1
|
Ma L, Lin X, Xu M, Ke X, Liu D, Chen Q. Exploring the biological mechanisms of severe COVID-19 in the elderly: Insights from an aged mouse model. Virulence 2025; 16:2487671. [PMID: 40228062 PMCID: PMC12005417 DOI: 10.1080/21505594.2025.2487671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 02/04/2025] [Accepted: 03/26/2025] [Indexed: 04/16/2025] Open
Abstract
The elderly population, who have increased susceptibility to severe outcomes, have been particularly impacted by the coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), leading to a global health crisis. However, definitive parameters or mechanisms underlying the severity of COVID-19 in elderly people remain unclear. Thus, this study seeks to elucidate the mechanism behind the increased vulnerability of elderly individuals to severe COVID-19. We employed an aged mouse model with a mouse-adapted SARS-CoV-2 strain to mimic the severe symptoms observed in elderly patients with COVID-19. Comprehensive analyses of the whole lung were performed using transcriptome and proteome sequencing, comparing data from aged and young mice. For transcriptome analysis, bulk RNA sequencing was conducted using an Illumina sequencing platform. Proteomic analysis was performed using mass spectrometry following protein extraction, digestion, and peptide labelling. We analysed the transcriptome and proteome profiles of young and aged mice and discovered that aged mice exhibited elevated baseline levels of inflammation and tissue damage repair. After SARS-CoV-2 infection, aged mice showed increased antiviral and inflammatory responses; however, these responses were weaker than those in young mice, with significant complement and coagulation cascade responses. In summary, our study demonstrates that the increased vulnerability of the elderly to severe COVID-19 may be attributed to an attenuated antiviral response and the overactivation of complement and coagulation cascades. Future research on antiviral and inflammatory responses is likely to yield treatments that reduce the severity of viral respiratory diseases in the elderly.
Collapse
Affiliation(s)
- Li Ma
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Xian Lin
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- Hubei Jiangxia Laboratory, Wuhan, China
| | - Meng Xu
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Xianliang Ke
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Di Liu
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Quanjiao Chen
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
2
|
Gheraibia S, Belattar N, Hassan ME, El-Nekeety AA, El-Sawy ER, Abdel-Wahhab MA. Molecular docking of polyphenol compounds and exploring the anticoagulant activity of Costus speciosus extracts in vitro and in vivo. Toxicol Rep 2025; 14:101961. [PMID: 40092046 PMCID: PMC11908605 DOI: 10.1016/j.toxrep.2025.101961] [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: 09/16/2024] [Revised: 01/29/2025] [Accepted: 02/10/2025] [Indexed: 03/19/2025] Open
Abstract
Anticoagulants have an important role in the prevention of cardiovascular disorders. Costus speciosus (Costaceae) is a medicinal herb used to treat COVID-19-induced thrombosis. The purpose of this study was to assess the anticoagulant activity of various C. speciosus aqueous (CSAE), ethanol (SCEE), and methanol (CSME) extracts in vivo and in vitro utilizing thrombin time (TT), activated partial thromboplastin time (aPTT), and prothrombin time (PT). Different concentrations of the three extracts were used to evaluate the anticoagulation effects in vitro. In the in vivo assay, male Sprague Dawley rats were used to test the CSME as in vivo anticoagulants. Three groups of rats included the control group and the groups that received CSME daily at a low (200 mg/kg) or high dose (400 mg/kg b.w) for 2 weeks. The molecular docking of the major bioactive constituents of the methanolic extract against the binding site of the thrombin inhibitor complex was evaluated. The HPLC detected 13, 10 and 11 polyphenols in the methanolic, ethanolic and aqueous extracts, respectively. The in vitro results showed that all the studied extracts had anticoagulant activity and increased aPTT, TT, and PT time. The in vivo experiment supported the in vitro results and demonstrated that CSME greatly prolonged the anticoagulant characteristics when compared to the negative control. Both findings suggested that these extracts have significant anticoagulant activity, with CSME being more effective and potentially useful in pharmaceutical applications as a natural anticoagulant medication.
Collapse
Affiliation(s)
- Sara Gheraibia
- Laboratory of Applied Biochemistry, Faculty of Sciences of Nature and Life, Ferhat Abbes University, Setif 1, Algeria
| | - Noureddine Belattar
- Laboratory of Applied Biochemistry, Faculty of Sciences of Nature and Life, Ferhat Abbes University, Setif 1, Algeria
| | - Marwa E. Hassan
- Toxicology Department, Research Institute of Medical Entomology, Giza, Egypt
| | - Aziza A. El-Nekeety
- Food Toxicology & Contaminants Department, National Research Centre, Dokki, Cairo, Egypt
| | - Eslam R. El-Sawy
- Chemistry of Natural Products Department, National Research Centre, Dokki, Cairo, Egypt
| | - Mosaad A. Abdel-Wahhab
- Food Toxicology & Contaminants Department, National Research Centre, Dokki, Cairo, Egypt
| |
Collapse
|
3
|
Sîrbu AC, Farcaș AD, Bocsan IC, Neag MA, Vesa ȘC, Suciu ȘM, Buzoianu AD. Biomarker Patterns and Their Association with Lung Injury in COVID-19 Patients. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:931. [PMID: 40428888 PMCID: PMC12113636 DOI: 10.3390/medicina61050931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 05/15/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025]
Abstract
Background and Objectives: The study investigates the relationship between accessible biomarkers and the extent of lung damage, assessed with computed tomography (CT) imaging, in patients hospitalized for COVID-19. Materials and Methods: This retrospective analysis was conducted in a hospital in Cluj-Napoca, Romania, and it includes 111 patients diagnosed with moderate to severe forms of COVID-19 during the Delta and Omicron waves. We evaluated the association of affordable lab works, such as C-reactive protein (CRP), procalcitonin, ferritin, neutrophil and lymphocyte counts, D-dimers, and albumin levels, with the extents of lung injury, pleural effusion, pulmonary embolism, and thoracic adenopathy. Results: Our data show that high CRP, neutrophil counts, ferritin, and procalcitonin levels, combined with lower lymphocyte and albumin levels, were significantly associated with >25% lung damage (p < 0.05). Elevated ferritin (≥274 ng/mL) and neutrophil counts (≥5.2 × 109/L) were independently associated with this threshold. CRP (≥2.67 mg/dL), CRP/albumin ratio (≥0.736), and ferritin had the highest sensitivity (86.8%). D-dimer was the sole biochemical marker associated with pulmonary embolism (p = 0.036). Pleural effusion was independently associated with lymphocyte count (cut-off < 0.605 × 109/L, p = 0.013). Thoracic lymphadenopathy was also associated with increased neutrophil counts and a heightened inflammatory response. Conclusions: These findings suggest that ferritin and the CRP/albumin ratio can serve as indicators for patients with extensive parenchymal damage. D-dimer levels were the only ones significantly associated with thromboembolic events, while lymphopenia appears to be a useful indicator of pleural involvement. Thus, these readily available biomarkers can prove useful in anticipating radiological severity in patients hospitalized with COVID-19.
Collapse
Affiliation(s)
- Alexandru Constantin Sîrbu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hațieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (A.C.S.); (I.C.B.)
| | - Anca Daniela Farcaș
- Department of Internal Medicine, Cardiology and Gastroenterology, Iuliu Hațieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
- 1st Cardiology Department, Cluj-Napoca Emergency County Hospital, 400006 Cluj-Napoca, Romania
| | - Ioana Corina Bocsan
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hațieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (A.C.S.); (I.C.B.)
| | - Maria Adriana Neag
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hațieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (A.C.S.); (I.C.B.)
| | - Ștefan Cristian Vesa
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hațieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (A.C.S.); (I.C.B.)
| | - Șoimița Mihaela Suciu
- Department of Physiology, Iuliu Hațieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania;
| | - Anca Dana Buzoianu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hațieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (A.C.S.); (I.C.B.)
| |
Collapse
|
4
|
Katayama H. Neutrophil Extracellular Traps Capturing SARS-CoV-2 in the Lung Tissue (Alveoli and Parenchyma) Cause Microthrombi - A Strategy to Eliminate SARS-CoV-2 From the Circulation as Degraded Fibrin Clots. Circ Rep 2025; 7:379-382. [PMID: 40352121 PMCID: PMC12061506 DOI: 10.1253/circrep.cr-24-0157] [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: 12/06/2024] [Revised: 02/02/2025] [Accepted: 02/03/2025] [Indexed: 05/14/2025] Open
Abstract
Background It has been thought that neutrophil extracellular traps (NETs) and thrombosis exacerbate COVID-19, but, on the other hand, NETs are an important player in innate immunity. The precise roles of NETs and thrombosis in the course of COVID-19 have not been fully elucidated. Methods and Results The roles were investigated in the literature and a new theory was formulated. When neutrophils encounter SARS-CoV-2 in the lung tissue, they undergo NETosis and capture the virus. This capture is triggered by electrostatic interaction between histones in NETs and SARS-CoV-2; histones are highly positively charged, and viruses, including SARS-CoV-2, have a net negative charge under physiological pH. NETs that capture SARS-CoV-2 fall into alveolar capillaries through the collapsed endothelium to spare the lung tissue from the toxicity of NETs. NETs in the microvessels cause microthrombosis; positively charged histones induce the aggregation of negatively charged platelets, which leads to microthrombi. Microthrombi engulfing SARS-CoV-2 are consolidated into fibrin clots, which are eventually degraded by increased fibrinolysis and eliminated from the circulation. Conclusions This novel theory suggests that NETosis and microthrombosis are phenomena inevitably elicited in COVID-19, and in combination they are a system newly termed "NETombosis". Undegraded fibrin clots remaining in the microcirculation may be the cause of the sequelae, because they cause long-lasting circulatory failure in various organs.
Collapse
|
5
|
Tsou PS, Ali RA, Lu C, Sule G, Carmona-Rivera C, Lucotti S, Ikari Y, Wu Q, Campbell PL, Gurrea-Rubio M, Maeda K, Fox SE, Brodie WD, Mattichak MN, Foster C, Tambralli A, Yalavarthi S, Amin MA, Kmetova K, Fonseca BM, Chong E, Zuo Y, Maile MD, Imberti L, Caruso A, Caccuri F, Quaresima V, Sottini A, Kuhns DB, Fink D, Castagnoli R, Delmonte OM, Kenney H, Zhang Y, Magliocco M, Su H, Notarangelo L, Zemans RL, Mao-Draayer Y, Matei IR, Salvatore M, Lyden D, Kanthi Y, Kaplan MJ, Knight JS, Fox DA. Soluble CD13 is a potential mediator of neutrophil-induced thrombogenic inflammation in SARS-CoV-2 infection. JCI Insight 2025; 10:e184975. [PMID: 40168094 DOI: 10.1172/jci.insight.184975] [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: 07/19/2024] [Accepted: 03/27/2025] [Indexed: 04/03/2025] Open
Abstract
The soluble variant of the ectopeptidase CD13 (sCD13), released from the cell surface by matrix metalloproteinase 14 (MMP14), is a potent pro-inflammatory mediator, displaying chemotactic, angiogenic, and arthritogenic properties through bradykinin receptor B1 (B1R). We revealed a link between sCD13 and amplified neutrophil-mediated inflammatory responses in SARS-CoV-2 infection. sCD13 was markedly elevated in patients with COVID-19 and correlated with disease severity and variants, ethnicity, inflammation markers, and neutrophil extracellular trap formation (NETosis). Neutrophils treated with sCD13 showed heightened NETosis and chemotaxis, which were inhibited by sCD13 receptor blockade. Meanwhile sCD13 did not induce platelet aggregation. Single-cell analysis of COVID-19 lungs revealed coexpression of CD13 and MMP14 by various cell types, and higher CD13 expression compared with controls. Neutrophils with high CD13 mRNA were enriched for genes associated with immaturity, though CD13 protein expression was lower. Histological examination of COVID-19 lungs revealed CD13-positive leukocytes trapped in vessels with fibrin thrombi. Flow cytometry verified the presence of B1R and a second sCD13 receptor, protease-activated receptor 4, on monocytes and neutrophils. These findings identify sCD13 as a potential instigator of COVID-19-associated NETosis, potentiating vascular stress and thromboembolic complications. The potent pro-inflammatory effects of sCD13 may contribute to severe COVID-19, suggesting that sCD13 and its receptors might be therapeutic targets.
Collapse
Affiliation(s)
- Pei-Suen Tsou
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Ramadan A Ali
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Chenyang Lu
- Division of Rheumatology, Department of Internal Medicine, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Gautam Sule
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Carmelo Carmona-Rivera
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland, USA
| | - Serena Lucotti
- Department of Pediatrics, Weill Cornell Medical College, New York, New York, USA
| | - Yuzo Ikari
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Qi Wu
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Phillip L Campbell
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Mikel Gurrea-Rubio
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Kohei Maeda
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Sharon E Fox
- Department of Pathology, Louisiana State University, Health Sciences Center, New Orleans, Louisiana, USA
| | - William D Brodie
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Megan N Mattichak
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Caroline Foster
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Ajay Tambralli
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Srilakshmi Yalavarthi
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - M Asif Amin
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Katarina Kmetova
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Bruna Mazetto Fonseca
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
- School of Medical Science, University of Campinas (UNICAMP), Campinas, Brazil
| | - Emily Chong
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Yu Zuo
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Michael D Maile
- Department of Anesthesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Luisa Imberti
- Section of Microbiology, University of Brescia, Brescia, Italy
| | - Arnaldo Caruso
- Section of Microbiology, University of Brescia, Brescia, Italy
| | | | - Virginia Quaresima
- Clinical Chemistry Laboratory, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Alessandra Sottini
- Clinical Chemistry Laboratory, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Douglas B Kuhns
- Leidos Biomedical Research, Inc.; Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Danielle Fink
- Leidos Biomedical Research, Inc.; Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Riccardo Castagnoli
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Ottavia M Delmonte
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Heather Kenney
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Yu Zhang
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Mary Magliocco
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Helen Su
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Luigi Notarangelo
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Rachel L Zemans
- Division of Pulmonary & Critical Care Medicine, Department of Internal Medicine; and Program in Cellular and Molecular Biology, School of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Yang Mao-Draayer
- Multiple Sclerosis Center of Excellence, Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Irina R Matei
- Department of Pediatrics, Weill Cornell Medical College, New York, New York, USA
| | - Mirella Salvatore
- Joan and Sanford I. Weill Department of Medicine and Department of Population Health Sciences, Weill Cornell Medical College, New York, New York, USA
| | - David Lyden
- Department of Pediatrics, Weill Cornell Medical College, New York, New York, USA
| | - Yogendra Kanthi
- Division of Intramural Research, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland, USA
| | - Jason S Knight
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - David A Fox
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
6
|
Zuo B, Jin L, Sun Z, Hu H, Yin Y, Yang S, Liu Z. Predicting Superaverage Length of Stay in COPD Patients with Hypercapnic Respiratory Failure Using Machine Learning. J Inflamm Res 2025; 18:5993-6008. [PMID: 40357373 PMCID: PMC12068394 DOI: 10.2147/jir.s511092] [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: 12/08/2024] [Accepted: 04/19/2025] [Indexed: 05/15/2025] Open
Abstract
Objective The purpose of this study was to develop and validate machine learning models that can predict superaverage length of stay in hypercapnic-type respiratory failure and to compare the performance of each model. Furthermore, screen and select the optimal individualized risk assessment model. This model is capable of predicting in advance whether an inpatient's length of stay will exceed the average duration, thereby enhancing its clinical application and utility. Methods The study included 568 COPD patients with hypercapnic respiratory failure, 426 inpatients from the Department of Respiratory and Critical Care Medicine of Yancheng First People's Hospital in the modeling group and 142 inpatients from the Department of Respiratory and Critical Care Medicine of Jiangsu Provincial People's Hospital in the external validation group. Ten machine learning algorithms were used to develop and validate a model for predicting superaverage length of stay, and the best model was evaluated and selected. Results We screened 83 candidate variables using the Boruta algorithm and identified 9 potentially important variables, including: cerebrovascular disease, white blood cell count, hematocrit, D-dimer, activated partial thromboplastin time, fibrin degradation products, partial pressure of carbon dioxide, reduced hemoglobin, and oxyhemoglobin. Cerebrovascular disease, hematocrit, activated partial thromboplastin time, partial pressure of carbon dioxide, reduced hemoglobin and oxyhemoglobin were independent risk factors for superaverage length of stay in COPD patients with hypercapnic respiratory failure. The Catboost model is the optimal model on both the modeling dataset and the external validation set. The interactive web calculator was developed using the Shiny framework, leveraging a predictive model based on Catboost. Conclusion The Catboost model has the most advantages and can be used for clinical evaluation and patient monitoring.
Collapse
Affiliation(s)
- Bingqing Zuo
- Department of Pulmonary and Critical Care Medicine, The Yancheng Clinical College of Xuzhou Medical University, The First People’s Hospital of Yancheng, Yancheng, Jiangsu, 224006, People’s Republic of China
| | - Lin Jin
- Third Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kuming, Yunnan, 650000, People’s Republic of China
| | - Zhixiao Sun
- Department of Pulmonary and Critical Care Medicine, The Yancheng Clinical College of Xuzhou Medical University, The First People’s Hospital of Yancheng, Yancheng, Jiangsu, 224006, People’s Republic of China
| | - Hang Hu
- Department of Pulmonary and Critical Care Medicine, The Yancheng Clinical College of Xuzhou Medical University, The First People’s Hospital of Yancheng, Yancheng, Jiangsu, 224006, People’s Republic of China
| | - Yuan Yin
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People’s Republic of China
| | - Shuanying Yang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shanxi, 710004, People’s Republic of China
| | - Zhongxiang Liu
- Department of Pulmonary and Critical Care Medicine, The Yancheng Clinical College of Xuzhou Medical University, The First People’s Hospital of Yancheng, Yancheng, Jiangsu, 224006, People’s Republic of China
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shanxi, 710004, People’s Republic of China
| |
Collapse
|
7
|
Anzic N, Stoiber W, Obermeyer A, Mertz KD, Stalder A, Haslbauer JD, Tzankov A. Two-sided effects of neutrophil extracellular traps and changes in the myeloid compartment in acute COVID-19: A histopathological study on autopsy cases from the first and second COVID-19 waves in Switzerland. J Leukoc Biol 2025; 117:qiaf056. [PMID: 40356380 DOI: 10.1093/jleuko/qiaf056] [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: 01/13/2025] [Revised: 03/05/2025] [Accepted: 05/12/2025] [Indexed: 05/15/2025] Open
Abstract
Severe COVID-19 is characterized by complex immunopathology that involves inflammation, endothelial dysfunction, and immunothrombosis. Neutrophil extracellular traps (NETs) have been recognized as key factors in the severity of the disease, with their emergence correlating to viral load, immmunothrombosis, and organ damage. In this study, we investigated the role of NETosis and macrophage activation in the course of severe COVID-19. We analyzed 23 autopsy samples from patients who died from COVID-19 and performed immunohistochemical staining and stereological point counting to quantify leukocyte infiltration and NET formation among other histopathological parameters. Our results showcase 2 evident immunophenotypes: lowNET and highNET. The lowNET group displayed lower NET formation, higher viral loads, and an increased incidence of secondary infections, as well as shorter survival times. In contrast, the highNET group exhibited increased neutrophil activation, pronounced endothelial damage and thrombotic complications, as well as prolonged survival times. Our data suggest a dual role of NETosis in COVID-19: initially protective, limiting viral replication, but later likely detrimental through immunothrombosis and tissue damage. These findings underline the need for tailored therapeutic actions, with early antiviral and immune-modulating interventions for lowNET patients and strategies aiming to limit excessive NETosis and coagulopathy in highNET patients. Further research is needed to define the timing of interventions based on the dynamics of NETosis.
