3 and 6/7dpi. Lisinopril-treated mice showed decreased levels of the pro-inflammatory cytokines IL-6 and TNF-α in the serum and lungs at 6/7dpi. Marginal improvements in body weight, clinical score and survival were observed in Lisinopril-treated mice. No differences between treated and untreated infected mice were observed in respiratory function and lung histology. Lisinopril treatment showed both deleterious (higher viral loads) and beneficial (anti-inflammatory and probably anti-constrictory and anti-coagulant) effects in experimental COVID-19. These effects seem to compensate each other, resulting in marginal beneficial effects in terms of outcome for Lisinopril-treated animals.
Collapse
Affiliation(s)
- Yasmin Silva-Santos
- Laboratory of Malaria Cellular and Molecular Immunopathology, Faculty of Pharmaceutical Sciences, Department of Clinical and Toxicological Analysis, University of São Paulo, São Paulo, Brazil
- Laboratory of Malaria Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Roberta Liberato Pagni
- Immunology Laboratory, Heart Institute, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Thais Helena Martins Gamon
- Laboratory of Clinical and Molecular Virology, Institute of Biomedical Sciences, Department of Microbiology, University of São Paulo, São Paulo, Brazil
| | - Marcela Santiago Pacheco de Azevedo
- Laboratory of Clinical and Molecular Virology, Institute of Biomedical Sciences, Department of Microbiology, University of São Paulo, São Paulo, Brazil
- Laboratory of Experimental Immunoparasitology, Institute of Biomedical Sciences, Department of Parasitology, University of São Paulo, São Paulo, Brazil
| | - Mônica Bielavsky
- Laboratory of Malaria Cellular and Molecular Immunopathology, Faculty of Pharmaceutical Sciences, Department of Clinical and Toxicological Analysis, University of São Paulo, São Paulo, Brazil
| | - Maria Laura Goussain Darido
- Laboratory of Clinical and Molecular Virology, Institute of Biomedical Sciences, Department of Microbiology, University of São Paulo, São Paulo, Brazil
| | - Danielle Bruna Leal de Oliveira
- Laboratory of Clinical and Molecular Virology, Institute of Biomedical Sciences, Department of Microbiology, University of São Paulo, São Paulo, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Edmarcia Elisa de Souza
- Unit for Drug Discovery, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Carsten Wrenger
- Unit for Drug Discovery, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Edson Luiz Durigon
- Laboratory of Clinical and Molecular Virology, Institute of Biomedical Sciences, Department of Microbiology, University of São Paulo, São Paulo, Brazil
| | | | - Hans Christian Ackerman
- Physiology Unit, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MD, United States
| | - Claudio Romero Farias Marinho
- Laboratory of Experimental Immunoparasitology, Institute of Biomedical Sciences, Department of Parasitology, University of São Paulo, São Paulo, Brazil
| | - Sabrina Epiphanio
- Laboratory of Malaria Cellular and Molecular Immunopathology, Faculty of Pharmaceutical Sciences, Department of Clinical and Toxicological Analysis, University of São Paulo, São Paulo, Brazil
| | | |
Collapse
|
7
|
Zhu J, Bouzid R, Travert B, Géri G, Cohen Y, Picod A, Heming N, Rottman M, Joly-Laffargue B, Veyradier A, Capron C, Coppo P. Combined coagulation and inflammation markers as predictors of venous thrombo-embolism and death in COVID-19. Front Med (Lausanne) 2024; 11:1399335. [PMID: 38915768 PMCID: PMC11194426 DOI: 10.3389/fmed.2024.1399335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 05/03/2024] [Indexed: 06/26/2024] Open
Abstract
Background The COVID-19 pandemic related to SARS-CoV-2 virus was responsible for global pandemic. The severe form of the disease was linked to excessive activation of immune pathways together with a systemic cytokine storm response and thrombotic venous or arterial complications. Factors predicting severe outcomes including venous and/or pulmonary thrombosis (VT) and death were identified, but the prognostic role of their combination was not addressed extensively. Objectives We investigated the role of prognostic factors from the coagulation or inflammatory pathways to better understand the outcome of the disease. Methods For this, we prospectively studied 167 SARS-CoV-2-positive patients from admission in intensive care units (ICU) or emergency departments from four academic hospitals over a 14-month period. Besides standard biology, we assessed serum concentrations of inflammatory markers, coagulation factors and peripheral blood cells immunophenotyping. Results Thirty-nine patients (23.3%) developed VT and 30 patients (18%) died. By univariate analysis, C-reactive protein (CRP) level > 150 mg/L, interleukin-6 (IL-6) ≥ 20 pg/mL, D-dimers > 1,500 μg/L, ADAMTS13 activity ≤ 50%, Von. Conclusion A combination of coagulation and inflammatory markers can refine the prognostication of severe outcome in COVID-19, and could be useful for the initial evaluation of other types of viral infection.
Collapse
Affiliation(s)
- Jaja Zhu
- Service d’Hématologie-Immunologie-Transfusion, AP-HP Paris Saclay, CHU Ambroise Paré, Université de Versailles Saint Quentin-Université Paris Saclay, Montigny-le-Bretonneux, France
- Laboratoire Cellules Souches et Applications Thérapeutiques, UMR INSERM 1184, Commissariat à l’Energie Atomique et aux Energies Alternatives, Fontenay-aux-Roses, France
| | - Raïda Bouzid
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), AP-HP, Paris, France
| | - Benoît Travert
- Service de Médecine Interne, Hôpital Ambroise-Paré, AP-HP, Boulogne-Billancourt, France
| | - Guillaume Géri
- Service de Médecine Intensive et Réanimation, Hôpital Ambroise-Paré, AP-HP, Boulogne-Billancourt, France
| | - Yves Cohen
- Service de Médecine Intensive et Réanimation, Hôpital Avicenne, AP-HP, Paris, France
| | - Adrien Picod
- Service de Médecine Intensive et Réanimation, Hôpital Avicenne, AP-HP, Paris, France
| | - Nicholas Heming
- Department of Intensive Care, Raymond Poincaré Hospital, APHP University Versailles Saint Quentin-University Paris Saclay, Garches, France
- Institut Hospitalo Universitaire PROMETHEUS, Garches, France
- Innovative Biomarkers Plateform, Laboratory of Infection & Inflammation-U1173, School of Medicine, INSERM, University Versailles Saint Quentin-University Paris Saclay, Garches, France
- FHU SEPSIS, Garches, France
| | - Martin Rottman
- Innovative Biomarkers Plateform, Laboratory of Infection & Inflammation-U1173, School of Medicine, INSERM, University Versailles Saint Quentin-University Paris Saclay, Garches, France; FHU SEPSIS, Garches, France
- General Intensive Care Unit, Raymond Poincaré Hospital (AP-HP), FHU SEPSIS, Laboratory of Infection and Inflammation-U1173, School of Medicine Simone Veil, Université Versailles Saint Quentin, University Paris Saclay, INSERM, Garches, France
| | - Bérangère Joly-Laffargue
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), AP-HP, Paris, France
- EA3518, Institut de Recherche Saint Louis, Université de Paris, Paris, France
- Service D’hématologie Biologique, Laboratoire ADAMTS13, Hôpital Lariboisière, AP-HP Nord, Université de Paris, Paris, France
| | - Agnès Veyradier
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), AP-HP, Paris, France
- EA3518, Institut de Recherche Saint Louis, Université de Paris, Paris, France
- Service D’hématologie Biologique, Laboratoire ADAMTS13, Hôpital Lariboisière, AP-HP Nord, Université de Paris, Paris, France
| | - Claude Capron
- Service d’Hématologie-Immunologie-Transfusion, AP-HP Paris Saclay, CHU Ambroise Paré, Université de Versailles Saint Quentin-Université Paris Saclay, Montigny-le-Bretonneux, France
- Université Paris-Saclay, Université de Versailles Saint Quentin en Yvelines (UVSQ), Biomarqueurs en cancérologie et onco-hématologie (BECCOH), Boulogne-Billancourt, France
| | - Paul Coppo
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), AP-HP, Paris, France
- Service d’Hématologie, Hôpital Saint-Antoine, AP-HP-Sorbonne Université, Paris, France
- NSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
| |
Collapse
|
8
|
Ni XX, Liu ZY, Zeng YY, Liu ZF. Heatstroke Comorbid with SARS-CoV-2 Infection: A Case Report and Literature Review. Int Med Case Rep J 2024; 17:555-563. [PMID: 38831931 PMCID: PMC11146621 DOI: 10.2147/imcrj.s461078] [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: 01/23/2024] [Accepted: 05/23/2024] [Indexed: 06/05/2024] Open
Abstract
Background Hyperthermia and multiple organ dysfunction syndrome (MODS) are the main characteristics of heatstroke and COVID-19. Differentiating between these illnesses is crucial during a summer COVID-19 pandemic, but cases of heatstroke comorbid with COVID-19 are rarely reported. Case description We report the first case of heatstroke comorbid with Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection in a 52-year-old male. After receiving intravenous antibiotics, organ protection measures, and treatment for coagulation disorders, his fever and coma resolved. However, he developed dyspnea and cerebral hemorrhage after several days. This patient experienced a multi-pathogen pulmonary infection and an intractable coagulopathy that ultimately resulted in MODS and death. Conclusion The combination of heatstroke and SARS-CoV-2 infection exacerbated inflammation, immune abnormalities, and coagulation disorders. The interaction between inflammation and coagulation disturbances contributed to the underlying mechanism in this case, highlighting the importance of early anti-infection, treatment for coagulopathy, immune regulation, and organ protection as crucial interventions.
Collapse
Affiliation(s)
- Xiao-xiao Ni
- Department of Hyperbaric Oxygen Medicine and Rehabilitation, General Hospital of Southern Theater Command of PLA, Guangzhou, Guangdong, People’s Republic of China
| | - Zhe-ying Liu
- Department of Medicine Intensive Care Unit, General Hospital of Southern Theater Command of PLA, Guangzhou, Guangdong, People’s Republic of China
| | - Yan-yan Zeng
- Department of Hyperbaric Oxygen Medicine and Rehabilitation, General Hospital of Southern Theater Command of PLA, Guangzhou, Guangdong, People’s Republic of China
| | - Zhi-feng Liu
- Department of Medicine Intensive Care Unit, General Hospital of Southern Theater Command of PLA, Guangzhou, Guangdong, People’s Republic of China
| |
Collapse
|
9
|
Shen J, Li J, Lei Y, Chen Z, Wu L, Lin C. Frontiers and hotspots evolution in cytokine storm: A bibliometric analysis from 2004 to 2022. Heliyon 2024; 10:e30955. [PMID: 38774317 PMCID: PMC11107250 DOI: 10.1016/j.heliyon.2024.e30955] [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: 07/17/2023] [Revised: 05/08/2024] [Accepted: 05/08/2024] [Indexed: 05/24/2024] Open
Abstract
Background As a fatal disease, cytokine storm has garnered research attention in recent years. Nonetheless, as the body of related studies expands, a thorough and impartial evaluation of the current status of research on cytokine storms remains absent. Consequently, this study aimed to thoroughly explore the research landscape and evolution of cytokine storm utilizing bibliometric and knowledge graph approaches. Methods Research articles and reviews centered on cytokine storms were retrieved from the Web of Science Core Collection database. For bibliometric analysis, tools such as Excel 365, CiteSpace, VOSviewer, and the Bibliometrix R package were utilized. Results This bibliometric analysis encompassed 6647 articles published between 2004 and 2022. The quantity of pertinent articles and citation frequency exhibited a yearly upward trend, with a sharp increase starting in 2020. Network analysis of collaborations reveals that the United States holds a dominant position in this area, boasting the largest publication count and leading institutions. Frontiers in Immunology ranks as the leading journal for the largest publication count in this area. Stephan A. Grupp, a prominent researcher in this area, has authored the largest publication count and has the second-highest citation frequency. Research trends and keyword evaluations show that the connection between cytokine storm and COVID-19, as well as cytokine storm treatment, are hot topics in research. Furthermore, research on cytokine storm and COVID-19 sits at the forefront in this area. Conclusion This study employed bibliometric analysis to create a visual representation of cytokine storm research, revealing current trends and burgeoning topics in this area for the first time. It will provide valuable insights, helping scholars pinpoint critical research areas and potential collaborators.
Collapse
Affiliation(s)
- Junyi Shen
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jiaming Li
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yuqi Lei
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zhengrui Chen
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Lingling Wu
- Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Chunyan Lin
- Department of Teaching and Research Section of Internal Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
10
|
Baye MF, Desta D, Hunegnaw S, Johar A, Bekele I, Angasu K, Bayleyegn NS, Abebe Getahun H, Sisay AL. Assessment of the hematological profiles among COVID-19 patients during the first and second waves in Ethiopia: A multicenter retrospective cohort study. SAGE Open Med 2024; 12:20503121241253522. [PMID: 38774743 PMCID: PMC11107322 DOI: 10.1177/20503121241253522] [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: 10/18/2023] [Accepted: 04/22/2024] [Indexed: 05/24/2024] Open
Abstract
Background The coronavirus disease 2019 is highly infectious and patients with coronavirus diseases demonstrated a series of clinical symptoms caused by severe acute respiratory syndrome coronavirus-2. Without a break of the wave's hematological profile of the patients is still ambiguous and differs from wave to wave. Objective This study aimed to assess the hematological profile among coronavirus disease 2019 patients during the first and the second waves in Ethiopia. Methods A multi-centered facility-based retrospective cohort study design was conducted at six coronavirus disease 2019 treatment centers. A total of 538 study participants were enrolled in the selected coronavirus disease 2019 treatment centers during the first and second waves of the pandemic. The demographic characteristics, underlying diseases, symptoms, and hematological parameters of patients were recorded. Data were entered into Epi-Data Manager 4.6 and analyzed using STATA 14.2. An independent sample t-test was used to assess the mean differences in hematological parameters across waves. Associations between categorical variables across waves were also determined using Chi-square and Fisher exact tests. Results Among the total study participants, 240 (44.6%) and 298 (55.4%) patients were taken from wave-1 and wave-2, respectively. The average age of the study participants was 56.44 ± 16.25 years. The most frequent comorbidities in this study were hypertension, diabetes mellitus, ischemic heart disease, and asthma. The most presenting symptoms of COVID-19 infection across the two waves were presented. In the first wave, dry cough 166 (69.2%), fatigue 153 (63.75%), shortness of breath 148 (61.67%), and fever accounted 116 (48.33%), while during the second wave, dry cough 242 (81.2%), fatigue 244 (81.88%) shortness of breath 204 (68.47%) and fever account 180 (60.40%). White blood cells (WBC), neutrophils, and lymphocytes had shown increment during the first wave. Platelet count and platelet distribution width (p < 0.001) had significant mean differences across the two waves, while the other had no significant mean difference. Conclusion In the present study, dry cough, fatigue, shortness of breath, and fever were found to be the most presenting symptoms of COVID-19 infection across the two waves. Only the platelet count and platelet distribution width had significant mean differences across the two waves, while the other had no significant mean difference across waves.
Collapse
Affiliation(s)
- Minale Fekadie Baye
- Department of Biochemistry, School of Biomedical Sciences, Jimma University, Jimma, Ethiopia
| | - Diliab Desta
- Department of Anatomy, School of Biomedical Sciences, Jimma University, Jimma, Ethiopia
| | - Samuel Hunegnaw
- Department of Internal Medicine, College of Medicine and Health Sciences, Bahir Dar University, Bahir Dar, Ethiopia
| | - Ahmed Johar
- Department of Biomedical Sciences, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| | - Israel Bekele
- Faculty of Health Sciences, School of Nursing, Jimma University, Jimma, Ethiopia
| | - Kebenesa Angasu
- Faculty of Health Sciences, School of Nursing, Jimma University, Jimma, Ethiopia
| | - Nebiyou Simegnew Bayleyegn
- Faculty of Health Sciences, Department of Surgery, Institute of Health, Jimma University, Jimma, Ethiopia
| | - Habtamu Abebe Getahun
- Public Health Faculty, Department of Epidemiology, Institute of Health, Jimma University, Jimma, Ethiopia
| | - Assefa Legesse Sisay
- Public Health Faculty, Department of Epidemiology, Institute of Health, Jimma University, Jimma, Ethiopia
| |
Collapse
|
11
|
Dong H, Hao Y, Gao P. Vitamin D level in COVID-19 patients has positive correlations with autophagy and negative correlations with disease severity. Front Pharmacol 2024; 15:1388348. [PMID: 38783947 PMCID: PMC11112027 DOI: 10.3389/fphar.2024.1388348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024] Open
Abstract
Background and Objectives There is still incomplete understanding of the pathogenesis of COVID-19. Calcitriol, the main form of vitamin D in serum, regulates immune responses and increases resistance to pathogens, but the mechanism by which it protects against COVID-19 is uncertain. Autophagy has antiviral effects and helps to maintain homeostasis, but its specific role in COVID-19 is also uncertain. Both vitamin D and autophagy have important functions in the lung microenvironment. This study examined the relationship of serum vitamin D and autophagy-related proteins in patients with COVID-19 and evaluated their potential use as biomarkers. Methods Blood samples from COVID-19 patients at the Second Hospital of Jilin University were collected. The levels of vitamin D, autophagy-related proteins (Becline 1 [BECN1] and autophagy-related 7 [ATG7]), and inflammatory markers (TNF-α and IL-1β) were measured using enzyme-linked immunosorbent assays. Results We examined 25 patients with mild/moderate COVID-19 and 27 patients with severe/critical COVID-19. The group with severe/critical COVID-19 had more abnormalities in many laboratory indicators, including lower levels of autophagy markers (BECN1 and ATG7) and vitamin D, and higher levels of inflammatory markers (TNF-α and IL-1β). Partial correlation analysis showed that vitamin D had strong positive correlations with ATG7 (r = 0.819, p < 0.001) and BECN1 (r = 0.900, p < 0.001). Conclusion Our results demonstrated that the vitamin D level had significant negative correlations with COVID-19 severity and strong positive correlations with autophagy. These findings enhance our understanding of the pathogenesis of COVID-19, and provide a theoretical basis for clinical interventions that target autophagy and vitamin D.
Collapse
Affiliation(s)
| | | | - Peng Gao
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
12
|
Bhavani SV, Robichaux C, Verhoef PA, Churpek MM, Coopersmith CM. Using Trajectories of Bedside Vital Signs to Identify COVID-19 Subphenotypes. Chest 2024; 165:529-539. [PMID: 37748574 PMCID: PMC10925543 DOI: 10.1016/j.chest.2023.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/23/2023] [Accepted: 09/18/2023] [Indexed: 09/27/2023] Open
Abstract
BACKGROUND Trajectories of bedside vital signs have been used to identify sepsis subphenotypes with distinct outcomes and treatment responses. The objective of this study was to validate the vitals trajectory model in a multicenter cohort of patients hospitalized with COVID-19 and to evaluate the clinical characteristics and outcomes of the resulting subphenotypes. RESEARCH QUESTION Can the trajectory of routine bedside vital signs identify COVID-19 subphenotypes with distinct clinical characteristics and outcomes? STUDY DESIGN AND METHODS The study included adult patients admitted with COVID-19 to four academic hospitals in the Emory Healthcare system between March 1, 2020, and May 31, 2022. Using a validated group-based trajectory model, we classified patients into previously defined vital sign trajectories using oral temperature, heart rate, respiratory rate, and systolic and diastolic BP measured in the first 8 h of hospitalization. Clinical characteristics, biomarkers, and outcomes were compared between subphenotypes. Heterogeneity of treatment effect to tocilizumab was evaluated. RESULTS The 7,065 patients with hospitalized COVID-19 were classified into four subphenotypes: group A (n = 1,429, 20%)-high temperature, heart rate, respiratory rate, and hypotensive; group B (1,454, 21%)-high temperature, heart rate, respiratory rate, and hypertensive; group C (2,996, 42%)-low temperature, heart rate, respiratory rate, and normotensive; and group D (1,186, 17%)-low temperature, heart rate, respiratory rate, and hypotensive. Groups A and D had higher ORs of mechanical ventilation, vasopressors, and 30-day inpatient mortality (P < .001). On comparing patients receiving tocilizumab (n = 55) with those who met criteria for tocilizumab but were admitted before its use (n = 461), there was significant heterogeneity of treatment effect across subphenotypes in the association of tocilizumab with 30-day mortality (P = .001). INTERPRETATION By using bedside vital signs available in even low-resource settings, we found novel subphenotypes associated with distinct manifestations of COVID-19, which could lead to preemptive and targeted treatments.
Collapse
Affiliation(s)
| | - Chad Robichaux
- Department of Biomedical Informatics, Emory University, Atlanta, GA
| | - Philip A Verhoef
- Department of Medicine, University of Hawaii John A. Burns School of Medicine, Honolulu, HI; Hawaii Permanente Medical Group, Honolulu, HI
| | | | - Craig M Coopersmith
- Emory Critical Care Center, Atlanta, GA; Department of Surgery, Emory University, Atlanta, GA
| |
Collapse
|
13
|
Queiroz MAF, Brito WRDS, Pereira KAS, Pereira LMS, Amoras EDSG, Lima SS, Santos EFD, Costa FPD, Sarges KMLD, Cantanhede MHD, Brito MTFMD, Silva ALSD, Leite MDM, Viana MDNDSDA, Rodrigues FBB, Silva RD, Viana GMR, Chaves TDSS, Veríssimo ADOL, Carvalho MDS, Henriques DF, Silva CPD, Nunes JAL, Costa IB, Cayres-Vallinoto IMV, Brasil-Costa I, Quaresma JAS, Falcão LFM, Santos EJMD, Vallinoto ACR. Severe COVID-19 and long COVID are associated with high expression of STING, cGAS and IFN-α. Sci Rep 2024; 14:4974. [PMID: 38424312 PMCID: PMC10904751 DOI: 10.1038/s41598-024-55696-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 02/27/2024] [Indexed: 03/02/2024] Open
Abstract
The cGAS-STING pathway appears to contribute to dysregulated inflammation during coronavirus disease 2019 (COVID-19); however, inflammatory factors related to long COVID are still being investigated. In the present study, we evaluated the association of cGAS and STING gene expression levels and plasma IFN-α, TNF-α and IL-6 levels with COVID-19 severity in acute infection and long COVID, based on analysis of blood samples from 148 individuals, 87 with acute COVID-19 and 61 in the post-COVID-19 period. Quantification of gene expression was performed by real-time PCR, and cytokine levels were quantified by ELISA and flow cytometry. In acute COVID-19, cGAS, STING, IFN-α, TNF-α, and IL-6 levels were higher in patients with severe disease than in those with nonsevere manifestations (p < 0.05). Long COVID was associated with elevated cGAS, STING and IFN-α levels (p < 0.05). Activation of the cGAS-STING pathway may contribute to an intense systemic inflammatory state in severe COVID-19 and, after infection resolution, induce an autoinflammatory disease in some tissues, resulting in long COVID.
Collapse
Affiliation(s)
- Maria Alice Freitas Queiroz
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil.
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil.
| | - Wandrey Roberto Dos Santos Brito
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Keise Adrielle Santos Pereira
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Leonn Mendes Soares Pereira
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | | | - Sandra Souza Lima
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Erika Ferreira Dos Santos
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Flávia Póvoa da Costa
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Kevin Matheus Lima de Sarges
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Marcos Henrique Damasceno Cantanhede
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | | | | | - Mauro de Meira Leite
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Maria de Nazaré do Socorro de Almeida Viana
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Fabíola Brasil Barbosa Rodrigues
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Rosilene da Silva
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Giselle Maria Rachid Viana
- Laboratory of Basic Research On Malaria, Parasitology Section, Evandro Chagas Institute, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua, Brazil
| | - Tânia do Socorro Souza Chaves
- Laboratory of Basic Research On Malaria, Parasitology Section, Evandro Chagas Institute, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua, Brazil
- School of Medicine, Institute of Medical Sciences, Federal University of Pará, Belém, Pará, Brazil
| | | | | | - Daniele Freitas Henriques
- Arbovirology and Hemorrhagic Fevers Section, Evandro Chagas Institute, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua, Brazil
| | - Carla Pinheiro da Silva
- Arbovirology and Hemorrhagic Fevers Section, Evandro Chagas Institute, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua, Brazil
| | - Juliana Abreu Lima Nunes
- Laboratory of Immunology, Section of Virology, Instituto Evandro Chagas, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua, Brazil
| | - Iran Barros Costa
- Laboratory of Immunology, Section of Virology, Instituto Evandro Chagas, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua, Brazil
- Graduate Program in Virology, Evandro Chagas Institute, Department of Science, Technology, Innovation and Strategic Health Inputs, Ministry of Health of Brazil, Ananindeua, Brazil
| | - Izaura Maria Vieira Cayres-Vallinoto
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Igor Brasil-Costa
- Laboratory of Immunology, Section of Virology, Instituto Evandro Chagas, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua, Brazil
- Graduate Program in Virology, Evandro Chagas Institute, Department of Science, Technology, Innovation and Strategic Health Inputs, Ministry of Health of Brazil, Ananindeua, Brazil
| | - Juarez Antônio Simões Quaresma
- Graduate Program in Virology, Evandro Chagas Institute, Department of Science, Technology, Innovation and Strategic Health Inputs, Ministry of Health of Brazil, Ananindeua, Brazil
- Center of Biological and Health Sciences, University of the State of Pará, Belém, Brazil
| | | | - Eduardo José Melo Dos Santos
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Antonio Carlos Rosário Vallinoto
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
- Graduate Program in Virology, Evandro Chagas Institute, Department of Science, Technology, Innovation and Strategic Health Inputs, Ministry of Health of Brazil, Ananindeua, Brazil
| |
Collapse
|
14
|
Hu C, Hu W, Tang B, Bao Q, Jiang X, Tang L, Wang H, He L, Lv M, Xiao Y, Liu C, Li X, Liu Y, Li J, Huang G, Dong Z, Li Z, Guo T, Yang S. Plasma and urine proteomics and gut microbiota analysis reveal potential factors affecting COVID-19 vaccination response. iScience 2024; 27:108851. [PMID: 38318387 PMCID: PMC10838952 DOI: 10.1016/j.isci.2024.108851] [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: 04/14/2023] [Revised: 10/15/2023] [Accepted: 01/03/2024] [Indexed: 02/07/2024] Open
Abstract
The efficacy of COVID-19 vaccination relies on the induction of neutralizing antibodies, which can vary among vaccine recipients. In this study, we investigated the potential factors affecting the neutralizing antibody response by combining plasma and urine proteomics and gut microbiota analysis. We found that activation of the LXR/FXR pathway in plasma was associated with the production of ACE2-RBD-inhibiting antibodies, while urine proteins related to complement system, acute phase response signaling, LXR/FXR, and STAT3 pathways were correlated with neutralizing antibody production. Moreover, we observed a correlation between the gut microbiota and plasma and urine proteins, as well as the vaccination response. Based on the above data, we built a predictive model for vaccination response (AUC = 0.85). Our study provides insights into characteristic plasma and urine proteins and gut microbiota associated with the ACE2-RBD-inhibiting antibodies, which could benefit our understanding of the host response to COVID-19 vaccination.
Collapse
Affiliation(s)
- Changjiang Hu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
- iMarkerlab, Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province, China
- Research Center for Industries of the Future, Westlake University, 600 Dunyu Road, Hangzhou 310030, Zhejiang, China
- Center for Infectious Disease Research, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang, China
| | - Weichao Hu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
- iMarkerlab, Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province, China
- Research Center for Industries of the Future, Westlake University, 600 Dunyu Road, Hangzhou 310030, Zhejiang, China
- Center for Infectious Disease Research, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang, China
| | - Bo Tang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Qiyu Bao
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Xingyu Jiang
- Laboratory Medicine Center, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Li Tang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - He Wang
- iMarkerlab, Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province, China
- Research Center for Industries of the Future, Westlake University, 600 Dunyu Road, Hangzhou 310030, Zhejiang, China
- Center for Infectious Disease Research, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang, China
| | - Lijiao He
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Moyang Lv
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Yufeng Xiao
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Cheng Liu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Xinzhe Li
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Yunyi Liu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Jie Li
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Guiping Huang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Zhen Dong
- iMarkerlab, Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province, China
- Research Center for Industries of the Future, Westlake University, 600 Dunyu Road, Hangzhou 310030, Zhejiang, China
- Center for Infectious Disease Research, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang, China
| | - Zhongjun Li
- Laboratory Medicine Center, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Tiannan Guo
- iMarkerlab, Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province, China
- Research Center for Industries of the Future, Westlake University, 600 Dunyu Road, Hangzhou 310030, Zhejiang, China
- Center for Infectious Disease Research, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang, China
| | - Shiming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| |
Collapse
|
15
|
Spinello I, Saulle E, Quaranta MT, Pelosi E, Castelli G, Cerio A, Pasquini L, Morsilli O, Dupuis ML, Labbaye C. AC-73 and Syrosingopine Inhibit SARS-CoV-2 Entry into Megakaryocytes by Targeting CD147 and MCT4. Viruses 2024; 16:82. [PMID: 38257782 PMCID: PMC10818282 DOI: 10.3390/v16010082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/20/2023] [Accepted: 12/23/2023] [Indexed: 01/24/2024] Open
Abstract
Coagulation disorders are described in COVID-19 and long COVID patients. In particular, SARS-CoV-2 infection in megakaryocytes, which are precursors of platelets involved in thrombotic events in COVID-19, long COVID and, in rare cases, in vaccinated individuals, requires further investigation, particularly with the emergence of new SARS-CoV-2 variants. CD147, involved in the regulation of inflammation and required to fight virus infection, can facilitate SARS-CoV-2 entry into megakaryocytes. MCT4, a co-binding protein of CD147 and a key player in the glycolytic metabolism, could also play a role in SARS-CoV-2 infection. Here, we investigated the susceptibility of megakaryocytes to SARS-CoV-2 infection via CD147 and MCT4. We performed infection of Dami cells and human CD34+ hematopoietic progenitor cells induced to megakaryocytic differentiation with SARS-CoV-2 pseudovirus in the presence of AC-73 and syrosingopine, respective inhibitors of CD147 and MCT4 and inducers of autophagy, a process essential in megakaryocyte differentiation. Both AC-73 and syrosingopine enhance autophagy during differentiation but only AC-73 enhances megakaryocytic maturation. Importantly, we found that AC-73 or syrosingopine significantly inhibits SARS-CoV-2 infection of megakaryocytes. Altogether, our data indicate AC-73 and syrosingopine as inhibitors of SARS-CoV-2 infection via CD147 and MCT4 that can be used to prevent SARS-CoV-2 binding and entry into megakaryocytes, which are precursors of platelets involved in COVID-19-associated coagulopathy.