Collapse
Affiliation(s)
- Nina Anzic
- Institute of Pathology, Cantonal Hospital Luzern, Spitalstrasse 16, 6000 Luzern, Switzerland
| | - Walter Stoiber
- Department of Environment and Biodiversity, University of Salzburg, Hellbrunnerstrasse 34, 5020 Salzburg, Austria
| | - Astrid Obermeyer
- Department of Environment and Biodiversity, University of Salzburg, Hellbrunnerstrasse 34, 5020 Salzburg, Austria
| | - Kirsten D Mertz
- Institute of Medical Genetics and Pathology, University Hospital Basel, Schönbeinstrasse 40, 4031 Basel, Switzerland
| | - Anna Stalder
- Institute of Medical Genetics and Pathology, University Hospital Basel, Schönbeinstrasse 40, 4031 Basel, Switzerland
| | - Jasmin D Haslbauer
- Institute of Medical Genetics and Pathology, University Hospital Basel, Schönbeinstrasse 40, 4031 Basel, Switzerland
| | - Alexandar Tzankov
- Institute of Medical Genetics and Pathology, University Hospital Basel, Schönbeinstrasse 40, 4031 Basel, Switzerland
| |
Collapse
|
8
|
Li J, Li SJ. Chondroitin sulfate binds to main protease of SARS-CoV-2 and efficaciously inhibits its activity. Int J Biol Macromol 2025; 306:141547. [PMID: 40020804 DOI: 10.1016/j.ijbiomac.2025.141547] [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: 11/11/2024] [Revised: 01/27/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is constantly mutating and spreading globally, posing a great threat to people's lives and health. The main protease of SARS-CoV-2 (Mpro, also called 3CLpro) is an attractive drug target for SARS-CoV-2, due to its crucial role in processing the viral replication. Here, we showed that chondroitin sulfate (CS) from pig, cattle and shark efficaciously inhibits Mpro activity of SARS-CoV-2 with half maximal inhibitory concentrations (IC50) of 0.148, 0.121 and 0.119 μM, respectively, through a fluorescence resonance energy transfer (FRET) assay. The inhibition pattern of CSs against Mpro activity is competitive inhibition, with inhibition constants (Ki) of CSs derived from pig, bovine and shark are 0.111, 0.096, and 0.107 μM, respectively, indicating significant inhibitory effects of CSs on Mpro activity. Protein fluorescence quenching demonstrated that porcine, bovine and shark CSs strongly bind to Mpro protein with dissociation constants (KD) of 28.31, 28.47 and 20.66 μM at 25 °C at a physiological condition, respectively, mainly through van der Waals and hydrogen bond interactions. Molecular docking and dynamics analysis provides an insight into structural information of the binding of the CSs with Mpro protein. Our findings suggested that CSs from different origins might be a promising food ingredient for the prevention of the SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Jinwen Li
- Department of Condensed Matter and Material Physics, School of Physics Science, Nankai University, Tianjin 300071, PR China.
| | - Shu Jie Li
- Department of Biophysics, School of Physics Science, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, PR China; Qilu Institute of Technology, Shandong 250200, PR China.
| |
Collapse
|
9
|
Yang Y, Li D, Nie J, Wang J, Huang H, Hang X. A Nomogram for Predicting Survival in Patients with SARS-CoV-2 Omicron Variant Pneumonia Based on Admission Data. Infect Drug Resist 2025; 18:2093-2104. [PMID: 40303607 PMCID: PMC12039831 DOI: 10.2147/idr.s509178] [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: 11/28/2024] [Accepted: 04/15/2025] [Indexed: 05/02/2025] Open
Abstract
Purpose Patients with severe SARS-CoV-2 omicron variant pneumonia pose a serious challenge. This study aimed to develop a nomogram for predicting survival using chest computed tomography (CT) imaging features and laboratory test results based on admission data. Patients and Methods A total of 436 patients with SARS-CoV-2 pneumonia (323 and 113 in the training and validation groups, respectively) were enrolled. Pneumonitis volume, assessed on chest CT scans at admission, was used to identify low- and high-risk groups. Risk analysis was performed using clinical symptoms, laboratory findings, and chest CT imaging features. A predictive algorithm was developed using Cox multivariate analysis. Results The high-risk group had a shorter survival duration than the low-risk group. Significant differences in mortality rate, neutrophil and lymphocyte counts, C-reactive protein (CRP) concentration, and urea nitrogen level were observed between the two groups. In the training group, age, pneumonia volume, total bilirubin, and blood urea nitrogen were independent prognostic factors. In the validation group, age, pneumonia volume, neutrophil count, and CRP were independent prognostic factors. A personalized prediction model for survival outcomes was developed using independent predictors. Conclusion A personalized prediction model was created to forecast the 5-, 10-, 15-, 20-, and 30-day survival rates of patients with COVID-19 omicron variant pneumonia based on admission data, and can be used to determine the survival rate and early treatment of severe patients.
Collapse
Affiliation(s)
- Yinghao Yang
- Department of Infectious Diseases, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
- Department of Infectious Diseases, the 988th Hospital of the Joint Logistic Support Force, Zhengzhou, People’s Republic of China
| | - Dong Li
- Department of Infectious Diseases, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
- Department of Gastroenterology, The 971th Hospital of PLA Navy, Qingdao, People’s Republic of China
| | - Jinqiu Nie
- Department of Infectious Diseases, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Junxue Wang
- Department of Infectious Diseases, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Huili Huang
- Department of Infectious Diseases, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Xiaofeng Hang
- Department of Infectious Diseases, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
| |
Collapse
|
10
|
Abudouleh E, Owaidah T, Alhamlan F, Al-Qahtani AA, Aljowaie RM, Al-Ghnnam F, Fe Bohol M, Al-Qahtani AA. SARS-COV-2 causes significant abnormalities in the fibrinolysis system of patients: correlation between viral mutations, variants and thrombosis. Front Cell Infect Microbiol 2025; 15:1531412. [PMID: 40302923 PMCID: PMC12037514 DOI: 10.3389/fcimb.2025.1531412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 03/19/2025] [Indexed: 05/02/2025] Open
Abstract
Background Coronavirus disease (COVID-19) is reported as a complex disorder affecting multiple systems and coagulopathy that can cause mortality. In this study, we investigated the correlation of SARS-CoV-2 mutations found in blood samples with various changes in the fibrinolysis system, as well as the severity of the disease based on outcome and whether or not these patients were admitted into the ICU. Materials and methods COVID-19 patients (n = 446) admitted to our institute between 2021 and 2022 were recruited. Blood samples were collected, and a sequence analysis of the SARS-CoV-2 spike gene was isolated from the blood. Measured several parameters of fibrinolysis and coagulation, including alpha-2-antiplasmin and plasminogen, thrombin activatable fibrinolysis inhibitor (TAFI), tissue plasminogen activator (tPA), plasminogen activator inhibitor-1 (PAI-1), D-dimer, and fibrinogen levels. Results SARS-CoV-2 RNA was found in 123/446 (27.6%) of the blood samples. The N501Y, D614G, K417N, and P681R mutations among COVID-19 patients were associated with higher admissions to the ICU (P = 0.0057, P = 0.0068, P = 0.0193, and P = 0.018, respectively). Omicron (BA.1.1) variant variants are highly associated with thrombosis (P = 0.002) in hospitalized COVID-19 patients that are unvaccinated and have comorbidity conditions. The plasma levels of tPA, aPTT, and D-dimer were significantly higher in participants who had the N501Y mutation (P = 0.044, P = 0.024, and P = 0.027, respectively). Conclusion Thrombosis was the most prevalent condition among severe COVID-19 patients. The correlation between specific SARS-CoV-2 new variants and thrombosis warrants more investigation.
Collapse
Affiliation(s)
- Esra’a Abudouleh
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Tarek Owaidah
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- Department of Pathology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Fatimah Alhamlan
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
- Department of Microbiology and Immunology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Arwa A. Al-Qahtani
- Department of Family Medicine, College of Medicine, Al-Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
| | - Reem M. Aljowaie
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Fatimah Al-Ghnnam
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Marie Fe Bohol
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Ahmed A. Al-Qahtani
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
- Department of Microbiology and Immunology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
11
|
Tcholadze G, Pantsulaia I, Ratiani L, Kopaleishvili L, Bolotashvili T, Jorbenadze A, Chikovani T. The Prognostic Value of Circulating Cytokines and Complete Blood Count-Based Inflammatory Markers in COVID-19 Patients With Atrial Fibrillation. Cardiol Res 2025; 16:153-160. [PMID: 40051670 PMCID: PMC11882233 DOI: 10.14740/cr2027] [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: 12/06/2024] [Accepted: 02/05/2025] [Indexed: 03/09/2025] Open
Abstract
Background Atrial fibrillation (AF) is associated with a high burden of cardiovascular disease, which has been worsened during the coronavirus disease 2019 (COVID-19) pandemic. The purpose of this study was to assess the association between clinical markers, especially interleukin-6 (IL-6) and other inflammatory biomarkers, and the severity of COVID-19 in patients with AF. Methods This retrospective cohort study categorized patients based on clinical presentations and laboratory results to investigate the prognostic significance of inflammatory markers in COVID-19 outcomes among those with AF. The study included 100 hospitalized COVID-19 patients aged between 40 to 80 years and was conducted at the Chapidze Hospital in Tbilisi, Georgia. Patients were then grouped by disease severity according to computed tomography (CT) scores, clinical symptoms, respiratory rate and oxygen saturation. Levels of IL-6 were obtained at three time points during hospitalization. A broad range of laboratory tests, including C-reactive protein (CRP), ferritin, and D-dimer, were also conducted. Results Patients with AF demonstrated significantly elevated levels of IL-6 (P = 0.024), CRP (P = 0.001), and ferritin (P < 0.001), suggesting a severe inflammatory response. D-dimer levels were also notably higher in the AF group (P < 0.005), indicating an increased risk of thrombotic complications. Oxygen saturation levels were significantly lower (P = 0.004) and CT scores higher in patients with AF. Furthermore, the length of hospitalization was longer among patients with AF (median duration significantly higher, P = 0.032), indicating a more severe disease course. Conclusions The proinflammatory markers such as IL-6 are independent predictive markers of COVID-19 severity in AF patients. Overall, it highlights urgent treatment approaches, such as available anti-inflammatory drugs, for COVID-19 patients with arrhythmias. Combining these biomarkers into clinical routines helps us better identify patients at risk and how to treat them.
Collapse
Affiliation(s)
- Giorgi Tcholadze
- Department of Immunology, Tbilisi State Medical University, Tbilisi 0177, Georgia
| | - Ia Pantsulaia
- Department of Immunology, Tbilisi State Medical University, Tbilisi 0177, Georgia
- Vl. Bakhutashvili Institute of Medical Biotechnology, Tbilisi State Medical University, Tbilisi 0159, Georgia
| | | | | | | | | | - Tinatin Chikovani
- Department of Immunology, Tbilisi State Medical University, Tbilisi 0177, Georgia
| |
Collapse
|
12
|
Nagoba BS, Dhotre SV, Gavkare AM, Mumbre SS, Dhotre PS. Convergence of COVID-19 and recurrent stroke: In-hospital mortality risks explored. World J Virol 2025; 14:99904. [PMID: 40134845 PMCID: PMC11612881 DOI: 10.5501/wjv.v14.i1.99904] [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: 08/02/2024] [Revised: 10/14/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
This editorial comments on the article by Desai et al, which investigates the impact of coronavirus disease 2019 (COVID-19) on in-hospital mortality among patients with recurrent stroke using data from the 2020 National Inpatient Sample. The findings reveal significantly higher mortality rates in COVID-19-positive patients compared to non-COVID-19 patients, particularly among middle-aged individuals, males, and ethnic minorities. This editorial explores the underlying mechanisms contributing to these outcomes and discusses the clinical implications for targeted management strategies in high-risk groups. The results emphasize the need for comprehensive approaches to mitigate the heightened risks faced by recurrent stroke patients during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Basavraj S Nagoba
- Department of Microbiology, Maharashtra Institute of Medical Sciences and Research (Medical College), Latur 413531, India
| | - Shree V Dhotre
- Department of Microbiology, Ashwini Rural Medical College, Solapur 413001, India
| | - Ajay M Gavkare
- Department of Physiology, Maharashtra Institute of Medical Sciences and Research (Medical College), Latur 413531, India
| | - Sachin S Mumbre
- Department of Community Medicine, Ashwini Rural Medical College, Solapur 413001, India
| | - Pradnya S Dhotre
- Department of Biochemistry, Ashwini Rural Medical College, Solapur 413001, India
| |
Collapse
|
13
|
Nab L, Visser C, van Bussel BCT, Beishuizen A, Bemelmans RHH, Ten Cate H, Croles FN, van Guldener C, de Jager CPC, Huisman MV, Nijziel MR, Kamphuisen PW, Klok FA, Koster SCE, Kuşadasi N, Meijer K, den Uil CA, Schutgens REG, Stam F, Vlaar APJ, Vlot EA, Linschoten MPM, Asselbergs FW, Kruip MJHA, le Cessie S, Cannegieter SC. Assessing differential application of thromboprophylaxis regimes related to risk of pulmonary embolism and mortality in COVID-19 patients through instrumental variable analysis. Sci Rep 2025; 15:10321. [PMID: 40133355 PMCID: PMC11937556 DOI: 10.1038/s41598-024-77858-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 10/25/2024] [Indexed: 03/27/2025] Open
Abstract
Thrombotic complications are common in Coronavirus disease 2019 (COVID-19) patients, with pulmonary embolism (PE) being the most frequent. Randomised trials have provided inconclusive results on the optimal dosage of thromboprophylaxis in critically ill COVID-19 patients. We utilized data from the multicentre CAPACITY-COVID patient registry to assess the effect of differential application of Low Molecular Weight Heparin (LMWH) dose protocols on PE and in-hospital mortality risk in critically ill COVID-19 patients. An instrumental variable analysis was performed to estimate the intention-to-treat effect, utilizing differences in thromboprophylaxis prescribing behaviour between hospitals. We included 939 patients with PCR confirmed SARS-CoV-2 infection from 34 hospitals. Two-hundred-and-one patients (21%) developed a PE. The adjusted cause-specific HR of PE was 0.92 (95% CI: 0.73-1.16) per doubling of LMWH dose. The adjusted cause-specific HR for in-hospital mortality was 0.82 (95% CI: 0.65-1.02) per doubling of LMWH dose. This dose-response relationship was shown to be non-linear. To conclude, this study did not find evidence for an effect of LMWH dose on the risk of PE, but suggested a non-linear decreased risk of in-hospital mortality for higher doses of LMWH. However, uncertainty remains, and the dose-response relationship between LMWH dose and in-hospital mortality needs further investigation in well-designed studies.
Collapse
Affiliation(s)
- Linda Nab
- Department of Clinical Epidemiology, Leiden University Medical Centre, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Chantal Visser
- Department of Haematology, Erasmus MC, Erasmus University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Bas C T van Bussel
- Department of Intensive Care, Maastricht University Medical Centre+, Maastricht, The Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
| | - Albertus Beishuizen
- Department of Intensive Care, Medical Spectrum Twente, Enschede, The Netherlands
| | - Remy H H Bemelmans
- Department of Internal Medicine, Hospital Gelderse Vallei, Ede, The Netherlands
| | - Hugo Ten Cate
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
- Department of Internal Medicine, Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands
- Department of Biochemistry, Laboratory for Clinical Thrombosis and Hemostasis, Maastricht University, Maastricht, The Netherlands
- Thrombosis Expertise Center, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - F Nanne Croles
- Department of Internal Medicine, Hospital St. Jansdal, Harderwijk, The Netherlands
| | - Coen van Guldener
- Department of Internal Medicine, Amphia Hospital, Breda, The Netherlands
| | - C Peter C de Jager
- Department of Intensive Care, Jeroen Bosch Hospital, 's-Hertogenbosch, The Netherlands
| | - Menno V Huisman
- Department of Medicine - Thrombosis and Haemostasis, Leiden University Medical Centre, Leiden, The Netherlands
- Central Diagnostic Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marten R Nijziel
- Department of Haematology-Oncology, Catherina Hospital, Eindhoven, The Netherlands
| | - Pieter W Kamphuisen
- Department of Internal Medicine, Tergooi Medical Center, Hilversum, The Netherlands
- Department of Vascular Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Frederikus A Klok
- Department of Medicine - Thrombosis and Haemostasis, Leiden University Medical Centre, Leiden, The Netherlands
| | | | - Nuray Kuşadasi
- Department of Intensive Care, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Karina Meijer
- Department of Haematology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Corstiaan A den Uil
- Department of Cardiology, Erasmus MC, Erasmus University Medical Centre Rotterdam, Rotterdam, The Netherlands
- Department of Intensive Care, Erasmus MC, Erasmus University Medical Centre Rotterdam, Rotterdam, The Netherlands
- Department of Intensive Care, Maasstad Ziekenhuis, Rotterdam, the Netherlands
| | - Roger E G Schutgens
- Centre for Benign Haematology, Thrombosis and Haemostasis, Van Creveldkliniek, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Frank Stam
- Department of Internal Medicine, Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands
| | - Alexander P J Vlaar
- Department of Intensive Care, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Eline A Vlot
- Department of Intensive Care, St. Antonius Hospital, Utrecht, The Netherlands
| | - Marijke P M Linschoten
- Department of Cardiology, Division of Heart and Lungs, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Folkert W Asselbergs
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- Institute of Health Informatics, University College London, London, UK
| | - Marieke J H A Kruip
- Department of Haematology, Erasmus MC, Erasmus University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Saskia le Cessie
- Department of Clinical Epidemiology, Leiden University Medical Centre, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
- Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, The Netherlands
| | - Suzanne C Cannegieter
- Department of Clinical Epidemiology, Leiden University Medical Centre, P.O. Box 9600, 2300 RC, Leiden, The Netherlands.
| |
Collapse
|
14
|
dos S. P. Andrade AC, Lacasse E, Dubuc I, Gudimard L, Gravel A, Puhm F, Campolina-Silva G, Queiroz-Junior C, Allaeys I, Prunier J, Azeggouar Wallen O, Dumais É, Belleannée C, Droit A, Flamand N, Boilard É, Flamand L. Deficiency in platelet 12-lipoxygenase exacerbates inflammation and disease severity during SARS-CoV-2 infection. Proc Natl Acad Sci U S A 2025; 122:e2420441122. [PMID: 40100623 PMCID: PMC11962506 DOI: 10.1073/pnas.2420441122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/27/2025] [Indexed: 03/20/2025] Open
Abstract
Platelets, known for maintaining blood balance, also participate in antimicrobial defense. Upon severeacute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, platelets become hyperactivated, releasing molecules such as cytokines, granule contents, and bioactive lipids. The key effector biolipids produced by platelets include 12-hydroxyeicosatetraenoic acid (12-HETE) and 12-hydroxyeicosatrienoic acid (12-HETrE), produced by 12-lipoxygenase (12-LOX), and prostaglandins and thromboxane, produced by cyclooxygenase-1. While prostaglandin E2 and thromboxane B2 were previously associated with lung inflammation in severe COVID-19, the role of platelet 12-LOX in SARS-CoV-2 infection remains unclear. Using mice deficient for platelets' 12-LOX, we report that SARS-CoV-2 infection resulted in higher lung inflammation characterized by histopathological tissue analysis, increased leukocyte infiltrates, and cytokine production relative to wild-type mice. In addition, distinct platelet and lung transcriptomic changes, including alterations in NOD-like receptor (NLR) family pyrin domain-containing 1 (NLRP1) inflammasome-related gene expression, were observed. Mass spectrometry lipidomic analysis in 12-LOX-deficient-infected mice revealed significant changes in bioactive lipid content, including reduced levels of 12-HETrE that inversely correlated with disease severity. Finally, platelet 12-LOX deficiency was associated with increased morbidity and lower survival rates relative to wild type (WT) mice. Overall, this study highlights the complex interplay between 12-LOX-related lipid metabolism and inflammatory responses during SARS-CoV-2 infection. The findings provide valuable insights into potential therapeutic targets aimed at mitigating severe outcomes, emphasizing the pivotal role of platelet enzymes in the host response to viral infections.
Collapse
Affiliation(s)
- Ana Claudia dos S. P. Andrade
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Emile Lacasse
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Isabelle Dubuc
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Leslie Gudimard
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Annie Gravel
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Florian Puhm
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Gabriel Campolina-Silva
- Division of Reproduction, mother and youth health, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Celso Queiroz-Junior
- Morphology Department, Universidade Federal de Minas Gerais, Belo Horizonte31270-901, Brazil
| | - Isabelle Allaeys
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Julien Prunier
- Division of Endocrinology and Nephrology, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Oumaima Azeggouar Wallen
- Centre de recherche de l’Institut Universitaire de cardiologie et pneumologie de Québec, Division of pneumology, Faculty of medicine, Université Laval, Québec City, QCG1V 4G5, Canada
| | - Élizabeth Dumais
- Centre de recherche de l’Institut Universitaire de cardiologie et pneumologie de Québec, Division of pneumology, Faculty of medicine, Université Laval, Québec City, QCG1V 4G5, Canada
| | - Clémence Belleannée
- Division of Reproduction, mother and youth health, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Arnaud Droit
- Division of Endocrinology and Nephrology, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Nicolas Flamand
- Centre de recherche de l’Institut Universitaire de cardiologie et pneumologie de Québec, Division of pneumology, Faculty of medicine, Université Laval, Québec City, QCG1V 4G5, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Université Laval, Québec City, QCG1V 4G5, Canada
| | - Éric Boilard
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
- Centre de Recherche ARThrite–Arthrite, Recherche, Traitements, Université Laval, Québec, QCG1V 4G2, Canada
- Department of microbiology, infectious disease and immunology, Faculty of Medicine, Université Laval, Québec City, QCG1V 0A6, Canada
| | - Louis Flamand
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
- Centre de Recherche ARThrite–Arthrite, Recherche, Traitements, Université Laval, Québec, QCG1V 4G2, Canada
- Department of microbiology, infectious disease and immunology, Faculty of Medicine, Université Laval, Québec City, QCG1V 0A6, Canada
| |
Collapse
|
15
|
Blagoeva V, Hodzhev V, Uchikov P, Dobreva-Yatseva B, Stoyanova R, Shterev M, Atiq S, Prasad A, Shankar Babu S. Clinical Course and Mortality Predictors in Adult Hospitalized Patients with COVID-19 Infection-A Retrospective Cohort Study. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:579. [PMID: 40282870 PMCID: PMC12028986 DOI: 10.3390/medicina61040579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/12/2025] [Accepted: 03/19/2025] [Indexed: 04/29/2025]
Abstract
Background and Objectives: Bulgaria had the highest mortality rate of COVID-19 in Europe and the second highest in the world based on statistical data. This study aimed to determine the mortality predictors in 306 adult patients with COVID-19 infection, treated at the COVID-19 Ward of St. George University Hospital in Plovdiv, Bulgaria in the period of August 2021-April 2022. Materials and Methods: All admitted and treated patients had a positive PCR test for SARS-CoV-2. They were assigned in three groups based on the severity rating scale published in NIH COVID-19 Treatment Guidelines by Stat Pearls Publishing, 2022. Demographic, clinical, and laboratory parameters and pre-existing comorbidities were investigated. Parametric and non-parametric methods were used for statistics. Logistic regression was applied for parameters significantly associated with mortality. Results: Data showed that demographic indicators were not significantly associated with poorer outcome. Among comorbidities, cardiovascular, chronic pulmonary and endocrine disorders were found to be related to poor survival rates (p = 0.003, p = 0.003 and p = 0.017 resp.) Clinical symptoms, such as sore throat, dry or productive cough and breathlessness, were determinants of poor outcome (p = 0.027, p = 0.029, p = 0.004 and p = 0.002 resp.). Laboratory parameters linked to mortality were elevated d-dimers (p = 0.015), ferritin (p = 0.009) and creatinine (p = 0.038). p02 < 50 and saturation < 90 also indicated a higher risk of death (p = 0.006 and p = 0021). Conclusions: Logistic regression showed that each stage of disease severity increased the risk of death 3.6 times, chronic pulmonary disorders increased it by 4.1, endocrine by 2.4 and dyspnea by 3.1 times.