Collapse
Affiliation(s)
- Isabella Spinello
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.S.); (E.S.); (M.T.Q.); (M.L.D.)
| | - Ernestina Saulle
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.S.); (E.S.); (M.T.Q.); (M.L.D.)
| | - Maria Teresa Quaranta
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.S.); (E.S.); (M.T.Q.); (M.L.D.)
| | - Elvira Pelosi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (E.P.); (G.C.); (A.C.)
| | - Germana Castelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (E.P.); (G.C.); (A.C.)
| | - Annamaria Cerio
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (E.P.); (G.C.); (A.C.)
| | - Luca Pasquini
- Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Ornella Morsilli
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Ageing, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Maria Luisa Dupuis
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.S.); (E.S.); (M.T.Q.); (M.L.D.)
| | - Catherine Labbaye
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.S.); (E.S.); (M.T.Q.); (M.L.D.)
| |
Collapse
|
16
|
Zanini G, Selleri V, Roncati L, Coppi F, Nasi M, Farinetti A, Manenti A, Pinti M, Mattioli AV. Vascular "Long COVID": A New Vessel Disease? Angiology 2024; 75:8-14. [PMID: 36652923 PMCID: PMC9895315 DOI: 10.1177/00033197231153204] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Vascular sequelae following (SARS-CoV-2 coronavirus disease) (COVID)-19 infection are considered as "Long Covid (LC)" disease, when occurring 12 weeks after the original infection. The paucity of specific data can be obviated by translating pathophysiological elements from the original Severe Acute Respiratory Syndrome-Corona Virus (SARS-CoV-2) infection (In a microcirculatory system, a first "endotheliitis," is often followed by production of "Neutrophil Extracellular Trap," and can evolve into a more complex leukocytoklastic-like and hyperimmune vasculitis. In medium/large-sized vessels, this corresponds to endothelial dysfunction, leading to an accelerated progression of pre-existing atherosclerotic plaques through an increased deposition of platelets, circulating inflammatory cells and proteins. Associated dysregulated immune and pro-coagulant conditions can directly cause thrombo-embolic arterial or venous complications. In order to implement appropriate treatment, physicians need to consider vascular pathologies observed after SARS-Cov-2 infections as possible "LC" disease.
Collapse
Affiliation(s)
- Giada Zanini
- Department of Life Sciences, University of Modena and Reggio
Emilia, Modena, Italy
| | - Valentina Selleri
- Department of Life Sciences, University of Modena and Reggio
Emilia, Modena, Italy
- Istituto Nazionale per le Ricerche
Cardiovascolari, University of Modena and Reggio
Emilia, Modena, Italy
| | - Luca Roncati
- Pathology Unit, University of Modena and Reggio
Emilia. Polyclinic Hospital, Modena, Italy
| | - Francesca Coppi
- Department of Medical and Surgical
Sciences for Children and Adults, University of Modena and. Reggio
Emilia, Modena, Italy
| | - Milena Nasi
- Department of Surgical, Medical and Dental Sciences
University of Modena and Reggio Emilia, Modena, Italy
| | - Alberto Farinetti
- Department of Medical and Surgical
Sciences for Children and Adults, University of Modena and. Reggio
Emilia, Modena, Italy
| | - Antonio Manenti
- Department of Medical and Surgical
Sciences for Children and Adults, University of Modena and. Reggio
Emilia, Modena, Italy
| | - Marcello Pinti
- Department of Life Sciences, University of Modena and Reggio
Emilia, Modena, Italy
| | - Anna Vittoria Mattioli
- Istituto Nazionale per le Ricerche
Cardiovascolari, University of Modena and Reggio
Emilia, Modena, Italy
- Department of Medical and Surgical
Sciences for Children and Adults, University of Modena and. Reggio
Emilia, Modena, Italy
| |
Collapse
|
17
|
Qidwai T. Cytokine storm in COVID-19 and malaria: Annals of pro-inflammatory cytokines. Cytokine 2024; 173:156420. [PMID: 37976701 DOI: 10.1016/j.cyto.2023.156420] [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: 07/24/2023] [Revised: 10/28/2023] [Accepted: 11/01/2023] [Indexed: 11/19/2023]
Abstract
Infectious diseases are affecting the people worldwide. Mostly, infectious agents activate excessive production of cytokines so called cytokine storm. Among the infectious diseases COVID-19 is one of the deadliest diseases affecting individuals all over the world, moreover, Plasmodium falciparum malaria and HIV are major killers. An excessive pro-inflammatory response is one of the major causes of pathological conditions in these diseases. It is important to investigate the pathophysiology in the infectious diseases such as COVID-19, malaria and HIV as there is no concrete therapy against them so far. Exploration of excessive pro-inflammation could be important for therapeutic intervention. In this article, an attempt has been made to analyze the pathological conditions arise due to excessive inflammatory response in COVID-19, malaria and other infectious diseases. Targeting excessive pro-inflammatory response/cytokine storm in infectious diseases could be a useful strategy.
Collapse
Affiliation(s)
- Tabish Qidwai
- Faculty of Biotechnology, Shri Ramswaroop Memorial University, Lucknow Deva Road, Uttar Pradesh 225003, India.
| |
Collapse
|
18
|
Yu B, Drelich A, Hsu J, Tat V, Peng BH, Wei Q, Wang J, Wang H, Wages J, Mendelsohn AR, Larrick JW, Tseng CT. Protective Efficacy of Novel Engineered Human ACE2-Fc Fusion Protein Against Pan-SARS-CoV-2 Infection In Vitro and in Vivo. J Med Chem 2023; 66:16646-16657. [PMID: 38100534 DOI: 10.1021/acs.jmedchem.3c01201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
Enduring occurrence of severe COVID-19 for unvaccinated, aged, or immunocompromised individuals remains an urgent need. Soluble human angiotensin-converting enzyme 2 (ACE2) has been used as a decoy receptor to inhibit SARS-CoV-2 infection, which is limited by moderate affinity. We describe an engineered, high-affinity ACE2 that is consistently effective in tissue cultures in neutralizing all strains tested, including Delta and Omicron. We also found that treatment of AC70 hACE2 transgenic mice with hACE2-Fc receptor decoys effectively reduced viral infection, attenuated tissue histopathology, and delayed the onset of morbidity and mortality caused by SARS-CoV-2 infection. We believe that using this ACE2-Fc protein would be less likely to promote the escape mutants of SARS-CoV-2 as frequently as did those neutralizing antibody therapies. Together, our results emphasize the suitability of our newly engineered hACE2-Fc fusion protein for further development as a potent antiviral agent against Pan-SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Bo Yu
- Larix Bioscience LLC, Sunnyvale, California 94089, United States
| | - Aleksandra Drelich
- Departments of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Jason Hsu
- Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Vivian Tat
- Pathology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Bi-Hung Peng
- Neuroscience, Cell Biology & Anatomy, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Qisheng Wei
- Larix Bioscience LLC, Sunnyvale, California 94089, United States
| | - Jianming Wang
- Larix Bioscience LLC, Sunnyvale, California 94089, United States
| | - Hong Wang
- Larix Bioscience LLC, Sunnyvale, California 94089, United States
| | - John Wages
- Larix Bioscience LLC, Sunnyvale, California 94089, United States
| | | | - James W Larrick
- Larix Bioscience LLC, Sunnyvale, California 94089, United States
| | - Chien-Te Tseng
- Departments of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas 77555, United States
- Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555, United States
- Pathology, University of Texas Medical Branch, Galveston, Texas 77555, United States
- Center for Biodefense and Emerging Infectious Disease, University of Texas Medical Branch, Galveston, Texas 77555, United States
| |
Collapse
|
19
|
Liu L, Zhou C, Jiang H, Wei H, Zhou Y, Zhou C, Ji X. Epidemiology, pathogenesis, and management of Coronavirus disease 2019-associated stroke. Front Med 2023; 17:1047-1067. [PMID: 38165535 DOI: 10.1007/s11684-023-1041-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 10/15/2023] [Indexed: 01/03/2024]
Abstract
The Coronavirus disease 2019 (COVID-19) epidemic has triggered a huge impact on healthcare, socioeconomics, and other aspects of the world over the past three years. An increasing number of studies have identified a complex relationship between COVID-19 and stroke, although active measures are being implemented to prevent disease transmission. Severe COVID-19 may be associated with an increased risk of stroke and increase the rates of disability and mortality, posing a serious challenge to acute stroke diagnosis, treatment, and care. This review aims to provide an update on the influence of COVID-19 itself or vaccines on stroke, including arterial stroke (ischemic stroke and hemorrhagic stroke) and venous stroke (cerebral venous thrombosis). Additionally, the neurovascular mechanisms involved in SARS-CoV-2 infection and the clinical characteristics of stroke in the COVID-19 setting are presented. Evidence on vaccinations, potential therapeutic approaches, and effective strategies for stroke management has been highlighted.
Collapse
Affiliation(s)
- Lu Liu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China
- Neurology and Intracranial Hypertension and Cerebral Venous Disease Center, National Health Commission of China, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China
| | - Chenxia Zhou
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China
- Neurology and Intracranial Hypertension and Cerebral Venous Disease Center, National Health Commission of China, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China
| | - Huimin Jiang
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Huimin Wei
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Yifan Zhou
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Chen Zhou
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, 100069, China.
- Department of Neurology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China.
| | - Xunming Ji
- Neurology and Intracranial Hypertension and Cerebral Venous Disease Center, National Health Commission of China, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China.
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, 100069, China.
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China.
| |
Collapse
|
20
|
Wang Y, Shen M, Li Y, Shao J, Zhang F, Guo M, Zhang Z, Zheng S. COVID-19-associated liver injury: Adding fuel to the flame. Cell Biochem Funct 2023; 41:1076-1092. [PMID: 37947373 DOI: 10.1002/cbf.3883] [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: 08/23/2023] [Revised: 10/19/2023] [Accepted: 10/21/2023] [Indexed: 11/12/2023]
Abstract
COVID-19 is mainly characterized by respiratory disorders and progresses to multiple organ involvement in severe cases. With expansion of COVID-19 and SARS-CoV-2 research, correlative liver injury has been revealed. It is speculated that COVID-19 patients exhibited abnormal liver function, as previously observed in the SARS and MERS pandemics. Furthermore, patients with underlying diseases such as chronic liver disease are more susceptible to SARS-CoV-2 and indicate a poor prognosis accompanied by respiratory symptoms, systemic inflammation, or metabolic diseases. Therefore, COVID-19 has the potential to impair liver function, while individuals with preexisting liver disease suffer from much worse infected conditions. COVID-19 related liver injury may be owing to direct cytopathic effect, immune dysfunction, gut-liver axis interaction, and inappropriate medication use. However, discussions on these issues are infancy. Expanding research have revealed that angiotensin converting enzyme 2 (ACE2) expression mediated the combination of virus and target cells, iron metabolism participated in the virus life cycle and the fate of target cells, and amino acid metabolism regulated immune response in the host cells, which are all closely related to liver health. Further exploration holds great significance in elucidating the pathogenesis, facilitating drug development, and advancing clinical treatment of COVID-19-related liver injury. This article provides a review of the clinical and laboratory hepatic characteristics in COVID-19 patients, describes the etiology and impact of liver injury, and discusses potential pathophysiological mechanisms.
Collapse
Affiliation(s)
- Yingqian Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Min Shen
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Yujia Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiangjuan Shao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Feng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Mei Guo
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zili Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shizhong Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
21
|
Dushianthan A, Bracegirdle L, Cusack R, Cumpstey AF, Postle AD, Grocott MPW. Alveolar Hyperoxia and Exacerbation of Lung Injury in Critically Ill SARS-CoV-2 Pneumonia. Med Sci (Basel) 2023; 11:70. [PMID: 37987325 PMCID: PMC10660857 DOI: 10.3390/medsci11040070] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/17/2023] [Accepted: 10/30/2023] [Indexed: 11/22/2023] Open
Abstract
Acute hypoxic respiratory failure (AHRF) is a prominent feature of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) critical illness. The severity of gas exchange impairment correlates with worse prognosis, and AHRF requiring mechanical ventilation is associated with substantial mortality. Persistent impaired gas exchange leading to hypoxemia often warrants the prolonged administration of a high fraction of inspired oxygen (FiO2). In SARS-CoV-2 AHRF, systemic vasculopathy with lung microthrombosis and microangiopathy further exacerbates poor gas exchange due to alveolar inflammation and oedema. Capillary congestion with microthrombosis is a common autopsy finding in the lungs of patients who die with coronavirus disease 2019 (COVID-19)-associated acute respiratory distress syndrome. The need for a high FiO2 to normalise arterial hypoxemia and tissue hypoxia can result in alveolar hyperoxia. This in turn can lead to local alveolar oxidative stress with associated inflammation, alveolar epithelial cell apoptosis, surfactant dysfunction, pulmonary vascular abnormalities, resorption atelectasis, and impairment of innate immunity predisposing to secondary bacterial infections. While oxygen is a life-saving treatment, alveolar hyperoxia may exacerbate pre-existing lung injury. In this review, we provide a summary of oxygen toxicity mechanisms, evaluating the consequences of alveolar hyperoxia in COVID-19 and propose established and potential exploratory treatment pathways to minimise alveolar hyperoxia.
Collapse
Affiliation(s)
- Ahilanandan Dushianthan
- NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK; (L.B.); (R.C.); (A.F.C.); (A.D.P.); (M.P.W.G.)
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Luke Bracegirdle
- NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK; (L.B.); (R.C.); (A.F.C.); (A.D.P.); (M.P.W.G.)
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Rebecca Cusack
- NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK; (L.B.); (R.C.); (A.F.C.); (A.D.P.); (M.P.W.G.)
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Andrew F. Cumpstey
- NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK; (L.B.); (R.C.); (A.F.C.); (A.D.P.); (M.P.W.G.)
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Anthony D. Postle
- NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK; (L.B.); (R.C.); (A.F.C.); (A.D.P.); (M.P.W.G.)
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Michael P. W. Grocott
- NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK; (L.B.); (R.C.); (A.F.C.); (A.D.P.); (M.P.W.G.)
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| |
Collapse
|
22
|
Abdullah M, Ali A, Usman M, Naz A, Qureshi JA, Bajaber MA, Zhang X. Post COVID-19 complications and follow up biomarkers. NANOSCALE ADVANCES 2023; 5:5705-5716. [PMID: 37881715 PMCID: PMC10597564 DOI: 10.1039/d3na00342f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 09/11/2023] [Indexed: 10/27/2023]
Abstract
Millions of people were infected by the coronavirus disease (COVID-19) epidemic, which left a huge burden on the care of post COVID-19 survivors around the globe. The self-reported COVID-19 symptoms were experienced by an estimated 1.3 million people in the United Kingdom (2% of the population), and these symptoms persisted for about 4 weeks from the beginning of the infection. The symptoms most frequently reported were exhaustion, shortness of breath, muscular discomfort, joint pain, headache, cough, chest pain, cognitive impairment, memory loss, anxiety, sleep difficulties, diarrhea, and a decreased sense of smell and taste in post-COVID-19 affected people. The post COVID-19 complications were frequently related to the respiratory, cardiac, nervous, psychological and musculoskeletal systems. The lungs, liver, kidneys, heart, brain and other organs had been impaired by hypoxia and inflammation in post COVID-19 individuals. The upregulation of substance "P" (SP) and various cytokines such as tumor necrosis factor-alpha (TNF-α), interleukin 6 (IL-6), interleukin 10 (IL-10), interleukin 1 beta (IL-1β), angiotensin-converting enzyme 2 (ACE2) and chemokine C-C motif ligand 3 (CCL3) has muddled respiratory, cardiac, neuropsychiatric, dermatological, endocrine, musculoskeletal, gastrointestinal, renal and genitourinary complications in post COVID-19 people. To prevent these complications from worsening, it was therefore important to study how these biomarkers were upregulated and block their receptors.