Collapse
Affiliation(s)
- Vesela Blagoeva
- Pulmonology and Tuberculosis Section, First Department of Internal Medicine, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
| | - Vladimir Hodzhev
- Pulmonology and Tuberculosis Section, First Department of Internal Medicine, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
| | - Petar Uchikov
- Department of Special Surgery, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
| | - Bistra Dobreva-Yatseva
- First Department of Internal Medicine, Cardiology Section, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
| | - Rumyana Stoyanova
- Department of Health Management and Health Economics, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
| | - Maritza Shterev
- Medical Faculty, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (M.S.); (S.A.); (S.S.B.)
| | - Samiya Atiq
- Medical Faculty, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (M.S.); (S.A.); (S.S.B.)
| | - Akanksha Prasad
- Medical Faculty, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (M.S.); (S.A.); (S.S.B.)
| | - Sriharini Shankar Babu
- Medical Faculty, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (M.S.); (S.A.); (S.S.B.)
| |
Collapse
|
16
|
Shakhidzhanov S, Filippova A, Bovt E, Gubkin A, Sukhikh G, Tsarenko S, Spiridonov I, Protsenko D, Zateyshchikov D, Vasilieva E, Kalinskaya A, Dukhin O, Novichkova G, Karamzin S, Serebriyskiy I, Lipets E, Kopnenkova D, Morozova D, Melnikova E, Rumyantsev A, Ataullakhanov F. Severely Ill COVID-19 Patients May Exhibit Hypercoagulability Despite Escalated Anticoagulation. J Clin Med 2025; 14:1966. [PMID: 40142778 PMCID: PMC11943368 DOI: 10.3390/jcm14061966] [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: 02/13/2025] [Revised: 03/07/2025] [Accepted: 03/09/2025] [Indexed: 03/28/2025] Open
Abstract
Introduction: Severely ill COVID-19 patients receiving prophylactic-dose anticoagulation exhibit high rates of thrombosis and mortality. The escalation of anticoagulation also does not reduce mortality and has an uncertain impact on thrombosis rates. The reasons why escalated doses fail to outperform prophylactic doses in reducing risks of thrombosis and death in severely ill COVID-19 patients remain unclear. We hypothesized that escalated anticoagulation would not effectively prevent hypercoagulability and, consequently, would not reduce the risk of thrombosis and death in some severely ill patients. Methods: We conducted a prospective multicenter study that enrolled 3860 COVID-19 patients, including 1654 severely ill. They received different doses of low-molecular-weight or unfractionated heparin, and their blood coagulation was monitored with activated partial thromboplastin time, D-dimer, and Thrombodynamics. A primary outcome was hypercoagulability detected by Thrombodynamics. Blood samples were collected at the trough level of anticoagulation. Results: We found that escalated anticoagulation did not prevent hypercoagulability in 28.3% of severely ill patients at the trough level of the pharmacological activity. Severely ill patients with such hypercoagulability had higher levels of inflammation markers and better creatinine clearance compared to severely ill patients without it. Hypercoagulability detected by Thrombodynamics was associated with a 1.68-fold higher hazard rate for death and a 3.19-fold higher hazard rate for thrombosis. Elevated D-dimer levels were also associated with higher hazard rates for thrombosis and death, while shortened APTTs were not. The simultaneous use of Thrombodynamics and D-dimer data enhanced the accuracy for predicting thrombotic events and fatal outcomes in severely ill patients. Conclusions: Thrombodynamics reliably detects hypercoagulability in COVID-19 patients and can be used in conjunction with D-dimer to assess the risk of thrombosis and death in severely ill patients. The pharmacological effect of LMWH at the trough level might be too low to prevent thrombosis in some severely ill patients with severe inflammation and better creatinine clearance, even if escalated doses are used.
Collapse
Affiliation(s)
- Soslan Shakhidzhanov
- Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, 117997 Moscow, Russia; (A.F.); (E.B.); (G.N.); (D.M.); (A.R.)
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Anna Filippova
- Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, 117997 Moscow, Russia; (A.F.); (E.B.); (G.N.); (D.M.); (A.R.)
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Elizaveta Bovt
- Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, 117997 Moscow, Russia; (A.F.); (E.B.); (G.N.); (D.M.); (A.R.)
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Andrew Gubkin
- Central Clinical Hospital No. 2 Named After N.A.Semashko “RZD-Medicine”, 121359 Moscow, Russia;
| | - Gennady Sukhikh
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I.Kulakov, 117997 Moscow, Russia;
| | - Sergey Tsarenko
- City Clinical Hospital No. 52 of Moscow Health Care Department, 123182 Moscow, Russia;
| | - Ilya Spiridonov
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Denis Protsenko
- Moscow Multiprofile Clinical Center “Kommunarka” of Moscow Healthcare Department, 142770 Moscow, Russia; (D.P.); (D.K.)
| | - Dmitriy Zateyshchikov
- City Clinical Hospital No. 51 of Moscow Health Care Department, 121309 Moscow, Russia;
| | - Elena Vasilieva
- City Clinical Hospital No. 23 of Moscow Health Care Department, 109004 Moscow, Russia; (E.V.); (A.K.); (O.D.)
| | - Anna Kalinskaya
- City Clinical Hospital No. 23 of Moscow Health Care Department, 109004 Moscow, Russia; (E.V.); (A.K.); (O.D.)
| | - Oleg Dukhin
- City Clinical Hospital No. 23 of Moscow Health Care Department, 109004 Moscow, Russia; (E.V.); (A.K.); (O.D.)
| | - Galina Novichkova
- Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, 117997 Moscow, Russia; (A.F.); (E.B.); (G.N.); (D.M.); (A.R.)
| | - Sergey Karamzin
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Ilya Serebriyskiy
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Elena Lipets
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Daria Kopnenkova
- Moscow Multiprofile Clinical Center “Kommunarka” of Moscow Healthcare Department, 142770 Moscow, Russia; (D.P.); (D.K.)
| | - Daria Morozova
- Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, 117997 Moscow, Russia; (A.F.); (E.B.); (G.N.); (D.M.); (A.R.)
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Evgeniya Melnikova
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Alexander Rumyantsev
- Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, 117997 Moscow, Russia; (A.F.); (E.B.); (G.N.); (D.M.); (A.R.)
| | - Fazoil Ataullakhanov
- Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, 117997 Moscow, Russia; (A.F.); (E.B.); (G.N.); (D.M.); (A.R.)
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| |
Collapse
|
17
|
Kang X, Wang G, Liu B, Wang Z. Knowledge mapping of Guillain-Barré syndrome from January 2013 to October 2023: A bibliometric analysis. Medicine (Baltimore) 2025; 104:e41830. [PMID: 40101082 PMCID: PMC11922404 DOI: 10.1097/md.0000000000041830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 02/21/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND With the COVID-19 pandemic and the serious sequelae, foreign factor-induced Guillain-Barré syndrome (GBS) has become a research focus in autoimmune peripheral neuropathies. The study employs a bibliometric system to illustrate the research hotspots and trends in GBS based on pertinent literature from January 2013 to October 2023. METHODS The Web of Science Core Collection retrieved articles on GBS from January 1, 2013, to October 28, 2023. These articles were then visualized and statistically evaluated using VOSviewer, CiteSpace software, R version 4.2.1, and Microsoft Office Excel 2019. RESULTS A total of 4269 articles on GBS were gathered. The United States of America produced the most publications (28.55%, 1219/4269), followed by China (14.22%, 607/4269). The world's leading country was the United States of America, with the most publications, the most substantial international cooperation, and the highest centrality (0.17). Union of French Research Universities (UDICE)-French Research Universities in France was the most productive organization (189 articles). Lancet was the highest cocited journal (2428), and Professor Jacobs, Bart C., was the most prolific author (93). The most significant increases were shown for the keywords coronavirus, respiratory failure, and coronavirus disease 2019. The novel coronavirus is an emerging virus that may cause GBS, indicating a promising area of research. CONCLUSIONS The study on GBS was illustrated using bibliometrics, and it covers trends in international collaboration, publications, and research hotspots. These findings allow the scientific community to pinpoint the novel ideas and directions that will drive future GBS research.
Collapse
Affiliation(s)
- Xue Kang
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Guowei Wang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, Chongqing, China
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bin Liu
- College of Biology, Hunan University, Changsha, China
| | - Zhenhai Wang
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, China
- Diagnosis and Treatment Engineering Technology Research Center of Nervous System Diseases of Ningxia Hui Autonomous Region, Yinchuan, China
- Neurology Center, General Hospital of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
18
|
Zhang Y, Wang H, Liu K, Sun R, Wang Y, Guo J, Zhou W, Zheng H, Qi Y. Manganese-Based Nanozyme Alleviates Acute Kidney Injury via Nrf2/HO-1 and PI3K/Akt/NF-κB Signaling Pathways. ACS Biomater Sci Eng 2025; 11:1751-1764. [PMID: 39878300 DOI: 10.1021/acsbiomaterials.4c02093] [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] [Indexed: 01/31/2025]
Abstract
Acute renal injury (AKI) has a high incidence rate and mortality, but current treatment methods are limited. As a kind of nanomaterial with enzyme-like activity, nanozyme has shown outstanding advantages in treating AKI according to recent reports. Herein, we assess the potential of manganese-based nanozymes (MnO2-BSA NPs) with excellent biosafety in effectively alleviating AKI. Our findings in vitro and in vivo reveal that MnO2-BSA NPs exert regulatory effects on oxidative stress, inflammation, and apoptosis. These effects are mediated through activation of the Nrf2/HO-1 and PI3K/Akt/NF-κB pathways. Notably, it was observed that the cytoprotective effect of MnO2-BSA NPs is abrogated upon inhibition of Nrf2 expression, highlighting the important role of this transcription factor in cellular protection. In summary, the study demonstrates the protective effect of MnO2-BSA NPs in AKI and provides the molecular mechanisms involved, which can contribute to the advancement of potential therapeutic interventions for nanozyme-based treatments.
Collapse
Affiliation(s)
- Yang Zhang
- School of Public Health, Jilin University, Changchun, Jilin, 130021, P. R. China
| | - Han Wang
- School of Public Health, Jilin University, Changchun, Jilin, 130021, P. R. China
| | - Ke Liu
- School of Public Health, Jilin University, Changchun, Jilin, 130021, P. R. China
| | - Ruimeng Sun
- School of Public Health, Jilin University, Changchun, Jilin, 130021, P. R. China
| | - Yurou Wang
- School of Public Health, Jilin University, Changchun, Jilin, 130021, P. R. China
| | - Jiayu Guo
- School of Public Health, Jilin University, Changchun, Jilin, 130021, P. R. China
| | - Wenxiang Zhou
- School of Public Health, Jilin University, Changchun, Jilin, 130021, P. R. China
| | - Haoran Zheng
- School of Public Health, Jilin University, Changchun, Jilin, 130021, P. R. China
| | - Yanfei Qi
- School of Public Health, Jilin University, Changchun, Jilin, 130021, P. R. China
| |
Collapse
|
19
|
Iroungou BA, Nze O A, M Kandet Y H, Longo-Pendy NM, Mezogho-Obame ND, Dikoumba AC, Mangouka GL. Interest of D-dimer level, severity of COVID-19 and cost of management in Gabon. World J Crit Care Med 2025; 14:100486. [DOI: 10.5492/wjccm.v14.i1.100486] [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: 08/18/2024] [Revised: 10/02/2024] [Accepted: 10/30/2024] [Indexed: 12/11/2024] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is strongly associated with an increased risk of thrombotic events, including severe outcomes such as pulmonary embolism. Elevated D-dimer levels are a critical biomarker for assessing this risk. In Gabon, early implementation of anticoagulation therapy and D-dimer testing has been crucial in managing COVID-19. This study hypothesizes that elevated D-dimer levels are linked to increased COVID-19 severity.
AIM To determine the impact of D-dimer levels on COVID-19 severity and their role in guiding clinical decisions.
METHODS This retrospective study analyzed COVID-19 patients admitted to two hospitals in Gabon between March 2020 and December 2023. The study included patients with confirmed COVID-19 diagnoses and available D-dimer measurements at admission. Data on demographics, clinical outcomes, D-dimer levels, and healthcare costs were collected. COVID-19 severity was classified as non-severe (outpatients) or severe (inpatients). A multivariable logistic regression model was used to assess the relationship between D-dimer levels and disease severity, with adjusted odds ratios (OR) and 95%CI.
RESULTS A total of 3004 patients were included, with a mean age of 50.17 years, and the majority were female (53.43%). Elevated D-dimer levels were found in 65.81% of patients, and 57.21% of these experienced severe COVID-19. Univariate analysis showed that patients with elevated D-dimer levels had 3.33 times higher odds of severe COVID-19 (OR = 3.33, 95%CI: 2.84-3.92, P < 0.001), and this association remained significant in the multivariable analysis, adjusted for age, sex, and year of collection. The financial analysis revealed a substantial burden, particularly for uninsured patients.
CONCLUSION D-dimer predicts COVID-19 severity and guides treatment, but the high cost of anticoagulant therapy highlights the need for policies ensuring affordable access in resource-limited settings like Gabon.
Collapse
Affiliation(s)
- Berthe A Iroungou
- Unité Mixte de Recherche, Centre International de Recherches Médicales de Franceville et le Service de Santé Militaire, Libreville 20404, Estuaire, Gabon
| | - Arnaud Nze O
- AP-HP Health Economics, Research Unit, Htel Dieu Hospital, Paris 75004, Ile-de-France, France
| | - Helga M Kandet Y
- Laboratory, Hôpital D'Instruction des Armées D'Akanda, Libreville 20404, Estuaire, Gabon
| | - Neil-Michel Longo-Pendy
- Unité de Recherche en Écologie de la Santé, Centre Interdisciplinaire de Recherches Médicales de Franceville, Franceville 769, Gabon
| | - Nina D Mezogho-Obame
- Laboratory, Hôpital D'Instruction des Armées Omar Bongo Ondimba, Libreville 20404, Estuaire, Gabon
| | - Annicet-Clotaire Dikoumba
- Unité Mixte de Recherche, Centre International de Recherches Médicales de Franceville et le Service de Santé Militaire, Libreville 20404, Estuaire, Gabon
| | - Guignali L Mangouka
- Department of Medecine Polyvalente, Hôpital D'Instruction des Armées Omar Bongo Ondimba, Libreville 20404, Estuaire, Gabon
| |
Collapse
|
20
|
Shergill S, Elshibly M, Hothi SS, Parke KS, England RJ, Wormleighton JV, Hudson GJ, Tunnicliffe EM, Wild J, Smith SM, Francis S, Toshner M, Sattar N, Khunti K, Brightling CE, Antoniades C, Berry C, Greenwood JP, Moss A, Neubauer S, McCann GP, Raman B, Arnold JR. Assessing the impact of COmorbidities and Sociodemographic factors on Multiorgan Injury following COVID-19: rationale and protocol design of COSMIC, a UK multicentre observational study of COVID-negative controls. BMJ Open 2025; 15:e089508. [PMID: 40050066 PMCID: PMC11887317 DOI: 10.1136/bmjopen-2024-089508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025] Open
Abstract
INTRODUCTION SARS-CoV-2 disease (COVID-19) has had an enormous health and economic impact globally. Although primarily a respiratory illness, multi-organ involvement is common in COVID-19, with evidence of vascular-mediated damage in the heart, liver, kidneys and brain in a substantial proportion of patients following moderate-to-severe infection. The pathophysiology and long-term clinical implications of multi-organ injury remain to be fully elucidated. Age, gender, ethnicity, frailty and deprivation are key determinants of infection severity, and both morbidity and mortality appear higher in patients with underlying comorbidities such as ischaemic heart disease, hypertension and diabetes. Our aim is to gain mechanistic insights into the pathophysiology of multiorgan dysfunction in people with COVID-19 and maximise the impact of national COVID-19 studies with a comparison group of COVID-negative controls. METHODS AND ANALYSIS COmorbidities and Sociodemographic factors on Multiorgan Injury following COVID-19 (COSMIC) is a prospective, multicentre UK study which will recruit 200 subjects without clinical evidence of prior COVID-19 and perform extensive phenotyping with multiorgan imaging, biobank serum storage, functional assessment and patient reported outcome measures, providing a robust control population to facilitate current work and serve as an invaluable bioresource for future observational studies. ETHICS AND DISSEMINATION Approved by the National Research Ethics Service Committee East Midlands (REC reference 19/EM/0295). Results will be disseminated via peer-reviewed journals and scientific meetings. TRIAL REGISTRATION NUMBER COSMIC is registered as an extension of C-MORE (Capturing Multi-ORgan Effects of COVID-19) on ClinicalTrials.gov (NCT04510025).
Collapse
Affiliation(s)
- Simran Shergill
- Department of Cardiovascular Sciences and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, University of Leicester, Leicester, UK
| | - Mohamed Elshibly
- Department of Cardiovascular Sciences and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, University of Leicester, Leicester, UK
| | - Sandeep S Hothi
- Department of Cardiology, Heart and Lung Centre, Royal Wolverhampton NHS Trust, Wolverhampton, UK
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Kelly S Parke
- Department of Cardiovascular Sciences and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, University of Leicester, Leicester, UK
- Department of Radiology, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Rachel J England
- Department of Cardiovascular Sciences and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, University of Leicester, Leicester, UK
- Department of Radiology, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Joanne V Wormleighton
- Department of Cardiovascular Sciences and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, University of Leicester, Leicester, UK
- Department of Radiology, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - George J Hudson
- Department of Cardiovascular Sciences and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, University of Leicester, Leicester, UK
| | - Elizabeth M Tunnicliffe
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford Centre for Clinical Magnetic Resonance Research, National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - James Wild
- POLARIS Imaging Group, The Department of Infection, Immunity and Cardiovascular Disease, The University of Sheffield Faculty of Medicine Dentistry and Health, Sheffield, UK
- Insigneo Institute for in silico Medicine, The University of Sheffield Faculty of Medicine Dentistry and Health, Sheffield, UK
| | - Stephen M Smith
- Oxford Centre for Functional MRI of the Brain, Wellcome Centre for Integrative Neuroimaging, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Sue Francis
- Sir Peter Mansfield Imaging Centre, School of Physics and Astronomy, University of Nottingham, Nottingham, UK
- National Institute for Health Research Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Mark Toshner
- National Institute for Health Research Cambridge Clinical Research Facility and Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences and British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Kamlesh Khunti
- Diabetes Research Centre, University of Leicester, Leicester, UK
| | - Christopher E Brightling
- Leicester National Institute for Health Research Biomedical Research Centre (Respiratory theme), Leicester, UK
- Infection, Inflammation and Immunity, University of Leicester, Leicester, UK
| | - Charalambos Antoniades
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford Centre for Clinical Magnetic Resonance Research, National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Colin Berry
- Institute of Cardiovascular and Medical Sciences and British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - John P Greenwood
- Baker Heart and Diabetes Institute South Australia, Melbourne, Victoria, Australia
| | - Alastair Moss
- Department of Cardiovascular Sciences and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, University of Leicester, Leicester, UK
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford Centre for Clinical Magnetic Resonance Research, National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Gerry P McCann
- Department of Cardiovascular Sciences and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, University of Leicester, Leicester, UK
| | - Betty Raman
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford Centre for Clinical Magnetic Resonance Research, National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Jayanth Ranjit Arnold
- Department of Cardiovascular Sciences and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, University of Leicester, Leicester, UK
| |
Collapse
|
21
|
Yang H, Li J, Zhang C, Sierra AP, Shen B. Predictive model for daily risk alerts in sepsis patients in the ICU: visualization and clinical analysis of risk indicators. PRECISION CLINICAL MEDICINE 2025; 8:pbaf003. [PMID: 40041421 PMCID: PMC11878768 DOI: 10.1093/pcmedi/pbaf003] [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: 11/17/2024] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 03/06/2025] Open
Abstract
This study introduces a novel Transformer-based time-series framework designed to revolutionize risk stratification in Intensive Care Units (ICUs) by predicting patient outcomes with high temporal precision. Leveraging sequential data from the eICU database, our two-stage architecture dynamically captures evolving health trajectories throughout a patient's ICU stay, enabling real-time identification of high-risk individuals and actionable insights for personalized interventions. The model demonstrated exceptional predictive power, achieving a progressive AUC increase from 0.87 (±0.021) on admission day to 0.92 (±0.009) by day 5, reflecting its capacity to assimilate longitudinal physiological patterns. Rigorous external validation across geographically diverse cohorts-including an 81.8% accuracy on Chinese sepsis data (AUC=0.73) and 76.56% accuracy on MIMIC-IV-3.1 (AUC=0.84)-confirmed robust generalizability. Crucially, SHAP-derived temporal heatmaps unveiled mortality-associated feature dynamics over time, bridging the gap between model predictions and clinically interpretable biomarkers. These findings establish a new paradigm for ICU prognostics, where data-driven temporal modeling synergizes with clinician expertise to optimize triage, reduce diagnostic latency, and ultimately improve survival outcomes in critical care.
Collapse
Affiliation(s)
- Hao Yang
- Information Center, West China Hospital of Sichuan University, Chengdu 610041, China
- Department of Computer Science and Information Technologies, Research Center for Information and Communications Technologies, University of A Coruña, Biomedical Research Institute of a Coruña, A Coruña 15071, Spain
| | - Jiaxi Li
- Department of Clinical Laboratory Medicine, Jinniu Maternity and Child Health Hospital of Chengdu, Chengdu 610031, China
| | - Chi Zhang
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Alejandro Pazos Sierra
- Department of Computer Science and Information Technologies, Research Center for Information and Communications Technologies, University of A Coruña, Biomedical Research Institute of a Coruña, A Coruña 15071, Spain
| | - Bairong Shen
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
22
|
Leitzke M, Roach DT, Hesse S, Schönknecht P, Becker GA, Rullmann M, Sattler B, Sabri O. Long COVID - a critical disruption of cholinergic neurotransmission? Bioelectron Med 2025; 11:5. [PMID: 40011942 DOI: 10.1186/s42234-025-00167-8] [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: 12/03/2024] [Accepted: 01/30/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Following the COVID-19 pandemic, there are many chronically ill Long COVID (LC) patients with different symptoms of varying degrees of severity. The pathological pathways of LC remain unclear until recently and make identification of path mechanisms and exploration of therapeutic options an urgent challenge. There is an apparent relationship between LC symptoms and impaired cholinergic neurotransmission. METHODS This paper reviews the current literature on the effects of blocked nicotinic acetylcholine receptors (nAChRs) on the main affected organ and cell systems and contrasts this with the unblocking effects of the alkaloid nicotine. In addition, mechanisms are presented that could explain the previously unexplained phenomenon of post-vaccination syndrome (PVS). The fact that not only SARS-CoV-2 but numerous other viruses can bind to nAChRs is discussed under the assumption that numerous other post-viral diseases and autoimmune diseases (ADs) may also be due to impaired cholinergic transmission. We also present a case report that demonstrates changes in cholinergic transmission, specifically, the availability of α4β2 nAChRs by using (-)-[18F]Flubatine whole-body positron emission tomography (PET) imaging of cholinergic dysfunction in a LC patient along with a significant neurological improvement before and after low-dose transcutaneous nicotine (LDTN) administration. Lastly, a descriptive analysis and evaluation were conducted on the results of a survey involving 231 users of LDTN. RESULTS A substantial body of research has emerged that offers a compelling explanation for the phenomenon of LC, suggesting that it can be plausibly explained because of impaired nAChR function in the human body. Following a ten-day course of transcutaneous nicotine administration, no enduring neuropathological manifestations were observed in the patient. This observation was accompanied by a significant increase in the number of free ligand binding sites (LBS) of nAChRs, as determined by (-)-[18F]Flubatine PET imaging. The analysis of the survey shows that the majority of patients (73.5%) report a significant improvement in the symptoms of their LC/MEF/CFS disease as a result of LDTN. CONCLUSIONS In conclusion, based on current knowledge, LDTN appears to be a promising and safe procedure to relieve LC symptoms with no expected long-term harm.
Collapse
Affiliation(s)
- Marco Leitzke
- Department of Nuclear Medicine, University of Leipzig Medical Centre, Leipzig, 04103, Germany.
- Department of Anesthesiology, Intensive Care Medicine, Pain- and Palliative Therapy Helios Clinics, Colditzer Straße 48, Leisnig, 04703, Germany.
| | - Donald Troy Roach
- School of Comillas University, Renegade Research, Madrid, 28015, Spain
| | - Swen Hesse
- Department of Nuclear Medicine, University of Leipzig Medical Centre, Leipzig, 04103, Germany
| | - Peter Schönknecht
- Department of Psychiatry and Neurology Altscherbitz, Schkeuditz, 04435, Germany
- Outpatient Department for Forensic-Psychiatric Research, University of Leipzig, Leipzig, 04103, Germany
| | - Georg-Alexander Becker
- Department of Nuclear Medicine, University of Leipzig Medical Centre, Leipzig, 04103, Germany
| | - Michael Rullmann
- Department of Nuclear Medicine, University of Leipzig Medical Centre, Leipzig, 04103, Germany
| | - Bernhardt Sattler
- Department of Nuclear Medicine, University of Leipzig Medical Centre, Leipzig, 04103, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University of Leipzig Medical Centre, Leipzig, 04103, Germany
| |
Collapse
|
23
|
Popazu C, Romila A, Petrea M, Grosu RM, Lescai AM, Vlad AL, Oprea VD, Baltă AAȘ. Overview of Inflammatory and Coagulation Markers in Elderly Patients with COVID-19: Retrospective Analysis of Laboratory Results. Life (Basel) 2025; 15:370. [PMID: 40141715 PMCID: PMC11943672 DOI: 10.3390/life15030370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Elderly patients with COVID-19 often exhibit a complex interplay between hypercoagulability and coagulopathy, key factors in determining the risk of severe complications and mortality. This study aimed to analyze coagulation and inflammatory markers to identify critical predictors of adverse outcomes in this vulnerable population. Material and Methods: The retrospective study was conducted on a sample of 1429 elderly patients (≥60 years) diagnosed with COVID-19, hospitalized in "Sf. Ap. Andrei" St. Apostle Andrew's County Emergency Hospital in various wards between March 2020 and August 2022. Data were collected from medical records and included inflammatory markers (C-reactive protein, procalcitonin, ESR) and coagulation markers (prothrombin time, INR, fibrinogen, D-dimer). The SPSS 2.0 statistical software was used to conduct the study. Results:Coagulation markers: Prothrombin activity averaged 74.22%, below normal levels, indicating a heightened bleeding risk, while fibrinogen levels were significantly elevated (mean: 531.69 mg/dL), reflecting hypercoagulability. Prolonged prothrombin time (mean: 17.28 s) and elevated INR (International normalized ratio) (mean: 1.51) were associated with increased mortality, emphasizing their role in risk stratification. Elevated D-dimer levels (mean: 2.75 mg/L) further highlighted thromboembolic risks. Inflammatory markers: C-reactive protein (CRP) and erythrocyte sedimentation rate (ESR) showed marked elevations (mean CRP: 92.09 mg/L, mean ESR: 58.47 mm/h), correlating with heightened systemic inflammation and poor outcomes. Bacterial infections: Elevated procalcitonin (mean: 1.98 ng/mL) suggested secondary bacterial infections, particularly in mechanically ventilated patients, significantly worsening prognosis. Conclusions: The duality of hypercoagulability and coagulopathy in elderly COVID-19 patients underscores the importance of consistently monitoring coagulation markers such as prothrombin time, INR, D-dimer, and fibrinogen. Simultaneously, elevated inflammatory markers and secondary bacterial infections require prompt therapeutic interventions. This study highlights the critical need for personalized management strategies to mitigate complications and reduce mortality in this high-risk population.
Collapse
Affiliation(s)
- Corina Popazu
- Clinical-Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, Str. Domnească 35, 800201 Galaţi, Romania; (C.P.); (A.L.V.); (A.A.Ș.B.)
| | - Aurelia Romila
- Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, Str. Domnească 35, 800201 Galaţi, Romania; (A.R.); (V.D.O.)
| | - Marius Petrea
- Pre-Clinical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, Str. Domnească 35, 800201 Galaţi, Romania; (M.P.); (R.M.G.)
| | - Robert Marius Grosu
- Pre-Clinical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, Str. Domnească 35, 800201 Galaţi, Romania; (M.P.); (R.M.G.)
| | - Alina-Maria Lescai
- Clinical-Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, Str. Domnească 35, 800201 Galaţi, Romania; (C.P.); (A.L.V.); (A.A.Ș.B.)
| | - Adriana Liliana Vlad
- Clinical-Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, Str. Domnească 35, 800201 Galaţi, Romania; (C.P.); (A.L.V.); (A.A.Ș.B.)
| | - Violeta Diana Oprea
- Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, Str. Domnească 35, 800201 Galaţi, Romania; (A.R.); (V.D.O.)
| | - Alexia Anastasia Ștefania Baltă
- Clinical-Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, Str. Domnească 35, 800201 Galaţi, Romania; (C.P.); (A.L.V.); (A.A.Ș.B.)
| |
Collapse
|
24
|
Dinoi G, Togo MV, Guida P, Deruvo C, Samarelli F, Imbrici P, Nicolotti O, De Luca A, Mastroianni F, Liantonio A, Altomare CD. Retrospective Clinical Investigation into the Association Between Abnormal Blood Clotting, Oral Anticoagulant Therapy, and Medium-Term Mortality in a Cohort of COVID-19 Patients. Biomedicines 2025; 13:535. [PMID: 40149514 PMCID: PMC11940371 DOI: 10.3390/biomedicines13030535] [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: 12/18/2024] [Revised: 02/14/2025] [Accepted: 02/16/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: People affected by COVID-19 are exposed to abnormal clotting and endothelial dysfunction, which may trigger thromboembolic events. This study aimed at retrospectively investigating whether oral anticoagulant therapy (OAT), encompassing either direct oral anticoagulants (DOACs), mainly apixaban, or the vitamin K antagonist (VKA) warfarin, could have impacted medium-term mortality in a cohort of SARS-CoV-2 patients. Methods: Among 1238 COVID-19 patients, hospitalized from 17 March 2020 to 15 June 2021, 247 survivors and 247 deceased within 90 days from hospitalization were matched 1:1 based on age, sex, and intensive care unit (ICU) admission within three days. Conditional logistic regression was used to estimate associations by means of odds ratio (OR) with a 95% confidence interval (CI). Results: A univariate regression analysis suggested that OAT, no differently from subcutaneous low-molecular-weight heparins (LMWHs) during hospitalization, has no significant impact (p value > 0.05) on medium-term mortality. A multivariate analysis, limited to baseline variables (i.e., comorbidities and pharmacotherapies at hospital admission) showing significant association (p < 0.05) to mortality in a univariate analysis, revealed that, compared to patients living at 90 days from hospitalization, deceased patients had cancer histories (OR 1.75, CI 1.06-2.90, p = 0.029) or suffered from asthma (OR 2.25, CI 1.13-4.47, p = 0.021). In contrast, heart failure (HF), atrial fibrillation (AF), arteriopathy, chronic obstructive pulmonary disease (COPD), and kidney failure (KF), which, in a univariate analysis, were found to be associated with the endpoint (p < 0.05), lost significance in a multivariate analysis. Therapy at admission with aldosterone antagonists also appeared to be associated with medium-term mortality (OR 2.49, CI 1.52-4.08, p < 0.001); whereas, vitamin D supplementation during hospitalization appeared to be beneficial. Although not conclusive, a search into the Eudravigilance database, combined with consulting a digital predictive platform (PLATO, polypharmacology platform prediction), suggested potential off-target activities, which might contribute to increasing the severity of SARS-CoV-2 infection. Conclusions: This retrospective clinical study furnished evidences of the impact of OAT, comorbidities and other pharmacological treatments on COVID-19 clinical course.
Collapse
Affiliation(s)
- Giorgia Dinoi
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| | - Maria Vittoria Togo
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| | - Pietro Guida
- Department of Internal Medicine, F. Miulli General Hospital, 70021 Bari, Italy; (P.G.); (F.M.)
| | - Caterina Deruvo
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| | - Francesco Samarelli
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| | - Paola Imbrici
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| | - Orazio Nicolotti
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| | - Annamaria De Luca
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| | - Franco Mastroianni
- Department of Internal Medicine, F. Miulli General Hospital, 70021 Bari, Italy; (P.G.); (F.M.)
| | - Antonella Liantonio
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| | - Cosimo Damiano Altomare
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| |
Collapse
|
25
|
Chang X, Lai Y, Zhao Y, Zhao J, Zhang Y, Qian X, Zhang G. Co-infections exacerbate inflammatory responses in COVID-19 patients, promoting coagulopathy and myocardial injury, leading to increased disease severity. Front Immunol 2025; 16:1522313. [PMID: 40046064 PMCID: PMC11879825 DOI: 10.3389/fimmu.2025.1522313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/05/2025] [Indexed: 05/13/2025] Open
Abstract
Objectives Severe COVID-19 infection is characterized by excessive inflammatory responses, hypercoagulation, and microvascular dysfunction. However, limited research has investigated the effects of co-infections on these characteristics in COVID-19 patients. This study aims to explore how co-infections influence inflammation, hypercoagulability, and microvascular dysfunction in hospitalized COVID-19 patients, and to assess their impact on disease progression. Methods This was a retrospective cohort study involving 630 COVID-19 inpatients who tested positive for SARS-CoV-2 RNA at Xi'an Ninth Hospital. The patients were categorized into two groups: a severe group (n = 176) and a mild group (n = 454). Additionally, they were further subdivided into a co-infected (n = 106) group and a non-co-infected group (n=524) based on the presence or absence of co-infections. Clinical characteristics and laboratory findings were analyzed and compared between the groups. Results In the co-infected group, 60 patients (56.6%) were classified as severe cases, and 15 (14.2%) died. By comparison, in the non-co-infected group, 97 patients (18.5%) were severe cases, with 4 (0.8%) deaths. The severity and mortality rates were significantly higher in co-infected patients compared to those non-co-infections. The severe and co-infected groups exhibited significantly higher levels of inflammatory cells, inflammatory factors, coagulation biomarkers, and myocardial injury markers compared to the mild and non-co-infected groups. Conversely, lymphocyte counts, RBC counts, HGB, HCT, TP, and ALB levels were significantly lower in the severe and co-infected groups than in the mild and non-co-infected groups. Furthermore, a notable positive correlation was observed among inflammatory factors, coagulation function, and myocardial injury biomarkers in COVID-19 patients. Conclusion Co-infections in COVID-19 patients can trigger severe inflammatory responses. This excessive inflammation may lead to coagulation disorders and myocardial injury, all of which are key contributors to disease progression and deterioration. Therefore, implementing infection prevention measures to minimize the spread of co-infections among hospitalized COVID-19 patients is crucial.
Collapse
Affiliation(s)
- Xiaoxia Chang
- Department of Clinical Laboratory, Ninth Hospital of Xi’an, Xi’an, Shannxi, China
| | - Yanjun Lai
- Department of Clinical Laboratory, Ninth Hospital of Xi’an, Xi’an, Shannxi, China
| | - Yingying Zhao
- Department of Pathology, Fenyang College of Shanxi Medical University, Fenyang, Shanxi, China
| | - Jing Zhao
- Department of Nephrotic Hemodialysis Center, Shannxi Provincial People’s Hospital, Xi’an, Shannxi, China
| | - Yunchao Zhang
- Department of Clinical Laboratory, Ninth Hospital of Xi’an, Xi’an, Shannxi, China
| | - Xiaotao Qian
- Department of Clinical Laboratory, Ninth Hospital of Xi’an, Xi’an, Shannxi, China
| | - Guochao Zhang
- Department of Clinical Laboratory, Ninth Hospital of Xi’an, Xi’an, Shannxi, China
| |
Collapse
|
26
|
Yao C, Dong Y, Zhou H, Zou X, Alhaskawi A, Ezzi SHA, Wang Z, Lai J, Kota VG, Abdulla MHAH, Liu Z, Abdalbary SA, Alenikova O, Lu H. COVID-19 and acute limb ischemia: latest hypotheses of pathophysiology and molecular mechanisms. J Zhejiang Univ Sci B 2025; 26:333-352. [PMID: 40274383 PMCID: PMC12021539 DOI: 10.1631/jzus.b2300512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 01/01/2024] [Indexed: 04/26/2025]
Abstract
Coronavirus disease 2019 (COVID-19) is a multi-system disease that can lead to various severe complications. Acute limb ischemia (ALI) has been increasingly recognized as a COVID-19-associated complication that often predicts a poor prognosis. However, the pathophysiology and molecular mechanisms underlying COVID-19-associated ALI remain poorly understood. Hypercoagulability and thrombosis are considered important mechanisms, but we also emphasize the roles of vasospasm, hypoxia, and acidosis in the pathogenesis of the disease. The angiotensin-converting enzyme 2 (ACE2) pathway, inflammation, and platelet activation may be important molecular mechanisms underlying these pathological changes induced by COVID-19. Furthermore, we discuss the hypotheses of risk factors for COVID-19-associated ALI from genetic, age, and gender perspectives based on our analysis of molecular mechanisms. Additionally, we summarize therapeutic approaches such as use of the interleukin-6 (IL-6) blocker tocilizumab, calcium channel blockers, and angiotensin-converting enzyme inhibitors, providing insights for the future treatment of coronavirus-associated limb ischemic diseases.
Collapse
Affiliation(s)
- Chengjun Yao
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yanzhao Dong
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Haiying Zhou
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xiaodi Zou
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310005, China
| | - Ahmad Alhaskawi
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Sohaib Hasan Abdullah Ezzi
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Department of Orthopaedics, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Zewei Wang
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jingtian Lai
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Vishnu Goutham Kota
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | | | - Zhenfeng Liu
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Sahar Ahmed Abdalbary
- Department of Orthopaedic Physical Therapy, Faculty of Physical Therapy, Nahda University, Beni Suef 2711860, Egypt
| | - Olga Alenikova
- Republic Scientific Practical Center of Neurology and Neurosurgery, Ministry of Health of the Republic of Belarus, Minsk 220004, Belarus
| | - Hui Lu
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
27
|
Sabit H, Arneth B, Altrawy A, Ghazy A, Abdelazeem RM, Adel A, Abdel-Ghany S, Alqosaibi AI, Deloukas P, Taghiyev ZT. Genetic and Epigenetic Intersections in COVID-19-Associated Cardiovascular Disease: Emerging Insights and Future Directions. Biomedicines 2025; 13:485. [PMID: 40002898 PMCID: PMC11852909 DOI: 10.3390/biomedicines13020485] [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: 12/19/2024] [Revised: 01/23/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
The intersection of COVID-19 and cardiovascular disease (CVD) has emerged as a significant area of research, particularly in understanding the impact of antiplatelet therapies like ticagrelor and clopidogrel. COVID-19 has been associated with acute cardiovascular complications, including myocardial infarction, thrombosis, and heart failure, exacerbated by the virus's ability to trigger widespread inflammation and endothelial dysfunction. MicroRNAs (miRNAs) play a critical role in regulating these processes by modulating the gene expressions involved in platelet function, inflammation, and vascular homeostasis. This study explores the potential of miRNAs such as miR-223 and miR-126 as biomarkers for predicting resistance or responsiveness to antiplatelet therapies in COVID-19 patients with cardiovascular disease. Identifying miRNA signatures linked to drug efficacy could optimize treatment strategies for patients at high risk of thrombotic events during COVID-19 infection. Moreover, understanding miRNA-mediated pathways offers new insights into how SARS-CoV-2 exacerbates CVD, particularly through mechanisms like cytokine storms and endothelial damage. The findings of this research could lead to personalized therapeutic approaches, improving patient outcomes and reducing mortality in COVID-19-associated cardiovascular events. With global implications, this study addresses the urgent need for effective management of CVD in the context of COVID-19, focusing on the integration of molecular biomarkers to enhance the precision of antiplatelet therapy.