Collapse
Affiliation(s)
- Muhammad Abdullah
- Institute of Molecular Biology and Biotechnology, University of Lahore Pakistan
| | - Amjed Ali
- University Institute of Physical Therapy, University of Lahore Pakistan
| | - Muhammad Usman
- Department of Bioinformatics, School of Medical Informatics and Engineering, Xuzhou Medical University Xuzhou China
| | - Anam Naz
- Institute of Molecular Biology and Biotechnology, University of Lahore Pakistan
| | - Javed Anver Qureshi
- Institute of Molecular Biology and Biotechnology, University of Lahore Pakistan
| | - Majed A Bajaber
- Department of Chemistry, Faculty of Science, King Khalid University P.O. Box 9004 Abha 61413 Saudi Arabia
| | - Xiao Zhang
- Department of Bioinformatics, School of Medical Informatics and Engineering, Xuzhou Medical University Xuzhou China
| |
Collapse
|
23
|
Berentschot JC, Drexhage HA, Aynekulu Mersha DG, Wijkhuijs AJM, GeurtsvanKessel CH, Koopmans MPG, Voermans JJC, Hendriks RW, Nagtzaam NMA, de Bie M, Heijenbrok-Kal MH, Bek LM, Ribbers GM, van den Berg-Emons RJG, Aerts JGJV, Dik WA, Hellemons ME. Immunological profiling in long COVID: overall low grade inflammation and T-lymphocyte senescence and increased monocyte activation correlating with increasing fatigue severity. Front Immunol 2023; 14:1254899. [PMID: 37881427 PMCID: PMC10597688 DOI: 10.3389/fimmu.2023.1254899] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/14/2023] [Indexed: 10/27/2023] Open
Abstract
Background Many patients with SARS-CoV-2 infection develop long COVID with fatigue as one of the most disabling symptoms. We performed clinical and immune profiling of fatigued and non-fatigued long COVID patients and age- and sex-matched healthy controls (HCs). Methods Long COVID symptoms were assessed using patient-reported outcome measures, including the fatigue assessment scale (FAS, scores ≥22 denote fatigue), and followed up to one year after hospital discharge. We assessed inflammation-related genes in circulating monocytes, serum levels of inflammation-regulating cytokines, and leukocyte and lymphocyte subsets, including major monocyte subsets and senescent T-lymphocytes, at 3-6 months post-discharge. Results We included 37 fatigued and 36 non-fatigued long COVID patients and 42 HCs. Fatigued long COVID patients represented a more severe clinical profile than non-fatigued patients, with many concurrent symptoms (median 9 [IQR 5.0-10.0] vs 3 [1.0-5.0] symptoms, p<0.001), and signs of cognitive failure (41%) and depression (>24%). Immune abnormalities that were found in the entire group of long COVID patients were low grade inflammation (increased inflammatory gene expression in monocytes, increased serum pro-inflammatory cytokines) and signs of T-lymphocyte senescence (increased exhausted CD8+ TEMRA-lymphocytes). Immune profiles did not significantly differ between fatigued and non-fatigued long COVID groups. However, the severity of fatigue (total FAS score) significantly correlated with increases of intermediate and non-classical monocytes, upregulated gene levels of CCL2, CCL7, and SERPINB2 in monocytes, increases in serum Galectin-9, and higher CD8+ T-lymphocyte counts. Conclusion Long COVID with fatigue is associated with many concurrent and persistent symptoms lasting up to one year after hospitalization. Increased fatigue severity associated with stronger signs of monocyte activation in long COVID patients and potentially point in the direction of monocyte-endothelial interaction. These abnormalities were present against a background of immune abnormalities common to the entire group of long COVID patients.
Collapse
Affiliation(s)
- Julia C. Berentschot
- Department of Respiratory Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Hemmo A. Drexhage
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | | | | | | | - Marion P. G. Koopmans
- Department of Viroscience, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Jolanda J. C. Voermans
- Department of Viroscience, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Rudi W. Hendriks
- Department of Respiratory Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Nicole M. A. Nagtzaam
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Maaike de Bie
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Majanka H. Heijenbrok-Kal
- Department of Rehabilitation Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- Rijndam Rehabilitation, Rotterdam, Netherlands
| | - L. Martine Bek
- Department of Rehabilitation Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Gerard M. Ribbers
- Department of Rehabilitation Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- Rijndam Rehabilitation, Rotterdam, Netherlands
| | | | - Joachim G. J. V. Aerts
- Department of Respiratory Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Willem A. Dik
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Merel E. Hellemons
- Department of Respiratory Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
24
|
Gómez-Sánchez L, Tamayo-Morales O, Suárez-Moreno N, Bermejo-Martín JF, Domínguez-Martín A, Martín-Oterino JA, Martín-González JI, González-Calle D, García-García Á, Lugones-Sánchez C, González-Sánchez S, Jiménez-Gómez R, García-Ortiz L, Gómez-Marcos MA, Navarro-Matías E. Relationship between the structure, function and endothelial damage, and vascular ageing and the biopsychological situation in adults diagnosed with persistent COVID (BioICOPER study). A research protocol of a cross-sectional study. Front Physiol 2023; 14:1236430. [PMID: 37772064 PMCID: PMC10523018 DOI: 10.3389/fphys.2023.1236430] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/17/2023] [Indexed: 09/30/2023] Open
Abstract
Background: SARS-CoV-2 infection affects the vascular endothelium, which mediates the inflammatory and thrombotic cascade. Moreover, alterations in the endothelium are related to arterial stiffness, which has been established as a marker of cardiovascular disease. The objective of this study is to analyse how the structure, vascular function, vascular ageing and endothelial damage are related to the biopsychological situation in adults diagnosed with persistent COVID and the differences by gender. Methods: This cross-sectional, descriptive, observational study will be carried out in the Primary Care Research Unit of Salamanca (APISAL) and in the BioSepsis laboratory of the University of Salamanca. The sample will be selected from the persistent COVID monographic office at the Internal Medicine Service of the University Hospital of Salamanca, and from the population of subjects diagnosed with persistent COVID in the clinical history of Primary Care. Through consecutive sampling, the study will include 300 individuals diagnosed with persistent COVID who meet the diagnosis criteria established by the WHO, after they sign the informed consent. Endothelial damage biomarkers will be measured using ELLA-SimplePlexTM technology (Biotechne). Their vascular structure and function will be analysed by measuring the carotid intima-media thickness (Sonosite Micromax); the pulse wave and carotid-femoral pulse wave velocity (cfPWV) will be recorded with Sphygmocor System®. Cardio Ankle Vascular Index (CAVI), brachial-ankle pulse wave velocity (baPWV) and ankle-brachial index will be analysed with Vasera VS-2000®. The integral assessment of the subjects with persistent COVID will be conducted with different scales that evaluate fatigue, sleep, dyspnea, quality of life, attention, nutrition state, and fragility. We will also evaluate their lifestyles (diet, physical activity, smoking habits and alcohol consumption), psychological factors, and cognitive deterioration, which will be gathered through validated questionnaires; moreover, physical activity will be objectively measured using a pedometer for 7 days. Body composition will be measured through impedance using an Inbody 230. Vascular ageing will be calculated with 10 and 90 percentiles of cfPWV and baPWV. Furthermore, we will analyse the presence of vascular injury in the retina, heart, kidneys and brain, as well as cardiovascular risk. Demographic and analytical variables will also be gathered. Discussion: Arterial stiffness reflects the mechanic and functional properties of the arterial wall, showing the changes in arterial pressure, blood flow, and vascular diameter that occur with each heartbeat. SARS-CoV-2 affects the endothelial cells that are infected with this virus, increasing the production of pro-inflammatory cytokines and pro-thrombotic factors, which can cause early vascular ageing and an increase of arterial stiffness. Persistent COVID is a complex heterogeneous disorder that affects the lives of millions of people worldwide. The identifications of potential risk factors to better understand who is at risk of developing persistent COVID is important, since this would enable early and appropriate clinical support. It is unknown whether vascular alterations caused by COVID-19 resolve after acute infection or remain over time, favouring the increase of arterial stiffness and early vascular ageing. Therefore, it is necessary to propose studies that analyse the evolution of persistent COVID in this group of patients, as well as the possible variables that influence it. Clinical Trial registration: ClinicalTrials.gov, identifier NCT05819840.
Collapse
Affiliation(s)
- Leticia Gómez-Sánchez
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Salamanca, Spain
- Hospital de la Paz de Madrid, Servicio de Urgencias, Madrid, Spain
| | - Olaya Tamayo-Morales
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), Salamanca, Spain
| | - Nuria Suárez-Moreno
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Castilla and León Health Service–SACYL, Gerencia Regional de Salud, Valladolid, Spain
| | - Jesus F. Bermejo-Martín
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Andrea Domínguez-Martín
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Castilla and León Health Service–SACYL, Gerencia Regional de Salud, Valladolid, Spain
| | - José A. Martín-Oterino
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Castilla and León Health Service–SACYL, Gerencia Regional de Salud, Valladolid, Spain
- Hospital Universitario de Salamanca, Internal Medicine Department, Salamanca, Spain
- Department of Biomedical and Diagnostic Sciences, University of Salamanca, Salamanca, Spain
| | - José I. Martín-González
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Castilla and León Health Service–SACYL, Gerencia Regional de Salud, Valladolid, Spain
- Hospital Universitario de Salamanca, Internal Medicine Department, Salamanca, Spain
- Department of Biomedical and Diagnostic Sciences, University of Salamanca, Salamanca, Spain
| | - David González-Calle
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Castilla and León Health Service–SACYL, Gerencia Regional de Salud, Valladolid, Spain
- Department of Biomedical and Diagnostic Sciences, University of Salamanca, Salamanca, Spain
- Hospital Universitario de Salamanca, Cardiology Department, Salamanca, Spain
| | - Ángel García-García
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Castilla and León Health Service–SACYL, Gerencia Regional de Salud, Valladolid, Spain
- Department of Biomedical and Diagnostic Sciences, University of Salamanca, Salamanca, Spain
- Hospital Universitario de Salamanca, Emergency Department, Salamanca, Spain
| | - Cristina Lugones-Sánchez
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), Salamanca, Spain
| | - Susana González-Sánchez
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), Salamanca, Spain
| | - Raquel Jiménez-Gómez
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Castilla and León Health Service–SACYL, Gerencia Regional de Salud, Valladolid, Spain
| | - Luis García-Ortiz
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), Salamanca, Spain
- Castilla and León Health Service–SACYL, Gerencia Regional de Salud, Valladolid, Spain
- Department of Biomedical and Diagnostic Sciences, University of Salamanca, Salamanca, Spain
| | - Manuel A. Gómez-Marcos
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), Salamanca, Spain
- Castilla and León Health Service–SACYL, Gerencia Regional de Salud, Valladolid, Spain
- Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Elena Navarro-Matías
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Castilla and León Health Service–SACYL, Gerencia Regional de Salud, Valladolid, Spain
| | | |
Collapse
|
25
|
Radkhah H, Mansouri ES, Rahimipour Anaraki S, Gholizadeh Mesgarha M, Sheikhy A, Khadembashiri MM, Khadembashiri MA, Eslami M, Mahmoodi T, Inanloo B, Pour Mohammad A. Predictive value of hematological indices on incidence and severity of pulmonary embolism in COVID-19 patients. Immun Inflamm Dis 2023; 11:e1012. [PMID: 37773719 PMCID: PMC10540144 DOI: 10.1002/iid3.1012] [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: 03/30/2023] [Revised: 07/28/2023] [Accepted: 08/31/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND Pulmonary thromboembolism (PTE) is a common complication of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which raises the COVID-19 disease's fatality rate from 3% to 45%. Nevertheless, due to fairly indistinguishable clinical symptoms and a lack of validated clinical prediction models, PTE diagnosis in COVID-19 patients is challenging. This study aims to investigate the applicability of hematological indices to predict PTE incidence and its severity in SARS-CoV-2 patients. METHODS A retrospective cohort study was conducted on hospitalized patients with a confirmed diagnosis of SARS-CoV-2 infection who underwent CT angiography to assess probable PTE in them. The correlation between complete blood count parameters 1 day before CT angiography and CT angiography outcomes, and simplified pulmonary embolism severity index (s-PESI) was investigated. RESULTS We discovered that among individuals with a probable PTE, males and those with higher platelet-to-lymphocyte (PLR) and neutrophil-to-lymphocyte (NLR) ratios had a greater likelihood of PTE incidence (p < .001, .027, and .037, respectively). PLR was a significant and independent predictor of PTE with a p value of .045. Moreover, a higher neutrophil count was associated with a higher s-PESI score in COVID-19 patients developing PTE (p: .038). CONCLUSIONS Among hematological indices, NLR and more precisely PLR are cost-effective and simply calculable markers that can assist physicians in determining whether or not COVID-19 patients with clinically probable PTE require CT angiography and the higher neutrophil count can be employed as an indicator of PTE severity in COVID-19 patients. Further large multicenter and prospective studies are warranted to corroborate these observations.
Collapse
Affiliation(s)
- Hanieh Radkhah
- Department of Internal Medicine, School of Medicine, Sina HospitalTehran University of Medical Sciences (TUMS)TehranIran
| | - Ensieh Sadat Mansouri
- Department of Internal Medicine, School of Medicine, Sina HospitalTehran University of Medical Sciences (TUMS)TehranIran
| | | | | | - Ali Sheikhy
- Students' Scientific Research CenterTehran University of Medical Sciences (TUMS)TehranIran
| | | | | | - Mohamad Eslami
- Students' Scientific Research CenterTehran University of Medical Sciences (TUMS)TehranIran
| | - Tara Mahmoodi
- Students' Scientific Research CenterTehran University of Medical Sciences (TUMS)TehranIran
| | - Behnaz Inanloo
- Sina HospitalTehran University of Medical Sciences (TUMS)TehranIran
| | | |
Collapse
|
26
|
Yao M, Ma J, Wu D, Fang C, Wang Z, Guo T, Mo J. Neutrophil extracellular traps mediate deep vein thrombosis: from mechanism to therapy. Front Immunol 2023; 14:1198952. [PMID: 37680629 PMCID: PMC10482110 DOI: 10.3389/fimmu.2023.1198952] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 08/10/2023] [Indexed: 09/09/2023] Open
Abstract
Deep venous thrombosis (DVT) is a part of venous thromboembolism (VTE) that clinically manifests as swelling and pain in the lower limbs. The most serious clinical complication of DVT is pulmonary embolism (PE), which has a high mortality rate. To date, its underlying mechanisms are not fully understood, and patients usually present with clinical symptoms only after the formation of the thrombus. Thus, it is essential to understand the underlying mechanisms of deep vein thrombosis for an early diagnosis and treatment of DVT. In recent years, many studies have concluded that Neutrophil Extracellular Traps (NETs) are closely associated with DVT. These are released by neutrophils and, in addition to trapping pathogens, can mediate the formation of deep vein thrombi, thereby blocking blood vessels and leading to the development of disease. Therefore, this paper describes the occurrence and development of NETs and discusses the mechanism of action of NETs on deep vein thrombosis. It aims to provide a direction for improved diagnosis and treatment of deep vein thrombosis in the near future.
Collapse
Affiliation(s)
- Mengting Yao
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jiacheng Ma
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Dongwen Wu
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Chucun Fang
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Zilong Wang
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Tianting Guo
- Department of Orthopedics, Guangdong Provincial People’s Hospital Ganzhou Hospital, Ganzhou Municipal Hospital, Ganzhou, Jiangxi, China
| | - Jianwen Mo
- Department of Orthopedic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
27
|
Pawar VA, Tyagi A, Verma C, Sharma KP, Ansari S, Mani I, Srivastva SK, Shukla PK, Kumar A, Kumar V. Unlocking therapeutic potential: integration of drug repurposing and immunotherapy for various disease targeting. Am J Transl Res 2023; 15:4984-5006. [PMID: 37692967 PMCID: PMC10492070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/31/2023] [Indexed: 09/12/2023]
Abstract
Drug repurposing, also known as drug repositioning, entails the application of pre-approved or formerly assessed drugs having potentially functional therapeutic amalgams for curing various disorders or disease conditions distinctive from their original remedial indication. It has surfaced as a substitute for the development of drugs for treating cancer, cardiovascular diseases, neurodegenerative disorders, and various infectious diseases like Covid-19. Although the earlier lines of findings in this area were serendipitous, recent advancements are based on patient centered approaches following systematic, translational, drug targeting practices that explore pathophysiological ailment mechanisms. The presence of definite information and numerous records with respect to beneficial properties, harmfulness, and pharmacologic characteristics of repurposed drugs increase the chances of approval in the clinical trial stages. The last few years have showcased the successful emergence of repurposed drug immunotherapy in treating various diseases. In this light, the present review emphasises on incorporation of drug repositioning with Immunotherapy targeted for several disorders.