Collapse
Affiliation(s)
- Hussein Sabit
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Borros Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Afaf Altrawy
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Aysha Ghazy
- Department of Agri-Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Rawan M. Abdelazeem
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Amro Adel
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Shaimaa Abdel-Ghany
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Amany I. Alqosaibi
- Department of Biology, College of Science, Imam Abdulrahman bin Faisal University, Dammam 31441, Saudi Arabia
| | - Panos Deloukas
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 4NS, UK;
| | - Zulfugar T. Taghiyev
- Department of Cardiovascular Surgery, Hospital of the Universities of Giessen and Marburg (UKGM), Justus Liebig University Giessen, 35392 Giessen, Germany
| |
Collapse
|
28
|
Nakahashi T, Tada H, Sakata K, Inaba S, Hashimoto M, Nomura A, Azuma S, Hirata M, Ito H, Takamura M. Impact of the Geriatric Nutritional Risk Index on In-hospital Thrombosis and Mortality in Patients Hospitalized with COVID-19. Intern Med 2025; 64:519-526. [PMID: 38987193 PMCID: PMC11904464 DOI: 10.2169/internalmedicine.2977-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 05/24/2024] [Indexed: 07/12/2024] Open
Abstract
Objective To determine whether nutritional status is related to the incidence of thrombosis and mortality in patients with coronavirus disease 2019 (COVID-19). Methods A total of 496 consecutive patients who were admitted and diagnosed with COVID-19 between April 2020 and March 2023 were retrospectively analyzed. The geriatric nutritional risk index (GNRI) on admission was calculated as follows: 14.89×serum albumin (g/dL) +41.7×body mass index/22. Patients were divided into two groups according to the median GNRI values. The endpoint of this study was a composite of in-hospital thrombotic events and mortality. Results The median GNRI value was 99.3. Patients in the low GNRI (≤99.3) group were older (75±21 vs. 51±20 years, p<0.001) and more likely to be female (55.6% vs. 41.1%, p<0.05). In addition, patients with a low GNRI often exhibited hypertension (43.5% vs. 28.2%, p<0.001) and had a history of cardiovascular disease (34.3% vs. 14.5%, p<0.001). Under these conditions, the median D-dimer levels on admission were significantly higher in patients with a low GNRI (0.90 μg/mL; interquartile range (IQR), 0.49-1.64 μg/mL) than those with high GNRI (0.36 μg/mL; IQR, 0.26-0.51 μg/mL, p<0.001). During hospitalization, the composite endpoint was observed in 32 patients. In the logistic regression analysis, a low GNRI was significantly associated with the composite endpoint adjusted using inverse probability of treatment weighting (odds ratio, 3.24; 95% confidence interval: 1.51-6.93, p<0.05). Conclusion Assessment of the GNRI provides useful information for predicting in-hospital thrombosis and mortality in COVID-19 patients.
Collapse
Affiliation(s)
| | - Hayato Tada
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Science, Japan
| | - Kenji Sakata
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Science, Japan
| | - Shota Inaba
- Department of Cardiology, Takaoka City Hospital, Japan
| | | | - Akihiro Nomura
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Science, Japan
| | - Shigeru Azuma
- Department of Internal Medicine, Takaoka City Hospital, Japan
| | | | - Hiroyuki Ito
- Department of Gastroenterology, Takaoka City Hospital, Japan
| | - Masayuki Takamura
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Science, Japan
| |
Collapse
|
29
|
Ayyoub S, Dhillon NK, Tura-Ceide O. Genetics of Long COVID: Exploring the Molecular Drivers of Persistent Pulmonary Vascular Disease Symptoms. Infect Dis Rep 2025; 17:15. [PMID: 39997467 PMCID: PMC11855385 DOI: 10.3390/idr17010015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/24/2025] [Accepted: 02/08/2025] [Indexed: 02/26/2025] Open
Abstract
Background/ Objectives: Long COVID or post-acute sequelae of SARS-CoV-2 infection (PASC) are symptoms that manifest despite passing the acute infection phase. These manifestations encompass a wide range of symptoms, the most common being fatigue, shortness of breath, and cognitive dysfunction. Genetic predisposition is clearly involved in the susceptibility of individuals to developing these persistent symptoms and the variation in the severity and forms. This review summarizes the role of genetic factors and gene polymorphisms in the development of major pulmonary vascular disorders associated with long COVID. Methods: A comprehensive review of current literature was conducted to examine the genetic contributions to pulmonary complications following SARS-CoV-2 infection. Studies investigating genetic polymorphisms linked to pulmonary hypertension, pulmonary thromboembolism, and pulmonary vascular endothelialitis were reviewed and summarized. Results: Findings show that specific genetic variants contribute to increased susceptibility to pulmonary vascular complications in long COVID patients. Variants associated with endothelial dysfunction, coagulation pathways, and inflammatory responses have been implicated in the development of pulmonary hypertension and thromboembolic events. Genetic predispositions influencing vascular integrity and immune responses appear to influence disease severity and progression. Conclusions: Understanding these mechanisms and genetic predispositions could pave the way for targeted therapeutic interventions to alleviate the burden on patients experiencing long COVID.
Collapse
Affiliation(s)
- Sana Ayyoub
- Department of Medical Sciences, Faculty of Medicine, University of Girona, 17004 Girona, Spain;
| | - Navneet Kaur Dhillon
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Kansas Medical Center, Mail Stop 3007, 3901 Rainbow Blvd, Kansas City, KS 66160, USA;
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Olga Tura-Ceide
- Translational Research Group on Cardiovascular Respiratory Diseases (CAREs), Girona Biomedical Research Institute (IDIBGI-CERCA), Martí i Julià, Hospital Park Building M2, 17190 Salt, Spain
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029 Madrid, Spain
| |
Collapse
|
30
|
Nalinthasnai N, Thammasudjarit R, Tassaneyasin T, Eksombatchai D, Sungkanuparph S, Boonsarngsuk V, Sutherasan Y, Junhasavasdikul D, Theerawit P, Petnak T. Unsupervised machine learning clustering approach for hospitalized COVID-19 pneumonia patients. BMC Pulm Med 2025; 25:70. [PMID: 39923003 PMCID: PMC11807335 DOI: 10.1186/s12890-025-03536-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: 10/18/2024] [Accepted: 01/28/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Identification of distinct clinical phenotypes of diseases can guide personalized treatment. This study aimed to classify hospitalized COVID-19 pneumonia subgroups using an unsupervised machine learning approach. METHODS We included hospitalized COVID-19 pneumonia patients from July to September 2021. K-means clustering, an unsupervised machine learning method, was performed to identify clinical phenotypes based on clinical and laboratory variables collected within 24 hours of admission. Variables were normalized before clustering to ensure equal contribution to the analysis. The optimal number of clusters was determined using the elbow method and Silhouette scores. Cox proportional hazard models were used to compare the risk of intubation and 90-day mortality across the identified clusters. RESULTS Three clinically distinct clusters were identified among 538 hospitalized COVID-19 pneumonia patients. Cluster 1 (N = 27) consisted predominantly of males and showed significantly elevated serum liver enzymes and LDH levels. Cluster 2 (N = 370) was characterized by lower chest x-ray scores and higher serum albumin levels. Cluster 3 (N = 141) was characterized by older age, diabetes mellitus, higher chest x-ray scores, more severe vital signs, higher creatinine levels, lower hemoglobin levels, lower lymphocyte counts, higher C-reactive protein, higher D-dimer, and higher LDH levels. When compared to cluster 2, cluster 3 was significantly associated with increased risk of 90-day mortality (HR, 6.24; 95% CI, 2.42-16.09) and intubation (HR, 5.26; 95% CI 2.37-11.72). In contrast, cluster 1 had a 100% survival rate with a non-significant increase in intubation risk compared to cluster 2 (HR, 1.40, 95% CI, 0.18-11.04). CONCLUSIONS We identified three distinct clinical phenotypes of COVID-19 pneumonia patients, with cluster 3 associated with an increased risk of respiratory failure and mortality. These findings may guide tailored clinical management strategies.
Collapse
Affiliation(s)
- Nuttinan Nalinthasnai
- Division of Pulmonary and Pulmonary Critical Care Medicine, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| | | | - Tanapat Tassaneyasin
- Division of Pulmonary and Pulmonary Critical Care Medicine, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
- Faculty of Medicine Ramathibodi Hospital, Chakri Naruebodindra Medical Institute, Mahidol University, Samutprakan, Thailand
| | - Dararat Eksombatchai
- Division of Pulmonary and Pulmonary Critical Care Medicine, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Somnuek Sungkanuparph
- Faculty of Medicine Ramathibodi Hospital, Chakri Naruebodindra Medical Institute, Mahidol University, Samutprakan, Thailand
| | - Viboon Boonsarngsuk
- Division of Pulmonary and Pulmonary Critical Care Medicine, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Yuda Sutherasan
- Division of Pulmonary and Pulmonary Critical Care Medicine, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Detajin Junhasavasdikul
- Division of Pulmonary and Pulmonary Critical Care Medicine, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Pongdhep Theerawit
- Division of Critical Care Medicine, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Tananchai Petnak
- Division of Pulmonary and Pulmonary Critical Care Medicine, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand.
| |
Collapse
|
31
|
Cosgrave D, McNicholas B, Hanley C, Sheehan JR, Calpin P, Kernan M, Murphy D, Alvarez-Iglesias A, Ferguson J, Giacomini C, Greene C, Cody C, McGeary S, Murphy M, Fitzgerald M, Curley G, Dixon B, Smith RJ, Masterson C, O'Toole D, van Haren F, Laffey JG. Can nebulised heparin reduce acute lung injury in patients with SARS‑CoV‑2 requiring advanced respiratory support in Ireland: the CHARTER‑Ireland phase Ib/IIa, randomised, parallel-group, open-label study. Intensive Care Med Exp 2025; 13:15. [PMID: 39920521 PMCID: PMC11806160 DOI: 10.1186/s40635-025-00727-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 01/29/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Nebulised unfractionated heparin may attenuate COVID-19 ARDS by reducing pulmonary microvascular thrombosis, blocking SARS-CoV-2 entry into cells, and decreasing lung inflammation. COVID-19 patients with a raised D-dimer have areas of pulmonary hypoperfusion on CT perfusion scans of the lung and have increased mortality risk. METHODS This was a phase Ib/IIa open-label multi-centre, randomised controlled trial. The study was designed to evaluate whether nebulised unfractionated heparin decreased D-dimer concentrations, with safety as a co-primary outcome. RESULTS Forty patients were recruited, with 20 patients into each group. Mean age was 56.6 (SD 11.5) in the heparin group and 51.3 (SD 14.7) in the standard care group, while 60% of participants were male. There was no change in D-dimers from baseline to day 10 (heparin group mean change - 316.5, [SD 1840.3] and control group mean change - 321.7 [SD 3589.4]; p = 0.996). Fourteen patients suffered at least one serious adverse event, 9 patients the Heparin group and 5 in the control group. Eight patients had one or more bleeding events, 5 in the heparin group and 3 in the control group, but were no cases of pulmonary bleeding, of severe haemorrhage or of heparin-induced thrombocytopenia. Patients receiving heparin therapy had lower PaO2/FiO2 ratios, increased oxygenation indices, and decreased ROX index profiles, up to day 10. The time to separation from respiratory support, and the time to ICU or hospital discharge was similar in both groups. There were 3 deaths in the Heparin group and 2 in the control group. CONCLUSIONS Nebulised unfractionated heparin was safe and well tolerated, but did not reduce D-dimer concentrations, and worsened oxygenation indices in patients with COVID-19 ARDS.
Collapse
Affiliation(s)
- David Cosgrave
- Department of Anaesthesia and Intensive Care Medicine, University Hospital Galway, Galway, Ireland
- Anaesthesia and Intensive Care Medicine, School of Medicine, University of Galway, Galway, Ireland
| | - Bairbre McNicholas
- Department of Anaesthesia and Intensive Care Medicine, University Hospital Galway, Galway, Ireland
- Anaesthesia and Intensive Care Medicine, School of Medicine, University of Galway, Galway, Ireland
| | - Ciara Hanley
- Department of Anaesthesia and Intensive Care Medicine, University Hospital Galway, Galway, Ireland
- Anaesthesia and Intensive Care Medicine, School of Medicine, University of Galway, Galway, Ireland
| | - John Robert Sheehan
- Department of Anaesthesia and Intensive Care Medicine, University Hospital Galway, Galway, Ireland
| | - Padraig Calpin
- Department of Anaesthesia and Intensive Care Medicine, University Hospital Galway, Galway, Ireland
| | - Maeve Kernan
- Anaesthesia and Intensive Care Medicine, School of Medicine, University of Galway, Galway, Ireland
| | - Darragh Murphy
- Anaesthesia and Intensive Care Medicine, School of Medicine, University of Galway, Galway, Ireland
| | | | - John Ferguson
- HRB Clinical Research Facility, School of Medicine, University of Galway, Galway, Ireland
- School of Mathematical & Statistical Sciences, University of Galway, Galway, Ireland
| | - Camilla Giacomini
- Department of Anaesthesia and Intensive Care Medicine, University Hospital Galway, Galway, Ireland
| | - Christine Greene
- Department of Anaesthesia and Intensive Care Medicine, Connolly Memorial Hospital Blanchardstown, Dublin, Ireland
| | - Catriona Cody
- Department of Anaesthesia and Intensive Care Medicine, Connolly Memorial Hospital Blanchardstown, Dublin, Ireland
| | - Shane McGeary
- Department of Anaesthesia and Intensive Care Medicine, Connolly Memorial Hospital Blanchardstown, Dublin, Ireland
| | - Marion Murphy
- Department of Anaesthesia and Intensive Care Medicine, University Hospital Galway, Galway, Ireland
| | - Marianne Fitzgerald
- Department of Anaesthesia and Intensive Care Medicine, Limerick University Hospital, Limerick, Ireland
| | - Gerard Curley
- Department of Anaesthesia and Intensive Care Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Barry Dixon
- Department of Critical Care Medicine, St Vincent's Hospital Melbourne, Fitzroy, VIC, Australia
| | - Roger J Smith
- Department of Critical Care Medicine, St Vincent's Hospital Melbourne, Fitzroy, VIC, Australia
| | - Claire Masterson
- Anaesthesia and Intensive Care Medicine, School of Medicine, University of Galway, Galway, Ireland
| | - Daniel O'Toole
- Anaesthesia and Intensive Care Medicine, School of Medicine, University of Galway, Galway, Ireland
| | - Frank van Haren
- Intensive Care Unit, St George Hospital, Sydney, Australia
- College of Health and Medicine, Australian National University, Canberra, Australia
| | - John G Laffey
- Department of Anaesthesia and Intensive Care Medicine, University Hospital Galway, Galway, Ireland.
- Anaesthesia and Intensive Care Medicine, School of Medicine, University of Galway, Galway, Ireland.
| |
Collapse
|
32
|
Zhu W, Zheng Y, Yu M, Witman N, Zhou L, Wei J, Zhang Y, Topchyan P, Nguyen C, Wang D, Janecke R, Padmanabhan A, Baumann Kreuziger L, White GC, Hari P, Gu T, Fields AT, Kornblith LZ, Aster R, Zhu J, Cui W, Jobe S, Graham MB, Wang D, Wen R. Prothrombotic antibodies targeting the spike protein's receptor-binding domain in severe COVID-19. Blood 2025; 145:635-647. [PMID: 39576992 PMCID: PMC11811936 DOI: 10.1182/blood.2024025010] [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: 04/17/2024] [Revised: 10/21/2024] [Accepted: 11/06/2024] [Indexed: 11/24/2024] Open
Abstract
ABSTRACT Thromboembolic complication is common in severe coronavirus disease 2019 (COVID-19), leading to an investigation into the presence of prothrombotic antibodies akin to those found in heparin-induced thrombocytopenia (HIT). In a study of samples from 130 hospitalized patients, collected 3.6 days after COVID-19 diagnosis, 80% had immunoglobulin G (IgG) antibodies recognizing complexes of heparin and platelet factor 4 (PF4; PF4/H), and 41% had antibodies inducing PF4-dependent P-selectin expression in CpG oligodeoxynucleotide-treated normal platelets. Unlike HIT, both PF4/H-reactive and platelet-activating antibodies were found in patients with COVID-19 regardless of recent heparin exposure. Notably, PF4/H-reactive IgG antibodies correlated with those targeting the receptor-binding domain (RBD) of the severe acute respiratory syndrome coronavirus 2 spike protein. Moreover, introducing exogenous RBD to or removing RBD-reactive IgG from COVID-19 plasma or IgG purified from COVID-19 plasma significantly reduced their ability to activate platelets. RBD-specific antibodies capable of platelet activation were cloned from peripheral blood B cells of patients with COVID-19. These antibodies possessed sequence motifs in the heavy-chain complementarity-determining region 3 (HCDR3), resembling those identified in pathogenic HIT antibodies. Furthermore, IgG+ B cells having these HCDR3 signatures were markedly expanded in patients with severe COVID-19. Importantly, platelet-activating antibodies present in patients with COVID-19 were associated with a specific elevation of platelet α-granule proteins in the plasma and showed a positive correlation with markers for inflammation and tissue damage, suggesting a functionality of these antibodies in patients. The demonstration of functional and structural similarities between certain RBD-specific antibodies in patients with COVID-19 and pathogenic antibodies typical of HIT suggests a novel mechanism by which RBD-specific antibodies might contribute to thrombosis in COVID-19.
Collapse
Affiliation(s)
- Wen Zhu
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | | | - Mei Yu
- Versiti Blood Research Institute, Milwaukee, WI
| | - Nathan Witman
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Lu Zhou
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Jianhui Wei
- Versiti Blood Research Institute, Milwaukee, WI
- Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Yongguang Zhang
- Versiti Blood Research Institute, Milwaukee, WI
- Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Paytsar Topchyan
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Christine Nguyen
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - David Wang
- School of Art and Science Undergraduate Program, Washington University in St. Louis, St. Louis, MO
| | - Rae Janecke
- Versiti Blood Research Institute, Milwaukee, WI
| | - Anand Padmanabhan
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Lisa Baumann Kreuziger
- Versiti Blood Research Institute, Milwaukee, WI
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | | | - Parameswaran Hari
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Tongjun Gu
- Versiti Blood Research Institute, Milwaukee, WI
| | - Alexander T. Fields
- Department of Surgery, University of California San Francisco, San Francisco, CA
| | - Lucy Z. Kornblith
- Department of Surgery, University of California San Francisco, San Francisco, CA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA
| | - Richard Aster
- Versiti Blood Research Institute, Milwaukee, WI
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Jieqing Zhu
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI
| | - Weiguo Cui
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Shawn Jobe
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI
| | - Mary Beth Graham
- Division of Infectious Disease, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Demin Wang
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Renren Wen
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
33
|
Mansor NF, Abdul Halim Zaki I, Kiok LC, Seng EK, Ravi T, Pathmanathan M, Goh KW, Ming LC, Razi P, Zulkifly HH. The prevalence of thromboembolic events among COVID-19 patients admitted to a single centre intensive care unit (ICU): an epidemiological study from a Malaysian population. J Pharm Policy Pract 2025; 18:2449044. [PMID: 39917475 PMCID: PMC11800336 DOI: 10.1080/20523211.2024.2449044] [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: 04/04/2024] [Accepted: 12/28/2024] [Indexed: 02/09/2025] Open
Abstract
Introduction Thromboembolic (TE) complications in COVID-19 patients are rising globally, contributing significantly to mortality, particularly in severe cases. However, their prevalence, characteristics, and impact on mortality in Malaysia remain unclear. Objectives This study aimed to determine the prevalence of thromboembolic (TE) events and associated mortality among COVID-19 patients admitted within a single centre intensive care unit (ICU). The proportions of patients with TE events who died, and factors associated with TE events were explored. Methods In this retrospective cohort study, patients with PCR confirmed SARS-CoV-2 virus and who received thromboprophylaxis within February 2020-2021 were included. TE event is a combination of venous [(deep vein thrombosis (DVT), pulmonary embolism (PE)] and arterial (myocardial infarction (MI), stroke) thromboembolism. Results Mean (SD) age 56.6 (13.7), 63.5% were male, 61.6% Malays, median (IQR) 7 (3-14) days of ICU stay, 64.2%, 53.2% and 20.9% had underlying hypertension, diabetes and obesity respectively. In total, 240 (44.9%) developed TE event. Significantly higher proportions of COVID-19 patients who developed complications of DVT (2.5% vs. 0.2%; p = 0.013), PE (47.5% vs 34.0%; p = 0.006), stroke (12.3% vs. 1.5; p<0.001) and MI (16.4% vs. 4.6%; p<0.001) died. Predictors of TE events were age [HR 1.01 (95% CI 1.00-1.02)], obesity [HR 1.98 (95% CI 1.51-2.6)], D-dimer [HR 1.01 (95% CI 1.00-1.01)], and duration of ICU stay [HR 0.98 (95% CI 0.97-0.99)]. Conclusion In severely ill COVID-19 patients, TE complications were common, and patients with DVT, PE, stroke, or MI faced increased mortality, even with thromboprophylaxis. Age, obesity, elevated D-Dimer levels, and longer ICU stays were significant predictors of TE events. Considering these findings, a more aggressive approach, combining thromboprophylaxis with enhanced anti-inflammatory treatments, may be necessary for high-risk COVID-19 ICU patients to reduce TE events and mortality.
Collapse
Affiliation(s)
| | - Izzati Abdul Halim Zaki
- Faculty of Pharmacy, Universiti Teknologi MARA, UiTM Kampus Puncak Alam, Puncak Alam, Malaysia
- Cardiology Therapeutics Research Group, Universiti Teknologi MARA, UiTM Kampus Puncak Alam, Puncak Alam, Malaysia
| | - Lee Chew Kiok
- Anesthesiology and Intensive Care Department, Sungai Buloh Hospital, Sungai Buloh, Malaysia
| | - Eng Kar Seng
- Anesthesiology and Intensive Care Department, Sungai Buloh Hospital, Sungai Buloh, Malaysia
| | - Tharmini Ravi
- Clinical Research Center, Sungai Buloh Hospital, Sungai Buloh, Malaysia
| | - Mohan Pathmanathan
- Institute for Clinical Research, National Institutes of Health, Shah Alam, Malaysia
| | - Khang Wen Goh
- Faculty of Data Science and Information Technology, INTI International University, Nilai, Malaysia
| | - Long Chiau Ming
- School of Medical and Life Sciences, Sunway University, Sunway City, Malaysia
- Datta Meghe College of Pharmacy, Datta Meghe Institute of Higher Education and Research (deemed to be University), Sawangi (M), Wardha, India
| | - Pakhrur Razi
- Center of Disaster Monitoring and Earth Observation, Physics Department, Universitas Negeri Padang, Padang, Indonesia
| | - Hanis Hanum Zulkifly
- Faculty of Pharmacy, Universiti Teknologi MARA, UiTM Kampus Puncak Alam, Puncak Alam, Malaysia
- Cardiology Therapeutics Research Group, Universiti Teknologi MARA, UiTM Kampus Puncak Alam, Puncak Alam, Malaysia
| |
Collapse
|
34
|
Arnold JR, Yeo JL, Budgeon CA, Shergill S, England R, Shiwani H, Artico J, Moon JC, Gorecka M, Roditi G, Morrow A, Mangion K, Shanmuganathan M, Miller CA, Chiribiri A, Alzahir M, Ramirez S, Lin A, Swoboda PP, McDiarmid AK, Sykes R, Singh T, Bucciarelli-Ducci C, Dawson D, Fontana M, Manisty C, Treibel TA, Levelt E, Young R, McConnachie A, Neubauer S, Piechnik SK, Davies RH, Ferreira VM, Dweck MR, Berry C, McCann GP, Greenwood JP. Myocardial ischaemia following COVID-19: a cardiovascular magnetic resonance study. Int J Cardiovasc Imaging 2025; 41:247-256. [PMID: 39738791 PMCID: PMC11811239 DOI: 10.1007/s10554-024-03304-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 11/27/2024] [Indexed: 01/02/2025]
Abstract
The pathophysiology of myocardial injury following COVID-19 remains uncertain. COVID-HEART was a prospective, multicentre study utilising cardiovascular magnetic resonance (CMR) to characterise COVID-related myocardial injury. In this pre-specified analysis, the objectives were to examine (1) the frequency of myocardial ischaemia following COVID-19, and (2) the association between ischaemia and myocardial injury. We studied 59 patients hospitalised with COVID-19 and elevated serum troponin (COVID + /troponin + , age 61 ± 11 years) and 37 control subjects without COVID-19 or elevated troponin and similar by age and cardiovascular comorbidities (COVID -/comorbidity + , 64 ± 10 years). Subjects underwent multi-parametric CMR (comprising assessment of ventricular volumes, stress perfusion, T1/T2 mapping and scar). The primary endpoint was the frequency of inducible myocardial ischaemia. Inducible ischaemia was evident in 11 (19%) COVID + /troponin + patients and in 8 (22%) control subjects (p = 0.72). In COVID + /troponin + patients with ischaemia, epicardial coronary disease pattern ischaemia was present in eight patients and microvascular disease pattern, in three patients. There was no significant difference in the frequency of inducible ischaemia in COVID + /troponin + patients with previous myocardial infarction and/or revascularisation compared to those without (2/12 [17%] vs. 9/47 [19%] respectively, p = 0.84), or in those with and without scar (7/27 [26%] vs. 4/32 [13%] respectively, p = 0.19). Myocardial ischaemia was present in ~ 20% of patients recently hospitalised with COVID-19 and with elevated cardiac troponin, but this was not different to matched comorbid controls. This finding coupled with the lack of an association between ischaemia and myocardial scar suggests that coronary artery abnormalities are unlikely to be the predominant mechanism underlying COVID-19 induced myocardial injury.
Collapse
Affiliation(s)
- J Ranjit Arnold
- University of Leicester and The NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK.
| | - Jian L Yeo
- University of Leicester and The NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Charley A Budgeon
- University of Leicester and The NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
- Cardiovascular Epidemiology Research Centre, School of Population and Global Health, University of Western Australia, Perth, Australia
| | - Simran Shergill
- University of Leicester and The NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Rachel England
- University of Leicester and The NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Hunain Shiwani
- Institute of Cardiovascular Science, University College London, London, UK
| | - Jessica Artico
- Institute of Cardiovascular Science, University College London, London, UK
| | - James C Moon
- Institute of Cardiovascular Science, University College London, London, UK
| | - Miroslawa Gorecka
- Institute of Cardiovascular and Metabolic Medicine, University of Leeds, and Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Giles Roditi
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Andrew Morrow
- Institute of Cardiovascular and Medical Sciences and British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Kenneth Mangion
- Institute of Cardiovascular and Medical Sciences and British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Mayooran Shanmuganathan
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford Centre for Clinical Magnetic Resonance Research, Oxford, UK
- British Heart Foundation Centre of Research Excellence, Oxford, UK
- Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Christopher A Miller
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Amedeo Chiribiri
- School of Biomedical Engineering and Imaging Sciences, King's College London, BHF Centre of Excellence and The NIHR Biomedical Research Centre at Guy's and St. Thomas' NHS Foundation Trust, The Rayne Institute, St. Thomas' Hospital, London, UK
| | - Mohammed Alzahir
- Institute of Cardiovascular Science, University College London, London, UK
| | - Sara Ramirez
- Institute of Cardiovascular Science, University College London, London, UK
| | - Andrew Lin
- Institute of Cardiovascular Science, University College London, London, UK
| | - Peter P Swoboda
- Institute of Cardiovascular and Metabolic Medicine, University of Leeds, and Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Adam K McDiarmid
- Adult Congenital and Paediatric Heart Unit, Freeman Hospital, Newcastle Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Robert Sykes
- Institute of Cardiovascular and Medical Sciences and British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Trisha Singh
- University of Edinburgh and British Heart Foundation Centre for Cardiovascular Science, Edinburgh, UK
| | - Chiara Bucciarelli-Ducci
- School of Biomedical Engineering and Imaging Sciences, King's College London, BHF Centre of Excellence and The NIHR Biomedical Research Centre at Guy's and St. Thomas' NHS Foundation Trust, The Rayne Institute, St. Thomas' Hospital, London, UK
- Royal Brompton and Harefield Hospitals, London, UK
- Guys' and St Thomas NHS Trust, London, UK
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Trust, Bristol, UK
| | - Dana Dawson
- Department of Cardiology, Aberdeen Cardiovascular and Diabetes Centre, Aberdeen Royal Infirmary and University of Aberdeen, Aberdeen, UK
| | - Marianna Fontana
- Division of Medicine, Royal Free Hospital, University College London, London, UK
| | - Charlotte Manisty
- Institute of Cardiovascular Science, University College London, London, UK
| | - Thomas A Treibel
- Institute of Cardiovascular Science, University College London, London, UK
| | - Eylem Levelt
- Institute of Cardiovascular and Metabolic Medicine, University of Leeds, and Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Robin Young
- Robertson Centre for Biostatistics, Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Alex McConnachie
- Robertson Centre for Biostatistics, Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford Centre for Clinical Magnetic Resonance Research, Oxford, UK
| | - Stefan K Piechnik
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford Centre for Clinical Magnetic Resonance Research, Oxford, UK
| | - Rhodri H Davies
- Institute of Cardiovascular Science, University College London, London, UK
| | - Vanessa M Ferreira
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford Centre for Clinical Magnetic Resonance Research, Oxford, UK
| | - Marc R Dweck
- University of Edinburgh and British Heart Foundation Centre for Cardiovascular Science, Edinburgh, UK
| | - Colin Berry
- Institute of Cardiovascular and Medical Sciences and British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Gerry P McCann
- University of Leicester and The NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - John P Greenwood
- Institute of Cardiovascular and Metabolic Medicine, University of Leeds, and Leeds Teaching Hospitals NHS Trust, Leeds, UK
- Baker Heart and Diabetes Institute, Melbourne, Australia
| |
Collapse
|
35
|
Selvarajan S, John JS, Tharyan P, Kirubakaran R, Singh B, George B, Mathew JL, Rupali P. Therapeutic Versus Non-Therapeutic Dose Anticoagulation in COVID-19 Infection: A Systematic Review and Meta-analysis of Randomised Controlled Trials. EJHAEM 2025; 6:e1100. [PMID: 39935487 PMCID: PMC11811394 DOI: 10.1002/jha2.1100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/14/2024] [Accepted: 12/19/2024] [Indexed: 02/13/2025]
Abstract
Background Abnormal coagulation and thrombotic complications prompted many guidelines to recommend thromboprophylaxis for patients hospitalised with COVID-19, but the dose required for prophylaxis remains unclear. This systematic review (SR) analyses the safety and efficacy of therapeutic dose anticoagulation (TDA) versus non-therapeutic dose anticoagulation (NDA) in COVID-19 patients. Methods According to the Cochrane Handbook of Systematic Review of Interventions, we performed an SR. The protocol is registered in Prospero (CRD42021269197, date 12 August 2021). Results In this SR of 18 studies, TDA was shown to reduce all-cause mortality (risk ratio [RR] 0.83; 95% confidence interval [95% CI] 0.70, 0.99) in COVID-19 infection. TDA also reduced thrombosis (RR 0.55; 95% CI 0.48, 0.72) but increased major bleeding (RR 1.87; 95% CI 1.29, 2.69). A stratified analysis according to severity revealed that, in non-critical patients, TDA resulted in mortality benefit (RR 0.79; 95% CI 0.67, 0.94). In critical patients, TDA did not affect all-cause mortality (RR 1.03; 95% CI 0.89, 1.18) but reduced thrombosis (RR 0.65; 95% CI 0.48, 0.86) and increased major bleeding (RR 1.85; 95% CI 1.06, 3.23). Conclusion TDA significantly reduced all-cause mortality and thrombosis in non-critical COVID-19 patients at the expense of increased major bleeding. In critical COVID-19, this mortality benefit was not observed.
Collapse
Affiliation(s)
- Sushil Selvarajan
- Department of Clinical HaematologyChristian Medical CollegeVelloreIndia
| | - Jisha Sara John
- Department of Infectious DiseasesChristian Medical CollegeVelloreIndia
| | - Prathap Tharyan
- Prof. BV Moses Centre for Evidence Informed HealthcareChristian Medical CollegeVelloreIndia
| | - Richard Kirubakaran
- Prof. BV Moses Centre for Evidence Informed HealthcareChristian Medical CollegeVelloreIndia
| | - Bhagteshwar Singh
- Department of Infectious DiseasesChristian Medical CollegeVelloreIndia
- Department of Clinical Infection Microbiology and ImmunologyInstitute of Infection Veterinary & Ecological SciencesUniversity of LiverpoolLiverpoolUK
- Centre for Evidence Synthesis in Global Health, Department of Clinical SciencesLiverpool School of Tropical MedicineLiverpoolUK
| | - Biju George
- Department of Clinical HaematologyChristian Medical CollegeVelloreIndia
| | - Joseph L. Mathew
- Advanced Paediatrics CentrePostgraduate Institute of Medical Education and ResearchChandigarhIndia
| | - Priscilla Rupali
- Department of Infectious DiseasesChristian Medical CollegeVelloreIndia
| |
Collapse
|
36
|
Marcos-Neira P, Morales-Indiano C, Fernández-Caballero M, Tomasa-Irriguible T, Bordejé-Laguna L, Ruíz-Artola V. Anti-Xa activity below range is related to thrombosis in patients with severe COVID-19. Med Intensiva 2025; 49:78-87. [PMID: 39054216 DOI: 10.1016/j.medine.2024.07.003] [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: 04/16/2024] [Revised: 06/05/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024]
Abstract
OBJECTIVE We aimed to anlayse the relationship between anti-Xa activity below range and thomboembolic events. DESIGN Single center prospective observational longitudinal cohort study (February-November 2021). SETTING Patients admitted to the ICU of a University Hospital. PARTICIPANTS Patients with severe COVID-19 pneumoniae. INTERVENTIONS Enoxaparin was used for prophylactic and therapeutic anticoagulation. Enoxaparin dosing and dose adjustment were based on anti-Xa activity according to the hospital protocol. MAIN VARIABLES OF INTEREST Target: thomboembolic events. PREDICTORS demographics, pharmacotherapy, anti-Xa measurements, clinical data, and laboratory results. Logistic regression was used to identify independent risk factors for thomboembolic events. RESULTS Data were available for 896 serum anti-Xa measurements from 228 subjects. Overall, 71.9% were male, with a median age of 62. Most patients needed invasive mechanical ventilation (87.7%) and mortality was 24.1%. A total of 28.9% new thomboembolic events were diagnosed. There were 27.1% anti-Xa measesurements below range. When multivariable logistic regression analysis was performed anti-Xa activity below range (RR, 4.2; p = 0.000), C-reactive protein (25 mg/L increase) (RR, 1.14; p = 0.005) and D-dimer (1000 ng/L increase) (RR, 1.06; p = 0.002) were the independent factors related to new thomboembolic events in patients with severe COVID-19. CONCLUSIONS Anti-Xa activity below range, C-reactive protein and D-dimer were the independent factors related to thomboembolic events in patients with severe COVID-19. Purposely designed clinical trials should be carried out to confirm the benefit of an anti-Xa monitoring.
Collapse
Affiliation(s)
- Pilar Marcos-Neira
- Intensive Care Unit, Germans Trias i Pujol University Hospital, Badalona, Barcelona, Spain.
| | - Cristian Morales-Indiano
- Department of Clinical Analysis and Biochemistry, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital, Badalona, Barcelona, Spain.
| | | | | | - Luisa Bordejé-Laguna
- Intensive Care Unit, Germans Trias i Pujol University Hospital, Badalona, Barcelona, Spain.
| | - Víctor Ruíz-Artola
- Intensive Care Unit, Germans Trias i Pujol University Hospital, Badalona, Barcelona, Spain.
| |
Collapse
|
37
|
Marrero Eligio De La Puente CE, Flota Ruiz D, Besalduch LS, Capó XF, Sala DG, Ibars CP, Mindiolaza IB, Chiscano Camon LS, Sanmartin AR, Ruiz-Rodríguez JC, Ferrer R, Montoya SB. Systematic Ultrasound Screening for Lower Extremity Deep Vein Thrombosis in ICU Patients with Severe COVID-19: A Randomized Clinical Trial. J Intensive Care Med 2025:8850666251313774. [PMID: 39838947 DOI: 10.1177/08850666251313774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
BACKGROUND Venous thromboembolism (VTE), whether pulmonary embolism (PE) or deep vein thrombosis (DVT), is common in patients with COVID-19. Recommendations on systematic screening in the intensive care unit (ICU) are lacking. RESEARCH QUESTION Is there any clinical benefit of systematic screening for DVT in critically ill patients with severe COVID-19? STUDY DESIGN AND METHODS Single-center randomized clinical trial (RCT) of COVID-19 cases admitted to the ICU. Patients were randomized into two groups: a study group that underwent ultrasound (US) screening for DVT Mondays and Thursdays, and a control group that was treated according to the unit protocol. The primary outcome was the presence of DVT. Secondary outcomes were ICU total stay, death within 21-day follow-up and bleeding complications (minor or major). A composite outcome of poor prognosis variables was analyzed. We tested a superiority hypothesis with a confidence level of 95% and an equivalence limit of 20%. RESULTS 163 patients (84 screening group, 79 control group) were enrolled between April and July 2021. There were 90 men (55.2%) with a mean ± SD age of 49.8 ± 13.58 years. In screening group 16.7% developed DVT versus 3.8% in control group (p = .007), and 3.6% versus 5.1% developed PE, respectively (p = 0.7). Poor outcome variables were male sex, age, COVID-19 vaccination status, Fibrinogen, Urea, Creatinine and Interleukin 6 (IL6) levels; Acute Physiology and Chronic Health Evaluation II (APACHE II) and Sequential Organ Failure Assessment (SOFA) scales. The superiority comparison, with a power of 95%, showed no statistically significant differences for a composite endpoint (p = .123). After adjusting by group, the OR for poor outcome is 1.966 (0.761-5.081) p = 0.163. INTERPRETATION Among these patients, a strategy of systematic US screening for DVT was not associated with any significant improvements to clinical outcomes compared with usual care. CLINICAL TRIAL REGISTRATION Clinicaltrials.org registration number: NCT05028244.
Collapse
Affiliation(s)
- Carlos Ernesto Marrero Eligio De La Puente
- Servicio de Angiología, cirugía vascular y endovascular. Hospital Universitario Vall d'Hebron, Barcelona, Spain
- Departamento de Cirugía y Ciencias Morfológicas, Universidad Autónoma de Barcelona, Barcelona, Spain
| | - David Flota Ruiz
- Servicio de Angiología, cirugía vascular y endovascular. Hospital Universitario Vall d'Hebron, Barcelona, Spain
| | - Lluis Sánchez Besalduch
- Servicio de Angiología, cirugía vascular y endovascular. Hospital Universitario Vall d'Hebron, Barcelona, Spain
| | - Xavier Faner Capó
- Servicio de Angiología, cirugía vascular y endovascular. Hospital Universitario Vall d'Hebron, Barcelona, Spain
| | - Daniel Gil Sala
- Servicio de Angiología, cirugía vascular y endovascular. Hospital Universitario Vall d'Hebron, Barcelona, Spain
| | - Clara Palmada Ibars
- Servicio de Medicina Intensiva. Hospital Universitario Vall d'Hebron, Barcelona, Spain
| | - Ivan Bajaña Mindiolaza
- Servicio de Medicina Intensiva. Hospital Universitario Vall d'Hebron, Barcelona, Spain
- Grupo de investigación Shock, Disfunción Orgánica y Resucitación. Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | - Luis Silvestre Chiscano Camon
- Servicio de Medicina Intensiva. Hospital Universitario Vall d'Hebron, Barcelona, Spain
- Grupo de investigación Shock, Disfunción Orgánica y Resucitación. Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | - Adolfo Ruiz Sanmartin
- Servicio de Medicina Intensiva. Hospital Universitario Vall d'Hebron, Barcelona, Spain
- Grupo de investigación Shock, Disfunción Orgánica y Resucitación. Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | - Juan Carlos Ruiz-Rodríguez
- Servicio de Medicina Intensiva. Hospital Universitario Vall d'Hebron, Barcelona, Spain
- Grupo de investigación Shock, Disfunción Orgánica y Resucitación. Vall d'Hebron Institut de Recerca, Barcelona, Spain
- Departamento de Medicina, Universidad Autónoma de Barcelona, Barcelona, Spain
| | - Ricard Ferrer
- Servicio de Medicina Intensiva. Hospital Universitario Vall d'Hebron, Barcelona, Spain
- Grupo de investigación Shock, Disfunción Orgánica y Resucitación. Vall d'Hebron Institut de Recerca, Barcelona, Spain
- Departamento de Medicina, Universidad Autónoma de Barcelona, Barcelona, Spain
| | - Sergi Bellmunt Montoya
- Servicio de Angiología, cirugía vascular y endovascular. Hospital Universitario Vall d'Hebron, Barcelona, Spain
- Departamento de Cirugía y Ciencias Morfológicas, Universidad Autónoma de Barcelona, Barcelona, Spain
| |
Collapse
|
38
|
Wu H, Yang J, Yan Y, Hu X, Li B, Cheng P, Song X. The effect of SARS-CoV-2 Omicron BA. 5.2 infection on perioperative physiological indicators in orthopedic patients. BMC Musculoskelet Disord 2025; 26:74. [PMID: 39833742 PMCID: PMC11744956 DOI: 10.1186/s12891-025-08327-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
OBJECTIVE The aim of this study was to investigate the effect of SARS-CoV-2 Omicron BA. 5.2 (hereafter referred to as Omicron BA. 5.2) infection on perioperative physiological indices and to provide clinical considerations for the precautions needed for patients who tested negative for SARS-CoV-2 infection perioperatively. METHODS Patients who underwent time-limited surgery (surgery that must be performed within a certain time) at the Department of Orthopedics of our hospital from September 1, 2022, to March 31, 2023, were divided into an observation group (those with Omicron BA 5.2 and a negative nucleic acid test after December 7, 2022, n = 100) and a control group (those negative for SARS-CoV-2 infection before December 7, 2022, n = 100). Changes in the following factors were compared within and between the two groups: heart rate, mean arterial pressure, oxygenation index at the time of entry into the operating room, incision of the skin and exit from the operating room and the neutrophil‒lymphocyte ratio (NLR), platelet‒lymphocyte ratio (PLR), systemic immune inflammation index (SII), D-dimer level, and fibrinogen degradation product (FDP) level. Changes in interleukin-6 and calcitonin levels were assessed the day after surgery, and multivariate regression analysis of the meaningful results was conducted. RESULTS Heart rate, mean arterial pressure, oxygenation index, NLR, PLR, SII, interleukin-6 and calcitonin were similar between the two groups (P > 0.05). The observation group had higher postoperative D-dimer and FDP levels (P < 0.05). Multivariate regression analysis revealed that Omicron BA 5.2 infection, older age and a history of cerebral infarction were associated with increased D-dimer and FDP levels, and their odds ratios (ORs) and confidence intervals are 3.339 [95% CI, 1.372-8.419], P = 0.008; OR, 1.080 [95% CI, 1.023-1.139]; P = 0.005; OR, 10.644 [95% CI, 1.352-83.320], P = 0.025, respectively. CONCLUSION Omicron BA. 5.2 Infection affects the perioperative coagulation function of orthopedic patients, thereby inducing a hypercoagulable state characterized by significant elevations in D-dimer and FDP levels and increasing the risk of venous thrombosis. This infection had no obvious effect on other physiological indices. Early surgery is feasible, but thrombotic events need to be considered.