Collapse
Affiliation(s)
| | - Anuradha Tyagi
- Department of cBRN, Institute of Nuclear Medicine and Allied ScienceDelhi 110054, India
| | - Chaitenya Verma
- Department of Pathology, Wexner Medical Center, Ohio State UniversityColumbus, Ohio 43201, USA
| | - Kanti Prakash Sharma
- Department of Nutrition Biology, Central University of HaryanaMahendragarh 123029, India
| | - Sekhu Ansari
- Division of Pathology, Cincinnati Children’s Hospital Medical CenterCincinnati, Ohio 45229, USA
| | - Indra Mani
- Department of Microbiology, Gargi College, University of DelhiNew Delhi 110049, India
| | | | - Pradeep Kumar Shukla
- Department of Biological Sciences, Faculty of Science, Sam Higginbottom University of Agriculture, Technology of SciencePrayagraj 211007, UP, India
| | - Antresh Kumar
- Department of Biochemistry, Central University of HaryanaMahendergarh 123031, Haryana, India
| | - Vinay Kumar
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical CenterColumbus, Ohio 43210, USA
| |
Collapse
|
28
|
Wei H, Xiao X, Zeng S, Liu Y, Liu X, Zeng T, Xu P, Xia W, Guo L, Hong S, Lv W, Chen Y, Xu R. Alterations in factors associated with diabetic retinopathy combined with thrombosis: A review. Medicine (Baltimore) 2023; 102:e34373. [PMID: 37543800 PMCID: PMC10403020 DOI: 10.1097/md.0000000000034373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/07/2023] Open
Abstract
Diabetic retinopathy (DR) is one of the most common and serious microvascular complications of diabetes mellitus, the incidence of which has been increasing annually, and it is the main cause of vision loss in diabetic patients and a common cause of blindness. It is now found that thrombosis plays a crucial role in the disease progression in DR patients, and the final vision loss in DR may be related to the occurrence of thrombosis in the retinal vessels, which is dominated by abnormal endothelial cell function, together with platelet dysfunction, imbalance of coagulation and fibrinolytic function, and related alterations of inflammatory factors leading to the main cause of thrombotic disease in DR patients. In this review, we examine the role between DR and thrombosis and the association of each factor, including endothelial dysfunction; platelet dysfunction; coagulation-fibrinolytic imbalance; and alterations in inflammatory factors.
Collapse
Affiliation(s)
- Haiyan Wei
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - Xiaoping Xiao
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, P.R. China
- Provincial Key Laboratory of Low-Carbon Solid Waste Recycling, Gannan Normal University, Ganzhou, P. R. China
| | - Shuqin Zeng
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - Ye Liu
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - Xiaofang Liu
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - Tianyu Zeng
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - Pengxiang Xu
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - Wenyan Xia
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - Li Guo
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - Shihua Hong
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - Weiming Lv
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - Yijian Chen
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - Rong Xu
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| |
Collapse
|
29
|
Koudriavtseva T, Lorenzano S, Cellerino M, Truglio M, Fiorelli M, Lapucci C, D’Agosto G, Conti L, Stefanile A, Zannino S, Filippi MM, Cortese A, Piantadosi C, Maschio M, Maialetti A, Galiè E, Salvetti M, Inglese M. Tissue factor as a potential coagulative/vascular marker in relapsing-remitting multiple sclerosis. Front Immunol 2023; 14:1226616. [PMID: 37583699 PMCID: PMC10424925 DOI: 10.3389/fimmu.2023.1226616] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 07/10/2023] [Indexed: 08/17/2023] Open
Abstract
Objectives Recent studies supported coagulation involvement in multiple sclerosis, an inflammatory-demyelinating and degenerative disease of the central nervous system. The main objectives of this observational study were to identify the most specific pro-coagulative/vascular factors for multiple sclerosis pathogenesis and to correlate them with brain hemodynamic abnormalities. Methods We compared i) serum/plasma levels of complement(C)/coagulation/vascular factors, viral/microbiological assays, fat-soluble vitamins and lymphocyte count among people with multiple sclerosis sampled in a clinical remission (n=30; 23F/7M, 40 ± 8.14 years) or a relapse (n=30; 24F/6M, age 41 ± 10.74 years) and age/sex-matched controls (n=30; 23F/7M, 40 ± 8.38 years); ii) brain hemodynamic metrics at dynamic susceptibility contrast-enhanced 3T-MRI during relapse and remission, and iii) laboratory data with MRI perfusion metrics and clinical features of people with multiple sclerosis. Two models by Partial Least Squares Discriminant Analysis were performed using two groups as input: (1) multiple sclerosis vs. controls, and (2) relapsing vs. remitting multiple sclerosis. Results Compared to controls, multiple sclerosis patients had a higher Body-Mass-Index, Protein-C and activated-C9; and a lower activated-C4. Levels of Tissue-Factor, Tie-2 and P-Selectin/CD62P were lower in relapse compared to remission and HC, whereas Angiopoietin-I was higher in relapsing vs. remitting multiple sclerosis. A lower number of total lymphocytes was found in relapsing multiple sclerosis vs. remitting multiple sclerosis and controls. Cerebral-Blood-Volume was lower in normal-appearing white matter and left caudatum while Cerebral-Blood-Flow was inferior in bilateral putamen in relapsing versus remitting multiple sclerosis. The mean-transit-time of gadolinium-enhancing lesions negatively correlated with Tissue-Factor. The top-5 discriminating variables for model (1) were: EBV-EBNA-1 IgG, Body-Mass-Index, Protein-C, activated-C4 and Tissue-Factor whereas for model (2) were: Tissue-Factor, Angiopoietin-I, MCHC, Vitamin A and T-CD3. Conclusion Tissue-factor was one of the top-5 variables in the models discriminating either multiple sclerosis from controls or multiple sclerosis relapse from remission and correlated with mean-transit-time of gadolinium-enhancing lesions. Tissue-factor appears a promising pro-coagulative/vascular biomarker and a possible therapeutic target in relapsing-remitting multiple sclerosis. Clinical trial registration ClinicalTrials.gov, identifier NCT04380220.
Collapse
Affiliation(s)
- Tatiana Koudriavtseva
- Medical Direction, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
- Department of Clinical Experimental Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Regina Elena National Cancer Institute, Istituti Fisioterapici Ospitalieri (IFO), Rome, Italy
| | - Svetlana Lorenzano
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | - Maria Cellerino
- Department of Clinical Experimental Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Regina Elena National Cancer Institute, Istituti Fisioterapici Ospitalieri (IFO), Rome, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Mauro Truglio
- Clinical Pathology and Cancer Biobank, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Marco Fiorelli
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | - Caterina Lapucci
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Giovanna D’Agosto
- Clinical Pathology and Microbiology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Gallicano Dermatological Institute, Rome, Italy
| | - Laura Conti
- Clinical Pathology and Cancer Biobank, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Annunziata Stefanile
- Clinical Pathology and Cancer Biobank, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Silvana Zannino
- Department of Clinical Experimental Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Regina Elena National Cancer Institute, Istituti Fisioterapici Ospitalieri (IFO), Rome, Italy
| | | | - Antonio Cortese
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | - Carlo Piantadosi
- Unità Operativa Complessa (UOC) Neurology, San Giovanni-Addolorata Hospital, Rome, Italy
| | - Marta Maschio
- Department of Clinical Experimental Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Regina Elena National Cancer Institute, Istituti Fisioterapici Ospitalieri (IFO), Rome, Italy
| | - Andrea Maialetti
- Department of Clinical Experimental Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Regina Elena National Cancer Institute, Istituti Fisioterapici Ospitalieri (IFO), Rome, Italy
| | - Edvina Galiè
- Department of Clinical Experimental Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Regina Elena National Cancer Institute, Istituti Fisioterapici Ospitalieri (IFO), Rome, Italy
| | - Marco Salvetti
- Department of Neuroscience Mental Health and Sensory Organs (NEMOS), Sapienza University, Rome, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy
| | - Matilde Inglese
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- Department of Neurology, Mount Sinai Hospital, New York, NY, United States
| |
Collapse
|
30
|
Lechuga GC, Morel CM, De-Simone SG. Hematological alterations associated with long COVID-19. Front Physiol 2023; 14:1203472. [PMID: 37565145 PMCID: PMC10411895 DOI: 10.3389/fphys.2023.1203472] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 07/10/2023] [Indexed: 08/12/2023] Open
Abstract
Long COVID-19 is a condition characterized by persistent symptoms lasting beyond the acute phase of COVID-19. Long COVID-19 produces diverse symptomatology and can impact organs and systems, including the hematological system. Several studies have reported, in COVID-19 patients, hematological abnormalities. Most of these alterations are associated with a higher risk of severe disease and poor outcomes. This literature review identified studies reporting hematological parameters in individuals with Long COVID-19. Findings suggest that Long COVID-19 is associated with a range of sustained hematological alterations, including alterations in red blood cells, anemia, lymphopenia, and elevated levels of inflammatory markers such as ferritin, D-dimer, and IL-6. These alterations may contribute to a better understanding of the pathophysiology of Long COVID-19 and its associated symptoms. However, further research is needed to elucidate the underlying mechanisms and potential treatments for these hematological changes in individuals with Long COVID-19.
Collapse
Affiliation(s)
- Guilherme C. Lechuga
- Center for Technological Development in Health (CDTS)/ National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
- Laboratory of Epidemiology and Molecular Systematics (LESM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
- Laboratory of Cellular Ultrastructure, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
| | - Carlos M. Morel
- Center for Technological Development in Health (CDTS)/ National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
| | - Salvatore Giovanni De-Simone
- Center for Technological Development in Health (CDTS)/ National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
- Laboratory of Epidemiology and Molecular Systematics (LESM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
- Post-Graduation Program in Science and Biotechnology, Department of Molecular and Cellular Biology, Biology Institute, Federal Fluminense University, Niterói, Brazil
| |
Collapse
|
31
|
Nabi AHMN, Ebihara A, Shekhar HU. Impacts of SARS-CoV-2 on diabetes mellitus: A pre and post pandemic evaluation. World J Virol 2023; 12:151-171. [PMID: 37396707 PMCID: PMC10311579 DOI: 10.5501/wjv.v12.i3.151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/24/2023] [Accepted: 04/13/2023] [Indexed: 06/21/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by the novel beta coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) crippled the whole world and has resulted in large number of morbidity and mortality. The origin of the SARS-CoV-2 is still disputed. The risk of infection with SARS-CoV-2 is dependent on several risk factors as observed in many studies. The severity of the disease depends on many factors including the viral strain, host immunogenetics, environmental factors, host genetics, host nutritional status and presence of comorbidities like hypertension, diabetes, Chronic Obstructive Pulmonary Disease, cardiovascular disease, renal impairment. Diabetes is a metabolic disorder mainly characterized by hyperglycemia. Diabetic individuals are intrinsically prone to infections. SARS-CoV-2 infection in patients with diabetes result in β-cell damage and cytokine storm. Damage to the cells impairs the equilibrium of glucose, leading to hyperglycemia. The ensuing cytokine storm causes insulin resistance, especially in the muscles and liver, which also causes a hyperglycemic state. All of these increase the severity of COVID-19. Genetics also play pivotal role in disease pathogenesis. This review article focuses from the probable sources of coronaviruses and SARS-CoV-2 to its impacts on individuals with diabetes and host genetics in pre- and post-pandemic era.
Collapse
Affiliation(s)
- A H M Nurun Nabi
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka 1000, Bangladesh
| | - Akio Ebihara
- Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193, Japan
| | - Hossain Uddin Shekhar
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka 1000, Bangladesh
| |
Collapse
|
32
|
Konarska-Bajda K, Ceranowicz P, Cieszkowski J, Ginter G, Stempniewicz A, Gałązka K, Kuśnierz-Cabala B, Dumnicka P, Bonior J, Warzecha Z. Administration of Warfarin Inhibits the Development of Cerulein-Induced Edematous Acute Pancreatitis in Rats. Biomolecules 2023; 13:948. [PMID: 37371528 DOI: 10.3390/biom13060948] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/19/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
Acute pancreatitis (AP) is a severe disease with high morbidity and mortality in which inflammation and coagulation play crucial roles. The development of inflammation leads to vascular injury, endothelium and leukocytes stimulation, and an increased level of tissue factor, which results in the activation of the coagulation process. For this reason, anticoagulants may be considered as a therapeutic option in AP. Previous studies have shown that pretreatment with heparin, low-molecular-weight heparin (LMWH), or acenocoumarol inhibits the development of AP. The aim of the present study was to check if pretreatment with warfarin affects the development of edematous pancreatitis evoked by cerulein. Warfarin (90, 180, or 270 µg/kg/dose) or saline were administered intragastrically once a day for 7 days consecutively before the induction of AP. AP was evoked by the intraperitoneal administration of cerulein. The pre-administration of warfarin at doses of 90 or 180 µg/kg/dose reduced the histological signs of pancreatic damage in animals with the induction of AP. Additionally, other parameters of AP, such as an increase in the serum activity of lipase and amylase, the plasma concentration of D-dimer, and interleukin-1β, were decreased. In addition, pretreatment with warfarin administered at doses of 90 or 180 µg/kg/dose reversed the limitation of pancreatic blood flow evoked by AP development. Warfarin administered at a dose of 270 µg/kg/dose did not exhibit a preventive effect in cerulein-induced AP. Conclusion: Pretreatment with low doses of warfarin inhibits the development of AP evoked by the intraperitoneal administration of cerulein.
Collapse
Affiliation(s)
- Katarzyna Konarska-Bajda
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Kraków, Poland
- Department of Pediatric Cardiology, University Children's Hospital in Cracow, 30-663 Kraków, Poland
| | - Piotr Ceranowicz
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Kraków, Poland
| | - Jakub Cieszkowski
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Kraków, Poland
| | - Grzegorz Ginter
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Kraków, Poland
| | - Agnieszka Stempniewicz
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Kraków, Poland
| | - Krystyna Gałązka
- Department of Pathology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Kraków, Poland
| | - Beata Kuśnierz-Cabala
- Chair of Clinical Biochemistry/Chair of Medical Biochemistry, Jagiellonian University Medical College, 31-034 Kraków, Poland
| | - Paulina Dumnicka
- Chair of Clinical Biochemistry/Chair of Medical Biochemistry, Jagiellonian University Medical College, 31-034 Kraków, Poland
| | - Joanna Bonior
- Department of Medical Physiology, Faculty of Health Sciences, Jagiellonian University Medical College, 31-126 Kraków, Poland
| | - Zygmunt Warzecha
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Kraków, Poland
| |
Collapse
|
33
|
Ragnoli B, Da Re B, Galantino A, Kette S, Salotti A, Malerba M. Interrelationship between COVID-19 and Coagulopathy: Pathophysiological and Clinical Evidence. Int J Mol Sci 2023; 24:ijms24108945. [PMID: 37240292 DOI: 10.3390/ijms24108945] [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: 04/25/2023] [Revised: 05/14/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Since the first description of COVID-19 infection, among clinical manifestations of the disease, including fever, dyspnea, cough, and fatigue, it was observed a high incidence of thromboembolic events potentially evolving towards acute respiratory distress syndrome (ARDS) and COVID-19-associated-coagulopathy (CAC). The hypercoagulation state is based on an interaction between thrombosis and inflammation. The so-called CAC represents a key aspect in the genesis of organ damage from SARS-CoV-2. The prothrombotic status of COVID-19 can be explained by the increase in coagulation levels of D-dimer, lymphocytes, fibrinogen, interleukin 6 (IL-6), and prothrombin time. Several mechanisms have been hypothesized to explain this hypercoagulable process such as inflammatory cytokine storm, platelet activation, endothelial dysfunction, and stasis for a long time. The purpose of this narrative review is to provide an overview of the current knowledge on the pathogenic mechanisms of coagulopathy that may characterize COVID-19 infection and inform on new areas of research. New vascular therapeutic strategies are also reviewed.