Collapse
Affiliation(s)
- Hao Wu
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, China
| | - JunZhe Yang
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, China
| | - YuQin Yan
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, China
| | - Xiao Hu
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, China
| | - BiXi Li
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, China
| | - PengFei Cheng
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, China.
| | - XiaoYang Song
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, China.
| |
Collapse
|
39
|
Wang L, Tian W, Wang S, Liu Y, Wang H, Xiao J, Yu Z, Xie L, Chen Y. Serum proteomics identifies biomarkers for predicting non-survivors in elderly COVID-19 patients. J Proteomics 2025; 311:105356. [PMID: 39547396 DOI: 10.1016/j.jprot.2024.105356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/29/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
In December 2022, China ceased the zero-COVID-19 policy, resulting in an increase in hospitalizations and deaths due to COVID-19, particularly among the elderly population. Predicting non-survivors aims to identify high-risk patients and enable targeted interventions to improve survival rates. Additionally, understanding factors affecting prognosis provides essential insights for further research and optimization of treatment strategies. We applied 4D-DIA mass spectrometry for serum proteome analysis and provided a comprehensive characterization of disease features in elderly patients within the Chinese population. Our study elucidated that immune disorders, lung damage, and cardiovascular disorders are predominant causes of death in these patients. Compared to clinical indices, proteomic analysis is more sensitive in tracing these disorders. We also provided a prediction panel for survival outcomes of elderly patients using levels of CXCL10, CXCL16 and IL1RA, which were validated by ELISA. These biomarkers will help improve predictive efficacy for survival outcomes in elderly patients.
Collapse
Affiliation(s)
- Lin Wang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China; College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing 100091, China
| | - Wenmin Tian
- Center for Precision Medicine Multi-Omics Research, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
| | - Sen Wang
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Yuhong Liu
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing 100091, China
| | - Hongli Wang
- Center for Precision Medicine Multi-Omics Research, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
| | - Junjie Xiao
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing 100091, China
| | - Zhongkuo Yu
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing 100091, China
| | - Lixin Xie
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing 100091, China.
| | - Yang Chen
- Center for Precision Medicine Multi-Omics Research, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China; Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|
40
|
de Sá NBR, Macieira KV, Coelho MRI, Goulart MN, Ribeiro-Alves M, Rosadas LADS, Geraldo KM, Ribeiro MPD, Cardoso SW, Grinsztejn B, Veloso VG, Cazote ADS, de Almeida DV, Giacoia-Gripp CBW, Côrtes FH, Morgado MG. COVID-19 and HIV: Clinical Outcomes and Inflammatory Markers in a Cohort from a Reference Hospital in Rio de Janeiro, Brazil. Viruses 2025; 17:91. [PMID: 39861879 PMCID: PMC11769093 DOI: 10.3390/v17010091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/07/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Severe COVID-19 presents a variety of clinical manifestations associated with inflammatory profiles. People living with HIV (PLWH) could face a higher risk of hospitalization and mortality from COVID-19, depending on their immunosuppression levels. This study describes inflammatory markers in COVID-19 clinical outcomes with and without HIV infection. METHODS We analyzed 112 inpatients of the Hospital Center for COVID-19 (INI/FIOCRUZ), including 22 cases of COVID-19 in PLWH (COVID/PLWH group). Plasma samples were tested for a panel of 15 cytokines by Luminex. Sociodemographic, clinical, and laboratory data were collected from patients' clinical records. RESULTS COVID-19 individuals were stratified according to the WHO clinical severity profiles at hospitalization. Significant differences in clinical scores, symptoms (coughs), and the occurrence of HIV infection were found among the groups. Clinical blood parameters and plasma cytokines were analyzed among COVID-19 groups with distinct severity profiles. Critical COVID-19 cases showed higher levels of inflammatory markers (Bilirubin, D-dimer, PCR, and urea, as well as IL-8, IL-10, TNF-α, INF-α, IL-1β, IL-17A, IL-23, IL-6) than moderate and severe groups. The COVID/PLWH group had lower CD4 counts (64 cells/mm3) and cytokine levels than other COVID-19 patients. CONCLUSIONS Overall, critically ill COVID-19 patients exhibited heightened inflammatory responses, while COVID/PLWH demonstrated unique immunological characteristics without increased mortality risk.
Collapse
Affiliation(s)
- Nathalia Beatriz Ramos de Sá
- Laboratório de AIDS & Imunologia Molecular, Instituto Oswaldo Cruz (IOC), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (K.V.M.); (M.R.I.C.); (M.N.G.); (A.d.S.C.); (D.V.d.A.); (C.B.W.G.-G.); (F.H.C.)
| | - Karine Venegas Macieira
- Laboratório de AIDS & Imunologia Molecular, Instituto Oswaldo Cruz (IOC), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (K.V.M.); (M.R.I.C.); (M.N.G.); (A.d.S.C.); (D.V.d.A.); (C.B.W.G.-G.); (F.H.C.)
| | - Mariana Rosa Inacio Coelho
- Laboratório de AIDS & Imunologia Molecular, Instituto Oswaldo Cruz (IOC), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (K.V.M.); (M.R.I.C.); (M.N.G.); (A.d.S.C.); (D.V.d.A.); (C.B.W.G.-G.); (F.H.C.)
| | - Milena Neira Goulart
- Laboratório de AIDS & Imunologia Molecular, Instituto Oswaldo Cruz (IOC), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (K.V.M.); (M.R.I.C.); (M.N.G.); (A.d.S.C.); (D.V.d.A.); (C.B.W.G.-G.); (F.H.C.)
| | - Marcelo Ribeiro-Alves
- Laboratório de Pesquisa Clínica em IST e AIDS, Instituto Nacional de Infectologia Evandro Chagas (INI), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (M.R.-A.); (L.A.d.S.R.); (K.M.G.); (M.P.D.R.); (S.W.C.); (B.G.); (V.G.V.)
| | - Leonardo Azevedo da Silva Rosadas
- Laboratório de Pesquisa Clínica em IST e AIDS, Instituto Nacional de Infectologia Evandro Chagas (INI), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (M.R.-A.); (L.A.d.S.R.); (K.M.G.); (M.P.D.R.); (S.W.C.); (B.G.); (V.G.V.)
| | - Kim Mattos Geraldo
- Laboratório de Pesquisa Clínica em IST e AIDS, Instituto Nacional de Infectologia Evandro Chagas (INI), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (M.R.-A.); (L.A.d.S.R.); (K.M.G.); (M.P.D.R.); (S.W.C.); (B.G.); (V.G.V.)
| | - Maria Pia Diniz Ribeiro
- Laboratório de Pesquisa Clínica em IST e AIDS, Instituto Nacional de Infectologia Evandro Chagas (INI), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (M.R.-A.); (L.A.d.S.R.); (K.M.G.); (M.P.D.R.); (S.W.C.); (B.G.); (V.G.V.)
| | - Sandra Wagner Cardoso
- Laboratório de Pesquisa Clínica em IST e AIDS, Instituto Nacional de Infectologia Evandro Chagas (INI), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (M.R.-A.); (L.A.d.S.R.); (K.M.G.); (M.P.D.R.); (S.W.C.); (B.G.); (V.G.V.)
| | - Beatriz Grinsztejn
- Laboratório de Pesquisa Clínica em IST e AIDS, Instituto Nacional de Infectologia Evandro Chagas (INI), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (M.R.-A.); (L.A.d.S.R.); (K.M.G.); (M.P.D.R.); (S.W.C.); (B.G.); (V.G.V.)
| | - Valdiléa G. Veloso
- Laboratório de Pesquisa Clínica em IST e AIDS, Instituto Nacional de Infectologia Evandro Chagas (INI), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (M.R.-A.); (L.A.d.S.R.); (K.M.G.); (M.P.D.R.); (S.W.C.); (B.G.); (V.G.V.)
| | - Andressa da Silva Cazote
- Laboratório de AIDS & Imunologia Molecular, Instituto Oswaldo Cruz (IOC), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (K.V.M.); (M.R.I.C.); (M.N.G.); (A.d.S.C.); (D.V.d.A.); (C.B.W.G.-G.); (F.H.C.)
| | - Dalziza Victalina de Almeida
- Laboratório de AIDS & Imunologia Molecular, Instituto Oswaldo Cruz (IOC), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (K.V.M.); (M.R.I.C.); (M.N.G.); (A.d.S.C.); (D.V.d.A.); (C.B.W.G.-G.); (F.H.C.)
| | - Carmem Beatriz Wagner Giacoia-Gripp
- Laboratório de AIDS & Imunologia Molecular, Instituto Oswaldo Cruz (IOC), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (K.V.M.); (M.R.I.C.); (M.N.G.); (A.d.S.C.); (D.V.d.A.); (C.B.W.G.-G.); (F.H.C.)
| | - Fernanda Heloise Côrtes
- Laboratório de AIDS & Imunologia Molecular, Instituto Oswaldo Cruz (IOC), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (K.V.M.); (M.R.I.C.); (M.N.G.); (A.d.S.C.); (D.V.d.A.); (C.B.W.G.-G.); (F.H.C.)
| | - Mariza Gonçalves Morgado
- Laboratório de AIDS & Imunologia Molecular, Instituto Oswaldo Cruz (IOC), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (K.V.M.); (M.R.I.C.); (M.N.G.); (A.d.S.C.); (D.V.d.A.); (C.B.W.G.-G.); (F.H.C.)
| |
Collapse
|
41
|
Gómez-Delgado I, López-Pastor AR, González-Jiménez A, Ramos-Acosta C, Hernández-Garate Y, Martínez-Micaelo N, Amigó N, Espino-Paisán L, Anguita E, Urcelay E. Long-term mitochondrial and metabolic impairment in lymphocytes of subjects who recovered after severe COVID-19. Cell Biol Toxicol 2025; 41:27. [PMID: 39792183 PMCID: PMC11723900 DOI: 10.1007/s10565-024-09976-0] [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: 08/23/2024] [Accepted: 12/21/2024] [Indexed: 01/12/2025]
Abstract
The underlying mechanisms explaining the differential course of SARS-CoV-2 infection and the potential clinical consequences after COVID-19 resolution have not been fully elucidated. As a dysregulated mitochondrial activity could impair the immune response, we explored long-lasting changes in mitochondrial functionality, circulating cytokine levels, and metabolomic profiles of infected individuals after symptoms resolution, to evaluate whether a complete recovery could be achieved. Results of this pilot study evidenced that different parameters of aerobic respiration in lymphocytes of individuals recuperated from a severe course lagged behind those shown upon mild COVID-19 recovery, in basal conditions and after simulated reinfection, and they also showed altered glycolytic capacity. The severe groups showed trends to enhanced superoxide production in parallel to lower OPA1-S levels. Unbalance of pivotal mitochondrial fusion (MFN2, OPA1) and fission (DRP1, FIS1) proteins was detected, suggesting a disruption in mitochondrial dynamics, as well as a lack of structural integrity in the electron transport chain. In serum, altered cytokine levels of IL-1β, IFN-α2, and IL-27 persisted long after clinical recovery, and growing amounts of the latter after severe infection correlated with lower basal and maximal respiration, ATP production, and glycolytic capacity. Finally, a trend for higher circulating levels of 3-hydroxybutyrate was found in individuals recovered after severe compared to mild course. In summary, long after acute infection, mitochondrial and metabolic changes seem to differ in a situation of full recovery after mild infection versus the one evolving from severe infection.
Collapse
Affiliation(s)
- Irene Gómez-Delgado
- Lab. Genetics and Molecular Bases of Complex Diseases, Health Research Institute of Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain
- Cooperative Research Networks Oriented to Health Results (RICORS, REI), ISCIII, 28029, Madrid, Spain
| | - Andrea R López-Pastor
- Lab. Genetics and Molecular Bases of Complex Diseases, Health Research Institute of Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain
- Cooperative Research Networks Oriented to Health Results (RICORS, REI), ISCIII, 28029, Madrid, Spain
| | - Adela González-Jiménez
- Lab. Genetics and Molecular Bases of Complex Diseases, Health Research Institute of Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain
- Cooperative Research Networks Oriented to Health Results (RICORS, REI), ISCIII, 28029, Madrid, Spain
| | - Carlos Ramos-Acosta
- Hematology Group, Health Research Institute of Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain
| | - Yenitzeh Hernández-Garate
- Lab. Genetics and Molecular Bases of Complex Diseases, Health Research Institute of Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain
| | | | - Núria Amigó
- Biosfer Teslab, 43201, Reus, Tarragona, Spain
- Department of Basic Medical Sciences, Rovira I Virgili University, 43007, Tarragona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Laura Espino-Paisán
- Lab. Genetics and Molecular Bases of Complex Diseases, Health Research Institute of Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain
- Cooperative Research Networks Oriented to Health Results (RICORS, REI), ISCIII, 28029, Madrid, Spain
| | - Eduardo Anguita
- Hematology Group, Health Research Institute of Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain
- Department of Medicine, Medical School, Universidad Complutense de Madrid, 28040, Madrid, Spain
- Hematology Department, IML, Hospital Clínico San Carlos, 28040, Madrid, Spain
| | - Elena Urcelay
- Lab. Genetics and Molecular Bases of Complex Diseases, Health Research Institute of Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain.
- Cooperative Research Networks Oriented to Health Results (RICORS, REI), ISCIII, 28029, Madrid, Spain.
| |
Collapse
|
42
|
Cheng HI, Chang KW, Wu BC, Teo MY, Hung WS, Wu HM, Huang ACC, Lin CW, Lin TY, Lin HC, Chiu CH, Lin SM. Comparison of Clinical Characteristics and Mortality Outcome in Critical COVID-19 Patients Infected with Alpha and Omicron Variants. Infect Drug Resist 2025; 18:151-160. [PMID: 39803308 PMCID: PMC11725234 DOI: 10.2147/idr.s479896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025] Open
Abstract
Objective Early reports have indicated that the Omicron variant of coronavirus disease 2019 (COVID-19) may be associated with low mortality. However, the mortality rate of critical patients in Taiwan with COVID-19 caused by different variants has not been well described. Methods This retrospective cohort study was conducted at the Linkou Branch of Chang Gung Memorial Hospital, Taiwan, from April 2020 to September 2022. Critically ill patients who had confirmed SARS-CoV-2 infection and were on mechanical ventilation (MV) were enrolled. Demographic data, laboratory results, and treatment information were collected and analyzed. In addition, clinical outcomes for different SARS-CoV-2 variants were analyzed. Results This study included 110 critical patients with COVID-19 who required intubation and intensive care unit (ICU) admission. Among these patients, 46 (41.8%) required intensive care during Alpha predominance period and 64 (58.2%) during the Omicron predominance period. The Alpha group had a higher body mass index, had a longer ICU stay, and included more patients with acute respiratory distress syndrome, and the Omicron group included more active smokers, had more comorbidities, had worse initial laboratory data (including higher white blood cell counts, prothrombin time [PT], activated partial prothrombin time, blood urine nitrogen levels, and creatine levels), and had higher in-hospital mortality rates (40.6% vs 15.2%, p = 0.004). The independent risk factors for in-hospital mortality, were Charlson Comorbidity Index (CCI) ≥ 3 and higher PT and creatine levels. Conclusion Our study discovered that CCI ≥ 3, elevated serum creatine levels, and prolonged PT were independently associated with a high mortality rate in patients with critical COVID-19. Patients with those risk factors may require intensive monitoring during their treatment course.
Collapse
Affiliation(s)
- Hsin-I Cheng
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkuo, Taiwan
| | - Ko-Wei Chang
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkuo, Taiwan
| | - Bing-Chen Wu
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkuo, Taiwan
| | - Mei-Yuan Teo
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkuo, Taiwan
| | - Wei-Syun Hung
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkuo, Taiwan
| | - Hao-Ming Wu
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkuo, Taiwan
| | | | - Chang-Wei Lin
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Ting-Yu Lin
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkuo, Taiwan
| | - Horng-Chyuan Lin
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkuo, Taiwan
| | - Cheng-Hsun Chiu
- Department of Pediatrics, Chang Gung Children’s Hospital, Chang Gung University College of Medicine, Taoyuan City, Taiwan
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shu-Min Lin
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkuo, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Linkuo, Taiwan
- School of Medicine, National Tsing Hua University, Hsin-Chu, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
43
|
Behar-Lagares R, Virseda-Berdices A, Martínez-González Ó, Blancas R, Homez-Guzmán M, Manteiga E, Churruca-Sarasqueta J, Manso-Álvarez M, Algaba Á, Resino S, Fernández-Rodríguez A, Jiménez-Sousa MA. Dynamics of coagulation proteins upon ICU admission and after one year of recovery from COVID-19: a preliminary study. Front Cell Infect Microbiol 2025; 14:1489936. [PMID: 39844842 PMCID: PMC11751041 DOI: 10.3389/fcimb.2024.1489936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 12/13/2024] [Indexed: 01/24/2025] Open
Abstract
Objectives This study aimed to investigate the association of baseline coagulation proteins with hospitalization variables in COVID-19 patients admitted to ICU, as well as coagulation system changes after one-year post-discharge, taking into account gender-specific bias in the coagulation profile. Methods We conducted a prospective longitudinal study on 49 ICU-admitted COVID-19 patients. Proteins were measured using a Luminex 200™. The association between coagulation protein levels and hospitalization variables was carried out by generalized linear models adjusted by the most relevant covariates. Results At ICU admission, lower factor XII, antithrombin, and protein C levels were linked to the need for invasive mechanical ventilation (IMV) or its duration (p=0.028; p=0.047 and p=0.015, respectively). Likewise, lower factor XII, antithrombin, and prothrombin levels were associated with longer ICU length of stay (ICU LOS) (p=0.045; p=0.022; p=0.036, respectively). From baseline to the end of the follow-up, factor XII, antithrombin, prothrombin, and protein C levels notably increased in patients with longer ICU LOS. One-year post-discharge, differences were found for factor IX, aPTT, and INR. Gender-stratified analysis showed sustained alterations in males. Conclusions Depleted specific coagulation factors on ICU admission are associated with increased severity in critically ill COVID-19 patients. Most coagulation alterations recover one-year post-discharge, except for factor IX, aPTT and INR, which remain reduced.
Collapse
Affiliation(s)
- Raquel Behar-Lagares
- Unit of Viral Infection and Immunity, National Center for Microbiology (CNM), Health Institute Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Ana Virseda-Berdices
- Unit of Viral Infection and Immunity, National Center for Microbiology (CNM), Health Institute Carlos III (ISCIII), Majadahonda, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Óscar Martínez-González
- Critical Care Department, Hospital Universitario del Tajo, Aranjuez, Madrid, Spain
- Universidad Alfonso X el Sabio, Villanueva de la Cañada, Madrid, Spain
- Fundación para la Investigación e Innovación Biomédica del Hospital Universitario Infanta Sofía y Hospital Universitario del Henares (FIB HUIS HHEN), San Sebastián de los Reyes, Madrid, Spain
| | - Rafael Blancas
- Critical Care Department, Hospital Universitario del Tajo, Aranjuez, Madrid, Spain
- Universidad Alfonso X el Sabio, Villanueva de la Cañada, Madrid, Spain
- Fundación para la Investigación e Innovación Biomédica del Hospital Universitario Infanta Sofía y Hospital Universitario del Henares (FIB HUIS HHEN), San Sebastián de los Reyes, Madrid, Spain
| | - Marcela Homez-Guzmán
- Critical Care Department, Hospital Universitario Infanta Cristina, Parla, Madrid, Spain
| | - Eva Manteiga
- Critical Care Department, Hospital Universitario Infanta Cristina, Parla, Madrid, Spain
| | | | - Madian Manso-Álvarez
- Critical Care Department, Hospital Universitario del Tajo, Aranjuez, Madrid, Spain
- Universidad Alfonso X el Sabio, Villanueva de la Cañada, Madrid, Spain
- Fundación para la Investigación e Innovación Biomédica del Hospital Universitario Infanta Sofía y Hospital Universitario del Henares (FIB HUIS HHEN), San Sebastián de los Reyes, Madrid, Spain
| | - Ángela Algaba
- Critical Care Department, Hospital Universitario del Tajo, Aranjuez, Madrid, Spain
- Universidad Alfonso X el Sabio, Villanueva de la Cañada, Madrid, Spain
- Fundación para la Investigación e Innovación Biomédica del Hospital Universitario Infanta Sofía y Hospital Universitario del Henares (FIB HUIS HHEN), San Sebastián de los Reyes, Madrid, Spain
| | - Salvador Resino
- Unit of Viral Infection and Immunity, National Center for Microbiology (CNM), Health Institute Carlos III (ISCIII), Majadahonda, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Amanda Fernández-Rodríguez
- Unit of Viral Infection and Immunity, National Center for Microbiology (CNM), Health Institute Carlos III (ISCIII), Majadahonda, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - María A. Jiménez-Sousa
- Unit of Viral Infection and Immunity, National Center for Microbiology (CNM), Health Institute Carlos III (ISCIII), Majadahonda, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
44
|
Ozturk M, Kumova Guler D, Oskan EE, Onder F. Long-Term Effects of COVID-19 on Optic Disc and Retinal Microvasculature Assessed by Optical Coherence Tomography Angiography. Diagnostics (Basel) 2025; 15:114. [PMID: 39795642 PMCID: PMC11720702 DOI: 10.3390/diagnostics15010114] [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: 11/07/2024] [Revised: 12/26/2024] [Accepted: 01/04/2025] [Indexed: 01/13/2025] Open
Abstract
Objectives: To evaluate the long-term effects of coronavirus disease (COVID-19) on optic disc and macular microvasculature. Methods: 40 post-COVID-19 and 40 healthy subjects were included. Optical coherence tomography angiography (OCTA) was performed for all subjects at the first visit and repeated in the fourth and twelfth months. Radial peripapillary capillary (RPC) vessel density (VD), retinal nerve fiber layer (RNFL) thickness, foveal avascular zone (FAZ) area, FAZ perimeter, VDs of the fovea, parafovea, and perifovea at superficial capillary plexus (SCP) and deep capillary plexus (DCP), and central macular thickness (CMT) were evaluated. The OCTA measurements of the COVID-19 group were compared with the control group. Results: The COVID-19 group showed lower VD values than the control group in the nasal parafoveal quadrant of the SCP at all visits (p = 0.009, p = 0.47, p = 0.042) and in the superior perifoveal quadrant of the DCP in the twelfth-month visit (p = 0.014). At all visits, FAZ area and FAZ perimeter were higher (p = 0.02, p = 0.02, p = 0.002; p = 0.002, p = 0.003, p = 0.005), foveal VD values of both SCP and DCP were lower (p < 0.001, p < 0.001, p < 0.001; p = 0.005, p = 0.001, p = 0.001), and CMT was lower (p < 0.001, p = 0.001, p = 0.001) in the COVID-19 group. The COVID-19 group had higher temporal quadrant RPC at all visits (p = 0.003, p = 0.003, p < 0.001) and higher average, superior and inferior RNFL at first and fourth-month visits (p = 0.014, p = 0.020; p = 0.001, p = 0.003; p = 0.021, p = 0.024). Conclusions: There are long-term changes that mainly point to the ischemia in the COVID-19 patients. We emphasize the need for long-term ophthalmologic and systemic follow-up of COVID-19 patients regarding potential complications.
Collapse
Affiliation(s)
- Mine Ozturk
- Department of Ophthalmology, Haseki Training and Research Hospital, 34096 Istanbul, Turkey; (D.K.G.); (F.O.)
| | - Deniz Kumova Guler
- Department of Ophthalmology, Haseki Training and Research Hospital, 34096 Istanbul, Turkey; (D.K.G.); (F.O.)
| | - Ekin Ece Oskan
- Department of Ophthalmology, Kirikhan State Hospital, 31440 Hatay, Turkey;
| | - Feyza Onder
- Department of Ophthalmology, Haseki Training and Research Hospital, 34096 Istanbul, Turkey; (D.K.G.); (F.O.)
| |
Collapse
|
45
|
Zhang S, Xie B, Lin Y, Chen C, Yang S, Xu S, Chen J, Li X, Yang F, Huang M. Impacts of the COVID-19 pandemic on pediatric and adult patients with hemophilia. Ann Hematol 2025; 104:95-104. [PMID: 39271522 PMCID: PMC11868306 DOI: 10.1007/s00277-024-05992-6] [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: 05/25/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19)-associated mortality rate of hemophilia patients is similar to that of the general population, but the risk of hospitalization and bleeding is higher. However, the specific impact of this infection on hemophilia patients remains unknown. We aimed to investigate the impact of the pandemic on the infection susceptibility, symptoms, drug use, and social intercourse of patients with hemophilia. METHODS A survey was distributed to 265 patients with hemophilia (185 adults and 80 children) in the Fujian hemophilia therapeutic center (Fuzhou City, China) during the COVID-19 pandemic, and data were collected between January 2022 and January 2023. The impacts of SARS-CoV-2 infection on hemophilia symptoms, drug use, and social intercourse of these patients were investigated, and the association between the recovery time and disease conditions was explored in infected patients. RESULTS During the pandemic, compared with adult patients, pediatric patients had significantly reduced social intercourse and outdoor activities because of the fear of contracting COVID-19 (85.0% vs. 66.5%). Bleeding events were also significantly fewer in children than in adults (61.2% vs. 81.1%). The SARS-CoV-2 infection rate was significantly higher in patients living in urban areas than in those living in rural areas (74.3% vs. 53.6%). The duration of symptomatic recovery from COVID-19 was not significantly associated with hemorrhage, type and classification of hemophilia, presence of inhibitors, complications, and vaccination status. CONCLUSION Having COVID-19 infection did not significantly influence the symptoms and treatments in patients with hemophilia. Pediatric patients had significantly fewer bleeding events than adults.
Collapse
Affiliation(s)
- Shuxia Zhang
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China
| | - Bangxiang Xie
- Medical oncology, Jianou Integrated traditional Chinese and western medicine hospital, Nanping, 353100, Fujian, China
| | - Yanfang Lin
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China
| | - Chunrong Chen
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China
| | - Shu Yang
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China
| | - Shujuan Xu
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China
| | - Jing Chen
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China
| | - Xin Li
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China
| | - Feng'e Yang
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China.
| | - Meijuan Huang
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China.
| |
Collapse
|
46
|
Okada K, Kin C, Yamashita Y, Kawamura S, Sato K, Chiba K, Miyake H. Possible mechanisms of spermatogenic dysfunction induced by viral infections: Insights from COVID-19. Reprod Med Biol 2025; 24:e12625. [PMID: 39845480 PMCID: PMC11751869 DOI: 10.1002/rmb2.12625] [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: 08/01/2024] [Accepted: 12/17/2024] [Indexed: 01/24/2025] Open
Abstract
Background As the COVID-19 pandemic nears resolution in 2024, the mechanisms by which SARS-CoV-2 and other viral infections induce spermatogenic dysfunction remain poorly understood. This review examines the mechanisms by which viral infections, particularly COVID-19, disrupt spermatogenesis and highlights the implications for male reproductive health. While reports suggest that spermatogenic dysfunction caused by COVID-19 is mild and transient, these findings may have broader applications in understanding and treating spermatogenic dysfunction caused by future viral infections. Methods The PubMed database was searched to identify original and review articles investigating the mechanisms by which viral infections, particularly SARS-CoV-2, contribute to spermatogenic dysfunction. Main Findings SARS-CoV-2 affects the testis through multiple mechanisms, including ACE2 receptor-mediated entry, direct viral damage, inflammatory response, blood-testis barrier disruption, hormonal imbalance, oxidative stress, and impaired spermatogenesis. The combination of these factors can disrupt testicular function and highlights the complexity of the effects of COVID-19 on male reproductive health. Conclusion COVID-19 may disrupt spermatogenesis through direct testicular infection, systemic inflammation, hormonal disruption, and oxidative stress. Ongoing research, vaccination efforts, and clinical vigilance are essential to address these challenges and develop effective treatment and prevention strategies.
Collapse
Affiliation(s)
- Keisuke Okada
- Department of UrologyKobe City Medical Center West HospitalKobeJapan
- Division of Urology, Department of Organs TherapeuticsKobe University Graduate School of MedicineKobeJapan
| | - Chanhyon Kin
- Division of Urology, Department of Organs TherapeuticsKobe University Graduate School of MedicineKobeJapan
| | - Yosuke Yamashita
- Division of Urology, Department of Organs TherapeuticsKobe University Graduate School of MedicineKobeJapan
| | - Shun Kawamura
- Division of Urology, Department of Organs TherapeuticsKobe University Graduate School of MedicineKobeJapan
| | - Katsuya Sato
- Division of Urology, Department of Organs TherapeuticsKobe University Graduate School of MedicineKobeJapan
| | - Koji Chiba
- Division of Urology, Department of Organs TherapeuticsKobe University Graduate School of MedicineKobeJapan
| | - Hideaki Miyake
- Division of Urology, Department of Organs TherapeuticsKobe University Graduate School of MedicineKobeJapan
| |
Collapse
|
47
|
Yu T, Zhang W, Wang Y, Chen R, Luo S. A clinical study on the screening efficacy of six thrombotic markers TAT, PIC, TM, t-PAI-C, D-D, and FDP for hypercoagulable state in pregnant women. Pak J Med Sci 2025; 41:189-193. [PMID: 39867778 PMCID: PMC11755280 DOI: 10.12669/pjms.41.1.9755] [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: 03/04/2024] [Revised: 10/11/2024] [Accepted: 11/16/2024] [Indexed: 01/28/2025] Open
Abstract
Objective To investigate the screening efficacy of six thrombotic markers for hypercoagulable state (HCS) in pregnant women, including thrombin-antithrombin III complex (TAT), plasmin-alpha-2 plasmin inhibitor complex (PIC), thrombomodulin (TM), tissue-type plasminogen activator inhibitor complex(t-PAI-C), D-dimer(D-D), and fibrinogen degradation products (FDP). Methods This was a retrospective study. Eighty-five high-risk pregnant women who underwent antenatal examination at Baoding maternal and Child Health Hospital from December 2022 to September 2023 were included as the observation group, while 85 healthy pregnant women without complications or comorbidities who underwent routine antenatal examinations at our hospital were randomly enrolled as the control group. Analyzed the screening value of these six thrombotic markers in pregnant women at risk of HCS. Results The levels of TAT, PIC, TM, FDP, and D-D in the observation group were significantly higher than in the control group (P < 0.05, respectively). Pearson correlation analysis showed a weak correlation between maternal age and TAT levels (r = -0.179, P< 0.05) as well as between BMI and FDP and D-D (r = 0.214, 0.232, P< 0.05, respectively). Binary logistic regression analysis revealed that abnormal D-D levels were a risk factor for HCS in pregnant women (OR = 0.117, 95% CI: 0.621-0.639, P< 0.05). The area under the joint curve was 0.757, indicating that the model is of certain predictive value for HCS in pregnant women. Conclusion TAT, PIC, TM, t-PAI-C, D-D, and FDP can used for the screening of HCS in pregnant women and have a certain predictive value for high-risk pregnant women.
Collapse
Affiliation(s)
- Tao Yu
- Tao Yu Clinical Laboratory, Baoding Maternal and Child Health Hospital, Baoding 071000, Hebei, China
| | - Wenjuan Zhang
- Wenjuan Zhang Clinical Laboratory, Baoding Maternal and Child Health Hospital, Baoding 071000, Hebei, China
| | - Yunshuang Wang
- Yunshuang Wang Clinical Laboratory, Baoding Maternal and Child Health Hospital, Baoding 071000, Hebei, China
| | - Ran Chen
- Ran Chen Clinical Laboratory, Baoding Maternal and Child Health Hospital, Baoding 071000, Hebei, China
| | - Shuo Luo
- Shuo Luo High-risk Obstetrics, Baoding Maternal and Child Health Hospital, Baoding 071000, Hebei, China
| |
Collapse
|
48
|
Eslamifar Z, Behzadifard M, Zare E. Investigation of homocysteine, D-dimer and platelet count levels as potential predictors of thrombosis risk in COVID-19 patients. Mol Cell Biochem 2025; 480:439-444. [PMID: 38502382 DOI: 10.1007/s11010-024-04967-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 02/13/2024] [Indexed: 03/21/2024]
Abstract
Thrombosis plays an important role in induction of Coronavirus disease 19 (COVID-19) complications including heart attack and stroke. Reliable biomarkers are needed to predict thrombosis risk for better management and improve patient outcomes. This study aimed to investigate the relationship between homocysteine, a thrombosis-related biomarker, and other thrombosis-related parameters, such as D-dimer and platelet count with disease outcome in COVID-19 patients. This case-control study including 50 intensive care unit hospitalized patients with Covid-19 with a positive RT-PCR test for SARS-CoV-2 infection and 50 healthy individuals as a control group was conducted. Both groups were matched for age and body mass index (BMI) and had no history of underlying diseases such as cardiovascular, liver, kidney or smoking. Blood samples were collected from both groups to measure serum homocysteine, platelet count and D-dimer levels. Data were analyzed using GraphPad Prism version 8.3 software. The study found no statistically significant difference in homocysteine levels between COVID-19 patients and the control group. However, D-dimer levels were significantly higher in the patient group. Platelet count analysis revealed a significant difference between patients who died and those who were discharged from the hospital (P < 0.05). Despite previous studies suggesting a link between homocysteine and thrombosis, this study found no significant difference in homocysteine levels between COVID-19 patients and the control group. The significantly elevated D-dimer levels in the death group patient suggest that D-dimer and thrombocytopenia may be more reliable predictors of thrombosis and worse outcome in COVID-19 patients without underlying diseases.
Collapse
Affiliation(s)
- Zahra Eslamifar
- Department of Medical Laboratory Sciences, School of Paramedical Sciences, Dezful University of Medical Sciences, Dezful, Iran
| | - Mahin Behzadifard
- Department of Medical Laboratory Sciences, School of Paramedical Sciences, Dezful University of Medical Sciences, Dezful, Iran.
| | - Ehsan Zare
- Student Research Committee, Dezful University of Medical Sciences, Dezful, Iran
| |
Collapse
|
49
|
Güven O, Karakurt G, Naser A, Selçuk H, Keleş DV, Gedik E, Avsever M, Köse FF. The Impact of COVID-19 Infection on the Development of Stroke, Pulmonary Embolism, and Myocardial Infarction: A Retrospective Study. Cureus 2025; 17:e77665. [PMID: 39968441 PMCID: PMC11835032 DOI: 10.7759/cureus.77665] [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: 01/18/2025] [Indexed: 02/20/2025] Open
Abstract
INTRODUCTION This study compares the period during which thromboembolic disease develops after contact with the virus before, during, and after the pandemic. METHODS In this study, the medical records of patients with a preliminary diagnosis of myocardial infarction (MI), pulmonary embolism (PE), and ischemic stroke who presented to the Emergency Department before, during, and after the pandemic (when vaccination rates increased) were retrospectively examined. Data on whether these patients had COVID-19 or were vaccinated, the time interval between infection/vaccination and the onset of these conditions, and the prognosis were analyzed. RESULTS In the MI group, patients developed embolism the longest after infection and the shortest after vaccination. Among MI patients, the rate of those who received the BioNTech vaccine during the normalization period was higher than that of those who received Sinovac (p = 0.005). In stroke patients, during the pandemic, the time to post-vaccine embolism was shorter (p < 0.001). Additionally, infection and vaccination increased the mortality rate in stroke and PE patients (p < 0.001). CONCLUSION This study demonstrates that thromboembolic events can occur at varying rates and durations after exposure to the virus. While the causes of thrombosis are multifactorial, contact with the virus may act as a triggering factor, even if COVID-19 does not have a direct effect.
Collapse
Affiliation(s)
- Oya Güven
- Department of Emergency Medicine, Kırklareli University Faculty of Medicine, Kırklareli, TUR
- Department of Emergency Medicine, Kırklareli Training and Research Hospital, Kırklareli, TUR
| | - Gökhan Karakurt
- Department of Pulmonology, Kırklareli Training and Research Hospital, Kırklareli, TUR
| | - Abdulrahman Naser
- Department of Cardiology, Kırklareli Training and Research Hospital, Kırklareli, TUR
| | - Hakan Selçuk
- Department of Emergency Medicine, Babaeskı State Hospital, Kırklareli, TUR
| | - Dilek V Keleş
- Department of Nursing, Kırklareli University Faculty of Health Sciences, Kırklareli, TUR
| | - Emre Gedik
- Department of Neurology, Kırklareli Training and Research Hospital, Kırklareli, TUR
| | - Mert Avsever
- Department of Emergency Medicine, St. John's Hospital, Edinburgh, GBR
| | - Fatih Furkan Köse
- Department of Emergency Medicine, Kırklareli Training and Research Hospital, Kırklareli, TUR
| |
Collapse
|
50
|
Martínez-Martinez AB, Tristancho-Baró A, Garcia-Rodriguez B, Clavel-Millan M, Palacian MP, Milagro A, Rezusta A, Arbones-Mainar JM. Impact of Obesity-Associated SARS-CoV-2 Mutations on COVID-19 Severity and Clinical Outcomes. Viruses 2024; 17:38. [PMID: 39861827 PMCID: PMC11769164 DOI: 10.3390/v17010038] [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: 11/15/2024] [Revised: 12/24/2024] [Accepted: 12/27/2024] [Indexed: 01/27/2025] Open
Abstract
This study explores the relationship between specific SARS-CoV-2 mutations and obesity, focusing on how these mutations may influence COVID-19 severity and outcomes in high-BMI individuals. We analyzed 205 viral mutations from a cohort of 675 patients, examining the association of mutations with BMI, hospitalization, and mortality rates. Logistic regression models and statistical analyses were applied to assess the impact of significant mutations on clinical outcomes, including inflammatory markers and antibody levels. Our findings revealed three key mutations-C14599T, A20268G, and C313T-that were associated with elevated BMI. Notably, C14599T appeared to be protective against hospitalization, suggesting context-dependent effects, while A20268G was linked to a 50% increase in hospitalization risk and elevated antibody levels, potentially indicating an adaptive immune response. C313T showed a 428% increase in mortality risk, marking it as a possible poor-prognosis marker. Interestingly, all three mutations were synonymous, suggesting adaptive roles in obesity-driven environments despite not altering viral protein structures. These results emphasize the importance of studying mutations within the broader context of comorbidities, other mutations, and regional factors to enhance our understanding of SARS-CoV-2 adaptation in high-risk groups. Further validation in larger cohorts is necessary to confirm these associations and to assess their clinical significance.
Collapse
Affiliation(s)
- Ana B. Martínez-Martinez
- Facultad de Ciencias de la Salud, Universidad de Zaragoza, 50009 Zaragoza, Spain;
- Instituto de Investigación Sanitaria Aragón, 50009 Zaragoza, Spain; (B.G.-R.); (M.C.-M.); (A.R.)
| | - Alexander Tristancho-Baró
- Department of Clinical Microbiology, Miguel Servet University Hospital, 50009 Zaragoza, Spain; (A.T.-B.); (M.P.P.); (A.M.)
| | - Beatriz Garcia-Rodriguez
- Instituto de Investigación Sanitaria Aragón, 50009 Zaragoza, Spain; (B.G.-R.); (M.C.-M.); (A.R.)
- Department of Clinical Biochemistry, Miguel Servet University Hospital, 50009 Zaragoza, Spain
| | - Marina Clavel-Millan
- Instituto de Investigación Sanitaria Aragón, 50009 Zaragoza, Spain; (B.G.-R.); (M.C.-M.); (A.R.)
- Adipocyte and Fat Biology Laboratory (AdipoFat), Instituto Aragonés de Ciencias de la Salud (IACS), 50009 Zaragoza, Spain
| | - Maria Pilar Palacian
- Department of Clinical Microbiology, Miguel Servet University Hospital, 50009 Zaragoza, Spain; (A.T.-B.); (M.P.P.); (A.M.)
| | - Ana Milagro
- Department of Clinical Microbiology, Miguel Servet University Hospital, 50009 Zaragoza, Spain; (A.T.-B.); (M.P.P.); (A.M.)
| | - Antonio Rezusta
- Instituto de Investigación Sanitaria Aragón, 50009 Zaragoza, Spain; (B.G.-R.); (M.C.-M.); (A.R.)
- Department of Clinical Microbiology, Miguel Servet University Hospital, 50009 Zaragoza, Spain; (A.T.-B.); (M.P.P.); (A.M.)
| | - Jose M. Arbones-Mainar
- Instituto de Investigación Sanitaria Aragón, 50009 Zaragoza, Spain; (B.G.-R.); (M.C.-M.); (A.R.)
- Adipocyte and Fat Biology Laboratory (AdipoFat), Instituto Aragonés de Ciencias de la Salud (IACS), 50009 Zaragoza, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|