Collapse
Affiliation(s)
| | - Beatrice Da Re
- Respiratory Unit, Sant'Andrea Hospital, 13100 Vercelli, Italy
| | | | - Stefano Kette
- Respiratory Unit, Sant'Andrea Hospital, 13100 Vercelli, Italy
| | - Andrea Salotti
- Respiratory Unit, Sant'Andrea Hospital, 13100 Vercelli, Italy
| | - Mario Malerba
- Respiratory Unit, Sant'Andrea Hospital, 13100 Vercelli, Italy
- Department of Traslational Medicine, University of Eastern Piedmont (UPO), 28100 Novara, Italy
| |
Collapse
|
34
|
Barbalho SM, Minniti G, Miola VFB, Haber JFDS, Bueno PCDS, de Argollo Haber LS, Girio RSJ, Detregiachi CRP, Dall'Antonia CT, Rodrigues VD, Nicolau CCT, Catharin VMCS, Araújo AC, Laurindo LF. Organokines in COVID-19: A Systematic Review. Cells 2023; 12:1349. [PMID: 37408184 DOI: 10.3390/cells12101349] [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: 02/22/2023] [Revised: 05/03/2023] [Accepted: 05/06/2023] [Indexed: 07/07/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a viral infection caused by SARS-CoV-2 that induces a generalized inflammatory state. Organokines (adipokines, osteokines, myokines, hepatokines, and cardiokines) can produce beneficial or harmful effects in this condition. This study aimed to systematically review the role of organokines on COVID-19. PubMed, Embase, Google Scholar, and Cochrane databases were searched, the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines were followed, and 37 studies were selected, comprising more than 2700 individuals infected with the virus. Among COVID-19 patients, organokines have been associated with endothelial dysfunction and multiple organ failure due to augmented cytokines and increased SARS-CoV-2 viremia. Changes in the pattern of organokines secretion can directly or indirectly contribute to aggravating the infection, promoting immune response alterations, and predicting the disease progression. These molecules have the potential to be used as adjuvant biomarkers to predict the severity of the illness and severe outcomes.
Collapse
Affiliation(s)
- Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Avenida Castro Alves, 62, Marília 17500-000, SP, Brazil
| | - Giulia Minniti
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
| | - Vitor Fernando Bordin Miola
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
| | - Jesselina Francisco Dos Santos Haber
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
- Centro Interdisciplinar em Diabetes (CENID), School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
| | - Patrícia Cincotto Dos Santos Bueno
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
- Department of Animal Sciences, School of Veterinary Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
| | - Luiza Santos de Argollo Haber
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
| | - Raul S J Girio
- Department of Animal Sciences, School of Veterinary Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
| | - Cláudia Rucco Penteado Detregiachi
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
- Department of Biochemistry and Pharmacology, Faculdade de Medicina de Marília (FAMEMA), School of Medicine, Avenida Monte Carmelo, 800, Marília 17519-030, SP, Brazil
| | - Camila Tiveron Dall'Antonia
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
| | - Victória Dogani Rodrigues
- Department of Biochemistry and Pharmacology, Faculdade de Medicina de Marília (FAMEMA), School of Medicine, Avenida Monte Carmelo, 800, Marília 17519-030, SP, Brazil
| | - Claudia C T Nicolau
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Avenida Castro Alves, 62, Marília 17500-000, SP, Brazil
| | - Virginia Maria Cavallari Strozze Catharin
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
| | - Adriano Cressoni Araújo
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
| | - Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
- Department of Biochemistry and Pharmacology, Faculdade de Medicina de Marília (FAMEMA), School of Medicine, Avenida Monte Carmelo, 800, Marília 17519-030, SP, Brazil
| |
Collapse
|
35
|
Shen J, Fan J, Zhao Y, Jiang D, Niu Z, Zhang Z, Cao G. Innate and adaptive immunity to SARS-CoV-2 and predisposing factors. Front Immunol 2023; 14:1159326. [PMID: 37228604 PMCID: PMC10203583 DOI: 10.3389/fimmu.2023.1159326] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 04/27/2023] [Indexed: 05/27/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus (SARS-CoV-2), has affected all countries worldwide. Although some symptoms are relatively mild, others are still associated with severe and even fatal clinical outcomes. Innate and adaptive immunity are important for the control of SARS-CoV-2 infections, whereas a comprehensive characterization of the innate and adaptive immune response to COVID-19 is still lacking and the mechanisms underlying immune pathogenesis and host predisposing factors are still a matter of scientific debate. Here, the specific functions and kinetics of innate and adaptive immunity involved in SARS-CoV-2 recognition and resultant pathogenesis are discussed, as well as their immune memory for vaccinations, viral-mediated immune evasion, and the current and future immunotherapeutic agents. We also highlight host factors that contribute to infection, which may deepen the understanding of viral pathogenesis and help identify targeted therapies that attenuate severe disease and infection.
Collapse
Affiliation(s)
- Jiaying Shen
- Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Junyan Fan
- Department of Epidemiology, Shanghai Key Laboratory of Medical Bioprotection, Key Laboratory of Biological Defense, Ministry of Education, Second Military Medical University, Shanghai, China
| | - Yue Zhao
- Department of Epidemiology, Shanghai Key Laboratory of Medical Bioprotection, Key Laboratory of Biological Defense, Ministry of Education, Second Military Medical University, Shanghai, China
| | - Doming Jiang
- Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Zheyun Niu
- Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Zihan Zhang
- Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Guangwen Cao
- Tongji University School of Medicine, Tongji University, Shanghai, China
- Department of Epidemiology, Shanghai Key Laboratory of Medical Bioprotection, Key Laboratory of Biological Defense, Ministry of Education, Second Military Medical University, Shanghai, China
| |
Collapse
|
36
|
Carvalho-Sauer R, Flores-Ortiz R, Costa MDCN, Teixeira MG, Saavedra R, Niag M, Paixao ES. Fetal death as an outcome of acute respiratory distress in pregnancy, during the COVID-19 pandemic: a population-based cohort study in Bahia, Brazil. BMC Pregnancy Childbirth 2023; 23:320. [PMID: 37147605 PMCID: PMC10161155 DOI: 10.1186/s12884-023-05601-w] [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: 01/28/2023] [Accepted: 04/12/2023] [Indexed: 05/07/2023] Open
Abstract
BACKGROUND Fetal loss is one of the most serious adverse outcomes of pregnancy. Since the onset of the COVID-19 pandemic, Brazil has recorded an unprecedented number of hospitalizations of pregnant women due to acute respiratory distress (ARD), thereby, we aimed to assess the risk of fetal deaths associated to ARD during pregnancy in Bahia state, Brazil, in the context of the COVID-19 pandemic. METHODS This is an observational population-based retrospective cohort study, developed with women at or after 20 weeks of pregnancy, residents in Bahia, Brazil. Women who had acute respiratory distress (ARD) in pregnancy during the COVID-19 pandemic (Jan 2020 to Jun 2021) were considered 'exposed'. Women who did not have ARD in pregnancy, and whose pregnancy occurred before the onset of the COVID-19 pandemic (Jan 2019 to Dec 2019) were considered 'non-exposed'. The main outcome was fetal death. We linked administrative data (under mandatory registration) on live births, fetal deaths, and acute respiratory syndrome, using a probabilistic linkage method, and analyzed them with multivariable logistic regression models. RESULTS 200,979 pregnant women participated in this study, 765 exposed and 200,214 unexposed. We found four times higher chance of fetal death in women with ARD during pregnancy, of all etiologies (adjusted odds ratio [aOR] 4.06 confidence interval [CI] 95% 2.66; 6.21), and due to SARS-CoV-2 (aOR 4.45 CI 95% 2.41; 8.20). The risk of fetal death increased more when ARD in pregnancy was accompanied by vaginal delivery (aOR 7.06 CI 95% 4.21; 11.83), or admission to Intensive Care Unit (aOR 8.79 CI 95% 4.96; 15.58), or use of invasive mechanical ventilation (aOR 21.22 CI 95% 9.93; 45.36). CONCLUSION Our findings can contribute to expanding the understanding of health professionals and managers about the harmful effects of SARS-CoV-2 on maternal-fetal health and alerts the need to prioritize pregnant women in preventive actions against SARS-CoV-2 and other respiratory viruses. It also suggests that pregnant women, infected with SARS-CoV-2, need to be monitored to prevent complications of ARD, including a careful assessment of the risks and benefits of early delivery to prevent fetal death.
Collapse
Affiliation(s)
- Rita Carvalho-Sauer
- Bahia State Health Department, Núcleo Regional de Saúde Leste, Avenida Esperança, 406. Maria Preta. Santo Antônio de Jesus., 44435-500 Bahia, Brazil
- Institute of Collective Health, Federal University of Bahia, Bahia, Brazil
| | - Renzo Flores-Ortiz
- Center for Data and Knowledge Integration for Health (CIDACS), Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Bahia, Brazil
| | | | | | - Ramon Saavedra
- Institute of Collective Health, Federal University of Bahia, Bahia, Brazil
| | - Marla Niag
- School of Medicine, Federal University of Recôncavo of Bahia, Bahia, Brazil
| | - Enny S. Paixao
- London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
37
|
Pan C, Sun X, Song J, Yu C, Guo Y, Wang S, Gao R, Ning F, Pang Z, Chen Z, Li L. The Prospective Associations of Egg Consumption with the Risk of Total Cerebrovascular Disease Morbidity among Chinese Adults. Nutrients 2023; 15:nu15081808. [PMID: 37111029 PMCID: PMC10142563 DOI: 10.3390/nu15081808] [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: 03/16/2023] [Revised: 04/02/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Studies investigating the relationship between egg consumption and the risk of cerebrovascular disease (CED) have yielded inconsistent results. This study evaluated the association between egg consumption and the risk of CED among Chinese adults. METHODS Data were obtained from China Kadoorie Biobank, Qingdao. A computerised questionnaire was used to collect information regarding egg consumption frequency. CED events were tracked through linkage with the Disease Surveillance Point System and the new national health insurance databases. Cox proportional hazards regression analyses were used to evaluate associations between egg consumption and CED risk controlling for potential confounders. RESULTS After a median follow-up of 9.2 years, 865 and 1083 CED events among men and women, respectively, were documented. More than 50% of participants consumed eggs daily with an average age of 52.0 (10.4) years at baseline. No association between egg consumption and CED were identified in the whole cohort and women. However, a 28% lower risk of CED was observed in those who consumed eggs at a higher frequency (HR = 0.72, 95% CI: 0.55-0.95) and a significant trend for the association (p for trend = 0.012) in a multivariable model in men. CONCLUSION Higher frequency of egg consumption was associated with a lower risk of total CED events among men but not women in Chinese adults. The beneficial effect on women warrants further investigations.
Collapse
Affiliation(s)
- Chi Pan
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Xiaohui Sun
- Qingdao Municipality Center for Disease Control and Prevention, Qingdao 266033, China
- Qingdao Institute of Preventive Medicine, Qingdao 266033, China
| | - Jiahui Song
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing 100191, China
| | - Yu Guo
- Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Shaojie Wang
- Qingdao Municipality Center for Disease Control and Prevention, Qingdao 266033, China
- Qingdao Institute of Preventive Medicine, Qingdao 266033, China
| | - Ruqin Gao
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Feng Ning
- Qingdao Municipality Center for Disease Control and Prevention, Qingdao 266033, China
- Qingdao Institute of Preventive Medicine, Qingdao 266033, China
| | - Zengchang Pang
- Qingdao Municipality Center for Disease Control and Prevention, Qingdao 266033, China
- Qingdao Institute of Preventive Medicine, Qingdao 266033, China
| | - Zhengming Chen
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing 100191, China
| |
Collapse
|
38
|
Akácsos-Szász OZ, Pál S, Nyulas KI, Nemes-Nagy E, Fárr AM, Dénes L, Szilveszter M, Bán EG, Tilinca MC, Simon-Szabó Z. Pathways of Coagulopathy and Inflammatory Response in SARS-CoV-2 Infection among Type 2 Diabetic Patients. Int J Mol Sci 2023; 24:4319. [PMID: 36901751 PMCID: PMC10001503 DOI: 10.3390/ijms24054319] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 02/14/2023] [Accepted: 02/18/2023] [Indexed: 02/24/2023] Open
Abstract
Chronic inflammation and endothelium dysfunction are present in diabetic patients. COVID-19 has a high mortality rate in association with diabetes, partially due to the development of thromboembolic events in the context of coronavirus infection. The purpose of this review is to present the most important underlying pathomechanisms in the development of COVID-19-related coagulopathy in diabetic patients. The methodology consisted of data collection and synthesis from the recent scientific literature by accessing different databases (Cochrane, PubMed, Embase). The main results are the comprehensive and detailed presentation of the very complex interrelations between different factors and pathways involved in the development of arteriopathy and thrombosis in COVID-19-infected diabetic patients. Several genetic and metabolic factors influence the course of COVID-19 within the background of diabetes mellitus. Extensive knowledge of the underlying pathomechanisms of SARS-CoV-2-related vasculopathy and coagulopathy in diabetic subjects contributes to a better understanding of the manifestations in this highly vulnerable group of patients; thus, they can benefit from a modern, more efficient approach regarding diagnostic and therapeutic management.
Collapse
Affiliation(s)
- Orsolya-Zsuzsa Akácsos-Szász
- Doctoral School, Faculty of Medicine, George Emil Palade University of Medicine Pharmacy, Science, and Technology of Târgu Mureş, 540142 Târgu-Mureș, Romania
| | - Sándor Pál
- Department of Transfusion Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Kinga-Ilona Nyulas
- Doctoral School, Faculty of Medicine, George Emil Palade University of Medicine Pharmacy, Science, and Technology of Târgu Mureş, 540142 Târgu-Mureș, Romania
| | - Enikő Nemes-Nagy
- Department of Chemistry and Medical Biochemistry, Faculty of Medicine in English, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureş, 540142 Târgu-Mureș, Romania
| | - Ana-Maria Fárr
- Department of Pathophysiology, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureş, 540142 Târgu-Mureș, Romania
| | - Lóránd Dénes
- Department of Anatomy, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureş, 540142 Târgu-Mureș, Romania
| | - Mónika Szilveszter
- Clinic of Plastic Surgery, Mureș County Emergency Hospital, 540136 Târgu Mureș, Romania
| | - Erika-Gyöngyi Bán
- Department of Pharmacology, Faculty of Medicine in English, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureş, 540142 Târgu-Mureș, Romania
| | - Mariana Cornelia Tilinca
- Department of Internal Medicine I, Faculty of Medicine in English, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureş, 540142 Târgu-Mureș, Romania
| | - Zsuzsánna Simon-Szabó
- Department of Pathophysiology, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureş, 540142 Târgu-Mureș, Romania
| |
Collapse
|
39
|
Silva MJA, Ribeiro LR, Gouveia MIM, Marcelino BDR, dos Santos CS, Lima KVB, Lima LNGC. Hyperinflammatory Response in COVID-19: A Systematic Review. Viruses 2023; 15:553. [PMID: 36851766 PMCID: PMC9962879 DOI: 10.3390/v15020553] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/12/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023] Open
Abstract
COVID-19 is a multisystemic disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The immunopathogenic conditions of the hyperinflammatory response that cause systemic inflammation are extremely linked to its severity. This research sought to review the immunopathological elements that contribute to its progression. This is a systematic review using the PUBMED, LILACS, MEDLINE, and SCIELO databases using articles between May 2020 and July 2022 with the following search terms in conjunction with "AND": "SARS-CoV-2"; "COVID-19"; "ARDS" and "Cytokine Storm". The quality appraisal and risk of bias were assessed by the JBI checklists and the Cochrane Collaboration's RoB 2.0 and ROBINS-I tools, respectively, and the risk of bias for in vitro studies by a pre-defined standard in the literature. The search resulted in 39 articles. The main actors in this response denote SARS-CoV-2 Spike proteins, cellular proteases, leukocytes, cytokines, and proteolytic cascades. The "cytokine storm" itself brings several complications to the host through cytokines such as IL-6 and chemokines (such as CCL2), which influence tissue inflammation through apoptosis and pyroptosis. The hyperinflammatory response causes several unfavorable outcomes in patients, and systemic inflammation caused largely by the dysregulation of the immune response should be controlled for their recovery.
Collapse
Affiliation(s)
- Marcos Jessé Abrahão Silva
- Graduate Program in Epidemiology and Health Surveillance (PPGEVS), Evandro Chagas Institute (IEC), Ananindeua 67030-000, PA, Brazil
- Bacteriology and Mycology Section, Evandro Chagas Institute (IEC), Ananindeua 67030-000, PA, Brazil
| | - Layana Rufino Ribeiro
- Bacteriology and Mycology Section, Evandro Chagas Institute (IEC), Ananindeua 67030-000, PA, Brazil
| | | | - Beatriz dos Reis Marcelino
- Graduate Program in Parasitic Biology in the Amazon (PPGBPA), University of Pará State (UEPA), Belém 66087-670, PA, Brazil
| | - Carolynne Silva dos Santos
- Federal Institute of Education, Science, and Technology of Pará (IFPA), Abaetetuba 68440-000, PA, Brazil
| | | | - Luana Nepomuceno Gondim Costa Lima
- Graduate Program in Epidemiology and Health Surveillance (PPGEVS), Evandro Chagas Institute (IEC), Ananindeua 67030-000, PA, Brazil
- Bacteriology and Mycology Section, Evandro Chagas Institute (IEC), Ananindeua 67030-000, PA, Brazil
| |
Collapse
|
40
|
Zhu G, Modepalli S, Anand M, Li H. Computational modeling of hypercoagulability in COVID-19. Comput Methods Biomech Biomed Engin 2023; 26:338-349. [PMID: 36154346 DOI: 10.1080/10255842.2022.2124858] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has infected more than 100 million people worldwide and claimed millions of lives. While the leading cause of mortality in COVID-19 patients is the hypoxic respiratory failure from acute respiratory distress syndrome, there is accumulating evidence that shows excessive coagulation also increases the fatalities in COVID-19. Thus, there is a pressing demand to understand the association between COVID-19-induced hypercoagulability and the extent of formation of undesired blood clots. Mathematical modeling of coagulation has been used as an important tool to identify novel reaction mechanisms and to identify targets for new drugs. Here, we employ the coagulation factor data of COVID-19 patients reported from published studies as inputs for two mathematical models of coagulation to identify how the concentrations of coagulation factors change in these patients. Our simulation results show that while the levels of many of the abnormal coagulation factors measured in COVID-19 patients promote the generation of thrombin and fibrin, two key components of blood clots, the increased level of fibrinogen and then the reduced level of antithrombin are the factors most responsible for boosting the level of fibrin and thrombin, respectively. Altogether, our study demonstrates the potential of mathematical modeling to identify coagulation factors responsible for the increased clot formation in COVID-19 patients where clinical data is scarce.
Collapse
Affiliation(s)
- Ge Zhu
- Center for Biomedical Engineering, Brown University, Providence, USA
| | | | - Mohan Anand
- Department of Chemical Engineering, Indian Institute of Technology Hyderabad, Hyderabad, India
| | - He Li
- School of Chemical, Materials & Biomedical Engineering, University of Georgia, Athens, USA
| |
Collapse
|
41
|
Varlas VN, Borș RG, Plotogea M, Iordache M, Mehedințu C, Cîrstoiu MM. Thromboprophylaxis in Pregnant Women with COVID-19: An Unsolved Issue. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:1949. [PMID: 36767320 PMCID: PMC9915713 DOI: 10.3390/ijerph20031949] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/15/2023] [Accepted: 01/18/2023] [Indexed: 06/18/2023]
Abstract
SARS-CoV-2 infection in pregnant women is of growing interest due to controversy over the use of antiplatelet and/or anticoagulant drugs during pregnancy and postpartum. Pregnant women are susceptible to develop severe forms of viral infections due to pregnancy-related immune alterations, changes in lung functions, and hypercoagulability. The association of pregnancy with SARS-CoV-2 infection can cause an increased incidence of thrombotic complications, especially in the case of patients with some genetic variants that favor inflammation and thrombosis. Compared to the general population, pregnant women may be at increased risk of thrombotic complications related to COVID-19. The lack of extensive clinical trials on thromboprophylaxis and extrapolating data from non-pregnant patients lead to major discrepancies in treating pregnant women with COVID-19. Currently, a multidisciplinary team should determine the dose and duration of prophylactic anticoagulant therapy for these patients, depending on the disease severity, the course of pregnancy, and the estimated due date. This narrative review aims to evaluate the protective effect of thromboprophylaxis in pregnant women with COVID-19. It is unknown at this time whether antiplatelet or anticoagulant therapy initiated at the beginning of pregnancy for various diseases (preeclampsia, intrauterine growth restriction, thrombophilia) offers a degree of protection. The optimal scheme for thromboprophylaxis in pregnant women with COVID-19 must be carefully established through an individualized decision concerning gestational age and the severity of the infection.
Collapse
Affiliation(s)
- Valentin Nicolae Varlas
- Department of Obstetrics and Gynecology, Filantropia Clinical Hospital, 011171 Bucharest, Romania
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 37 Dionisie Lupu St., 050451 Bucharest, Romania
| | - Roxana Georgiana Borș
- Department of Obstetrics and Gynecology, Filantropia Clinical Hospital, 011171 Bucharest, Romania
| | - Mihaela Plotogea
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 37 Dionisie Lupu St., 050451 Bucharest, Romania
- Department of Obstetrics and Gynecology, Nicolae Malaxa Clinical Hospital Bucharest, 022441 Bucharest, Romania
| | - Madalina Iordache
- Doctoral School, “Carol Davila”, University of Medicine and Pharmacy, 4192910 Bucharest, Romania
| | - Claudia Mehedințu
- Department of Obstetrics and Gynecology, Filantropia Clinical Hospital, 011171 Bucharest, Romania
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 37 Dionisie Lupu St., 050451 Bucharest, Romania
| | - Monica Mihaela Cîrstoiu
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 37 Dionisie Lupu St., 050451 Bucharest, Romania
- Department of Obstetrics and Gynecology, University Emergency Hospital Bucharest, 050098 Bucharest, Romania
| |
Collapse
|
42
|
Dos Santos F, Li JB, Juocys N, Mazor R, Beretta L, Coufal NG, Lam MTY, Odish MF, Irigoyen MC, O’Donoghue AJ, Aletti F, Kistler EB. Plasma enzymatic activity, proteomics and peptidomics in COVID-19-induced sepsis: A novel approach for the analysis of hemostasis. Front Mol Biosci 2023; 9:1051471. [PMID: 36710882 PMCID: PMC9874325 DOI: 10.3389/fmolb.2022.1051471] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 12/20/2022] [Indexed: 01/13/2023] Open
Abstract
Introduction: Infection by SARS-CoV-2 and subsequent COVID-19 can cause viral sepsis. We investigated plasma protease activity patterns in COVID-19-induced sepsis with bacterial superinfection, as well as plasma proteomics and peptidomics in order to assess the possible implications of enhanced proteolysis on major protein systems (e.g., coagulation). Methods: Patients (=4) admitted to the intensive care units (ICUs) at the University of California, San Diego (UCSD) Medical Center with confirmed positive test for COVID-19 by real-time reverse transcription polymerase chain reaction (RT-PCR) were enrolled in a study approved by the UCSD Institutional Review Board (IRB# 190699, Protocol #20-0006). Informed consent was obtained for the collection of blood samples and de-identified use of the data. Blood samples were collected at multiple time points and analyzed to quantify a) the circulating proteome and peptidome by mass spectrometry; b) the aminopeptidase activity in plasma; and c) the endopeptidase activity in plasma using fluorogenic substrates that are cleaved by trypsin-like endopeptidases, specific clotting factors and plasmin. The one patient who died was diagnosed with bacterial superinfection on day 7 after beginning of the study. Results: Spikes in protease activity (factor VII, trypsin-like activity), and corresponding increases in the intensity of peptides derived by hydrolysis of plasma proteins, especially of fibrinogen degradation products and downregulation of endogenous protease inhibitors were detected on day 7 for the patient who died. The activity of the analyzed proteases was stable in survivors. Discussion: The combination of multiomics and enzymatic activity quantification enabled to i) hypothesize that elevated proteolysis occurs in COVID-19-induced septic shock with bacterial superinfection, and ii) provide additional insight into malfunctioning protease-mediated systems, such as hemostasis.
Collapse
Affiliation(s)
- Fernando Dos Santos
- Department of Anesthesiology, School of Medicine, University of California, San Diego, CA, United States
| | - Joyce B. Li
- Department of Bioengineering, University of California, San Diego, CA, United States
| | - Nathalia Juocys
- Department of Anesthesiology, School of Medicine, University of California, San Diego, CA, United States
- Instituto do Coração, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (InCor-FMUSP), São Paulo, Brazil
| | - Rafi Mazor
- Department of Anesthesiology, School of Medicine, University of California, San Diego, CA, United States
| | - Laura Beretta
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA, United States
| | - Nicole G. Coufal
- Department of Pediatrics, School of Medicine, University of California, San Diego, CA, United States
| | - Michael T. Y. Lam
- Department of Medicine, School of Medicine, University of California, San Diego, CA, United States
| | - Mazen F. Odish
- Department of Medicine, School of Medicine, University of California, San Diego, CA, United States
| | - Maria Claudia Irigoyen
- Instituto do Coração, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (InCor-FMUSP), São Paulo, Brazil
| | - Anthony J. O’Donoghue
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA, United States
| | - Federico Aletti
- Instituto de Ciência e Tecnologia, Universidade Federal de São Paulo, São Josê dos Campos, Brazil
| | - Erik B. Kistler
- Department of Anesthesiology, School of Medicine, University of California, San Diego, CA, United States
- Department of Anesthesiology and Critical Care, VA San Diego Healthcare System, San Diego, CA, United States
| |
Collapse
|
43
|
Khreefa Z, Barbier MT, Koksal AR, Love G, Del Valle L. Pathogenesis and Mechanisms of SARS-CoV-2 Infection in the Intestine, Liver, and Pancreas. Cells 2023; 12:cells12020262. [PMID: 36672197 PMCID: PMC9856332 DOI: 10.3390/cells12020262] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/30/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
The novel coronavirus, SARS-CoV-2, rapidly spread worldwide, causing an ongoing global pandemic. While the respiratory system is the most common site of infection, a significant number of reported cases indicate gastrointestinal (GI) involvement. GI symptoms include anorexia, abdominal pain, nausea, vomiting, and diarrhea. Although the mechanisms of GI pathogenesis are still being examined, viral components isolated from stool samples of infected patients suggest a potential fecal-oral transmission route. In addition, viral RNA has been detected in blood samples of infected patients, making hematologic dissemination of the virus a proposed route for GI involvement. Angiotensin-converting enzyme 2 (ACE2) receptors serve as the cellular entry mechanism for the virus, and these receptors are particularly abundant throughout the GI tract, making the intestine, liver, and pancreas potential extrapulmonary sites for infection and reservoirs sites for developing mutations and new variants that contribute to the uncontrolled spread of the disease and resistance to treatments. This transmission mechanism and the dysregulation of the immune system play a significant role in the profound inflammatory and coagulative cascades that contribute to the increased severity and risk of death in several COVID-19 patients. This article reviews various potential mechanisms of gastrointestinal, liver, and pancreatic injury.
Collapse
Affiliation(s)
- Zaid Khreefa
- Department of Pathology, School of Medicine, Louisiana State University Health School of Medicine, New Orleans, LA 70112, USA
| | - Mallory T. Barbier
- Louisiana Cancer Research Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Ali Riza Koksal
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Gordon Love
- Department of Pathology, School of Medicine, Louisiana State University Health School of Medicine, New Orleans, LA 70112, USA
| | - Luis Del Valle
- Department of Pathology, School of Medicine, Louisiana State University Health School of Medicine, New Orleans, LA 70112, USA
- Louisiana Cancer Research Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- Correspondence:
| |
Collapse
|
44
|
Volod O, Bunch CM, Miller J, Moore EE, Moore HB, Kwaan HC, Patel SS, Wiarda G, Aboukhaled M, Thomas SG, Fulkerson D, Erdman L, Tincher A, Walsh MM. Reply to Bareille et al. Are Viscoelastometric Assays of Old Generation Ready for Disposal? Comment on "Volod et al. Viscoelastic Hemostatic Assays: A Primer on Legacy and New Generation Devices. J. Clin. Med. 2022, 11, 860". J Clin Med 2023; 12:jcm12020478. [PMID: 36675408 PMCID: PMC9862366 DOI: 10.3390/jcm12020478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/16/2022] [Accepted: 12/27/2022] [Indexed: 01/11/2023] Open
Abstract
We are pleased to see that Bareille et al. have written a Commentary: "Are viscoelastometric assays of old generation ready for disposal?" [...].
Collapse
Affiliation(s)
- Oksana Volod
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Correspondence:
| | - Connor M. Bunch
- Department of Emergency Medicine, Henry Ford Hospital, Detroit, MI 48402, USA
- Department of Internal Medicine, Saint Joseph Regional Medical Center, Mishawaka, IN 46545, USA
| | - Joseph Miller
- Department of Emergency Medicine, Henry Ford Hospital, Detroit, MI 48402, USA
| | - Ernest E. Moore
- Department of Surgery, Ernest E. Moore Shock Trauma Center at Denver Health, University of Colorado Health Sciences Center, Denver, CO 80204, USA
| | - Hunter B. Moore
- Department of Surgery, Ernest E. Moore Shock Trauma Center at Denver Health, University of Colorado Health Sciences Center, Denver, CO 80204, USA
| | - Hau C. Kwaan
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Shivani S. Patel
- Department of Internal Medicine, Saint Joseph Regional Medical Center, Mishawaka, IN 46545, USA
| | - Grant Wiarda
- Department of Internal Medicine, Saint Joseph Regional Medical Center, Mishawaka, IN 46545, USA
| | - Michael Aboukhaled
- Department of Internal Medicine, Saint Joseph Regional Medical Center, Mishawaka, IN 46545, USA
| | - Scott G. Thomas
- Department of Trauma Surgery, Memorial Leighton Trauma Center, Beacon Health System, South Bend, IN 46601, USA
| | - Daniel Fulkerson
- Department of Trauma Surgery, Memorial Leighton Trauma Center, Beacon Health System, South Bend, IN 46601, USA
| | - Lee Erdman
- Department of Internal Medicine, Saint Joseph Regional Medical Center, Mishawaka, IN 46545, USA
| | - Anna Tincher
- Department of Internal Medicine, Saint Joseph Regional Medical Center, Mishawaka, IN 46545, USA
| | - Mark M. Walsh
- Department of Internal Medicine, Saint Joseph Regional Medical Center, Mishawaka, IN 46545, USA
| |
Collapse
|
45
|
Singh M, Pushpakumar S, Bard N, Zheng Y, Homme RP, Mokshagundam SPL, Tyagi SC. Simulation of COVID-19 symptoms in a genetically engineered mouse model: implications for the long haulers. Mol Cell Biochem 2023; 478:103-119. [PMID: 35731343 PMCID: PMC9214689 DOI: 10.1007/s11010-022-04487-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 05/30/2022] [Indexed: 01/24/2023]
Abstract
The ongoing pandemic (also known as coronavirus disease-19; COVID-19) by a constantly emerging viral agent commonly referred as the severe acute respiratory syndrome corona virus 2 or SARS-CoV-2 has revealed unique pathological findings from infected human beings, and the postmortem observations. The list of disease symptoms, and postmortem observations is too long to mention; however, SARS-CoV-2 has brought with it a whole new clinical syndrome in "long haulers" including dyspnea, chest pain, tachycardia, brain fog, exercise intolerance, and extreme fatigue. We opine that further improvement in delivering effective treatment, and preventive strategies would be benefited from validated animal disease models. In this context, we designed a study, and show that a genetically engineered mouse expressing the human angiotensin converting enzyme 2; ACE-2 (the receptor used by SARS-CoV-2 agent to enter host cells) represents an excellent investigative resource in simulating important clinical features of the COVID-19. The ACE-2 mouse model (which is susceptible to SARS-CoV-2) when administered with a recombinant SARS-CoV-2 spike protein (SP) intranasally exhibited a profound cytokine storm capable of altering the physiological parameters including significant changes in cardiac function along with multi-organ damage that was further confirmed via histological findings. More importantly, visceral organs from SP treated mice revealed thrombotic blood clots as seen during postmortem examination. Thus, the ACE-2 engineered mouse appears to be a suitable model for studying intimate viral pathogenesis thus paving the way for identification, and characterization of appropriate prophylactics as well as therapeutics for COVID-19 management.
Collapse
Affiliation(s)
- Mahavir Singh
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
| | - Sathnur Pushpakumar
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Nia Bard
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Yuting Zheng
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Rubens P Homme
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Sri Prakash L Mokshagundam
- Division of Endocrinology, Metabolism and Diabetes and Robley Rex VA Medical Center, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Suresh C Tyagi
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| |
Collapse
|
46
|
Chikomba C, Dlamini S, George JA, Pillay T. COVID Diagnostics: From Molecules to Omics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1412:141-158. [PMID: 37378765 DOI: 10.1007/978-3-031-28012-2_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
The identification and genetic sequencing of a novel coronavirus was key to the diagnosis and management of the global pandemic. An understanding of the SARS-CoV-2 structure and mechanism of injury is vital to explaining the disease course and the pathophysiology of the signs and symptoms observed. This particularly as the presentation, disease course, and severity are noted to be highly variable. The role of the spike protein and angiotensin-converting enzyme 2 (ACE-2) receptor in immune response and viral entry provides great insight into current and future diagnostics and therapeutics. This article reviews the traditional diagnostic methods, which include molecular testing methods, antigen testing, and antibody testing. The gold standard for diagnosis of COVID-19 is reverse transcriptase polymerase chain reaction (RT-PCR). There have been multiple improvements to these principles to help optimize the sensitivity, specificity, and user friendliness of the method. In addition, advancements in gene sequencing and identification have been integral to identifying variants and managing outbreaks. Serological and immunological testing have made significant contributions to the management of the COVID-19 pandemic, each with its unique benefits and limitations. A growing role of the laboratory is in triaging patients to determine which patients will most benefit from hospitalization and specialized care. This is imperative for rationalizing resources during outbreaks. As we learn to live with the pandemic, novel testing methods include the use of multiomic technologies and the greater utility of point of care.
Collapse
Affiliation(s)
- Chemedzai Chikomba
- Department of Chemical Pathology. National Health Laboratory Services and University of Witwatersrand, Johannesburg, South Africa
| | - Siphelele Dlamini
- Department of Chemical Pathology. National Health Laboratory Services and University of Witwatersrand, Johannesburg, South Africa
| | - Jaya A George
- Wits Diagnostic and Innovation Hub, University of Witwatersrand, Johannesburg, South Africa.
| | - Taryn Pillay
- Department of Chemical Pathology. National Health Laboratory Services and University of Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
47
|
Jun C, Cho YJ, Quinn PM, Song SK. Acute respiratory distress syndrome after treating ipsilateral femoral shaft and neck fracture with reamed intramedullary nailing in an asymptomatic COVID-19 patient. SAGE Open Med Case Rep 2023; 11:2050313X231168290. [PMID: 37113390 PMCID: PMC10119647 DOI: 10.1177/2050313x231168290] [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: 06/13/2022] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
Currently, it is quite difficult to determine the best timeframe of operative treatment, as well as conduct surgery on asymptomatic COVID-19 patients due to misunderstandings and unawareness of the patient's inflammation state. Caution needs to be taken in specific patient cohorts, particularly those with femoral shaft fractures, who are more likely to develop diseases like acute respiratory distress syndrome after a procedure like intramedullary nailing. In this case report, a 36-year-old patient suffered a motorcycle accident in which he suffered an ipsilateral femoral shaft and neck fracture of the hip. The patient's screening test for COVID-19 was positive before admission. Because the patient did not display any COVID-19-related symptoms upon arrival to the hospital, surgical fixation with a reamed intramedullary femoral nail was performed. Despite having a successful post-surgical outcome, the patient was diagnosed with acute respiratory distress syndrome 36 h after surgery, recovering fully after about 2 weeks. To prevent subsequent complications such as acute respiratory distress syndrome in a high inflammatory state patient like COVID-19, the respiratory status and the degree of systemic inflammation should be considered precisely when determining the surgical timing and method.
Collapse
Affiliation(s)
- Chungmu Jun
- Department of Orthopaedic Surgery, Daegu Catholic University Medical Center, Daegu, Korea
| | - Young-Jae Cho
- Department of Orthopaedic Surgery, Daegu Catholic University Medical Center, Daegu, Korea
| | - Patrick Michael Quinn
- Department of Orthopaedic Surgery, Wakamatsu Hospital, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Suk-Kyoon Song
- Department of Orthopaedic Surgery, Daegu Catholic University Medical Center, Daegu, Korea
- Suk-Kyoon Song, Department of Orthopaedic Surgery, Daegu Catholic University Medical Center, 33 Duryugongwon-ro 17-gil, Nam-gu, Daegu 42472, Korea.
| |
Collapse
|
48
|
Liang H, Lu R, Li Y, Pan P. Clinical characteristics of severe coronavirus disease 2019 patients with chronic obstructive pulmonary disease. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2022; 47:1695-1703. [PMID: 36748380 PMCID: PMC10930268 DOI: 10.11817/j.issn.1672-7347.2022.210596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Indexed: 02/08/2023]
Abstract
OBJECTIVES Coronavirus disease 2019 (COVID-19) in elderly and patients with chronic respiratory diseases (COPD) had a poor prognosis. COPD is one of the most common chronic respiratory diseases. We explore the epidemiological characteristics of patients with severe COVID-19 with COPD patients in order to provide medical evidence for the prevention and treatment of severe COVID-19. METHODS We retrospectively analyzed the clinical baseline characteristics, treatment strategies, disease progression and prognosis of 557 severe COVID-19 patients admitted to the West Court of Union Hospital of Huazhong University of Science and Technology from January 29, 2020 to April 8, 2020. RESULTS A total of 465 patients with severe COVID-19 were enrolled in the study, including 248 (53.3%) males and 217 (46.7%) females. The median age of severe COVID-19 patients was 62.0 years, and 53 patients were complicated with COPD. Common symptoms at the onset included fever (78.5%), dry cough (67.1%), shortness of breath (47.3%) and fatigue (40.9%). Compared with non-COPD patients, patients with COPD had significantly lower levels of SpO2 in admission (90.0% vs 92.0%, P=0.014). In terms of laboratory examinations, patients with COPD had higher levels of C-reactive protein, interleukin-6, procalcitonin, total bilirubin, blood urea nitrogen, serum creatinine, lipoprotein (a), high-sensitivity troponin I, and D-dimer, while had lower levels of platelet counts, albumin and apolipoprotein AI. Severe COVID-19 patients with COPD had higher Sequential Organ Failure Assessment scores [3.0(2.0, 3.0) vs 2.0(2.0, 3.0), P=0.038] and CURB-65 score [1.0(1.0, 2.0) vs1.0(0.0, 1.0), P<0.001], and a higher proportion of progressing to critical illness (28.3% vs 10.0%, P<0.001) with more complications [e.g. septic shock (15.1% vs 6.1%, P=0.034)], had higher incidence rates of antibiotic therapies (90.6% vs 77.2%, P=0.025), non-invasive (11.3% vs 1.7%, P<0.001) and invasive mechanical ventilation (17.0% vs 8.3%, P=0.039), ICU admission (17.0% vs 7.5%, P=0.021) and death (15.1% vs 6.1%, P=0.016). Cox proportion hazard model was carried out, and the results showed that comorbid COPD was an independent risk factor for severe COVID-19 patients progressing to critical type, after adjusting for age and gender [adjusted hazard ratio (AHR)=2.38(1.30-4.37), P=0.005] and additionally adjusting for chronic kidney diseases, hypertension, coronary heart disease [AHR=2.63(1.45-4.77), P<0.001], or additionally adjusting for some statistically significant laboratory findings [AHR=2.10(1.13-3.89), P=0.018]. CONCLUSIONS Severe COVID-19 patients with COPD have higher levels of disease severity, proportion of progression to critical illness and mortality rate. Individualized treatment strategies should be adopted to improve the prognosis of severe COVID-19 patients.
Collapse
Affiliation(s)
- Huaying Liang
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Rongli Lu
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yi Li
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Pinhua Pan
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
49
|
Faisal M, Alzahraa Al-Hattab F, Mohammed Al-boinin A, Mahmoud Al-Qudah B, Waheed MA, Danjuma M. The effects of thrombocytopenia, type 2 diabetes mellitus, and endothelial dysfunction on clinical outcomes in patients with COVID-19. Qatar Med J 2022; 2023:3. [PMID: 36588776 PMCID: PMC9800283 DOI: 10.5339/qmj.2023.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/30/2022] [Indexed: 12/29/2022] Open
Abstract
Diabetes mellitus is a well-recognized contributor to increased COVID-19 severity. Endothelial dysfunction has been implicated in the pathogenesis of COVID-19, while thrombocytopenia has been identified as a potential risk factor for severe COVID-19. In this study, we evaluated the combined effect of thrombocytopenia and other markers of endothelial dysfunction on disease outcomes in patients with type 2 diabetes and active COVID-19 infection. Our aim was to risk stratify patients with COVID-19 and type 2 diabetes mellitus, which can help identify patients with high-risk features who will benefit the most from hospital admission and a high level of care. This cross-sectional study was performed after reviewing secondary data of 932 patients with COVID-19 and type 2 diabetes mellitus in the outpatient and inpatient settings across Qatar between March 1, 2020 and May 7, 2020. Univariate and multivariate analyses, with adjustment for low platelet counts, were performed for the following variables: age, hemoglobin, white blood cells (WBC), lymphocytes, monocytes, eosinophils, alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, ferritin, D-dimer, and interleukin 6. Increasing age was associated with an increased risk for death and/or intensive care unit admission in diabetic patients with COVID-19 who have low platelet counts. These findings support the evidence found in the literature and give special attention to COVID-19 patients with low platelet counts and diabetes mellites. These results can guide physicians in making clinical decisions regarding hospital admission and escalation of care during follow-up in this population of patients.
Collapse
Affiliation(s)
- Mohanad Faisal
- Hamad Medical Corporation, Doha, Qatar. E-mail: ORCID: E-mail: https://orcid.org/0000-0002-2086-2957,E-mail: ORCID: E-mail: https://orcid.org/0000-0002-2086-2957
| | - Fatima Alzahraa Al-Hattab
- Hamad Medical Corporation, Doha, Qatar. E-mail: ORCID: E-mail: https://orcid.org/0000-0002-2086-2957
| | - Aisha Mohammed Al-boinin
- Hamad Medical Corporation, Doha, Qatar. E-mail: ORCID: E-mail: https://orcid.org/0000-0002-2086-2957
| | - Bara Mahmoud Al-Qudah
- Hamad Medical Corporation, Doha, Qatar. E-mail: ORCID: E-mail: https://orcid.org/0000-0002-2086-2957
| | - Muhammad Aamir Waheed
- Hamad Medical Corporation, Doha, Qatar. E-mail: ORCID: E-mail: https://orcid.org/0000-0002-2086-2957
| | - Mohammed Danjuma
- Hamad Medical Corporation, Doha, Qatar. E-mail: ORCID: E-mail: https://orcid.org/0000-0002-2086-2957
| |
Collapse
|
50
|
Moiseiwitsch N, Zwennes N, Szlam F, Sniecinski R, Brown A. COVID-19 patient fibrinogen produces dense clots with altered polymerization kinetics, partially explained by increased sialic acid. J Thromb Haemost 2022; 20:2909-2920. [PMID: 36111490 PMCID: PMC9537908 DOI: 10.1111/jth.15882] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/23/2022] [Accepted: 09/15/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND Thrombogenicity is a known complication of COVID-19, resulting from SARS-CoV-2 infection, with significant effects on morbidity and mortality. OBJECTIVE We aimed to better understand the effects of COVID-19 on fibrinogen and the resulting effects on clot structure, formation, and degradation. METHODS Fibrinogen isolated from COVID-19 patients and uninfected subjects was used to form uniformly concentrated clots (2 mg/ml), which were characterized using confocal microscopy, scanning electron microscopy, atomic force microscopy, and endogenous and exogenous fibrinolysis assays. Neuraminidase digestion and subsequent NANA assay were used to quantify sialic acid residue presence; clots made from the desialylated fibrinogen were then assayed similarly to the original fibrinogen clots. RESULTS Clots made from purified fibrinogen from COVID-19 patients were shown to be significantly stiffer and denser than clots made using fibrinogen from noninfected subjects. Endogenous and exogenous fibrinolysis assays demonstrated that clot polymerization and degradation dynamics were different for purified fibrinogen from COVID-19 patients compared with fibrinogen from noninfected subjects. Quantification of sialic acid residues via the NANA assay demonstrated that SARS-CoV-2-positive fibrinogen samples contained significantly more sialic acid. Desialylation via neuraminidase digestion resolved differences in clot density. Desialylation did not normalize differences in polymerization, but did affect rate of exogenous fibrinolysis. DISCUSSION These differences noted in purified SARS-CoV-2-positive clots demonstrate that structural differences in fibrinogen, and not just differences in gross fibrinogen concentration, contribute to clinical differences in thrombotic features associated with COVID-19. These structural differences are at least in part mediated by differential sialylation.
Collapse
Affiliation(s)
- Nina Moiseiwitsch
- Joint Department of Biomedical Engineering of University of North Carolina - Chapel Hill and North Carolina State University, Raleigh, North Carolina, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| | - Nicole Zwennes
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Fania Szlam
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Roman Sniecinski
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ashley Brown
- Joint Department of Biomedical Engineering of University of North Carolina - Chapel Hill and North Carolina State University, Raleigh, North Carolina, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
- Department of Material Science and Engineering, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|