1
|
Wang C, Yu T, Xia Y, Tao F, Sun J, Zhao J, Mao X, Tang M, Yin L, Yang Y, Tan W, Shen L, Zhang S. Serum metabolomic characteristics of COVID-19 patients co-infection with echovirus. Virulence 2025; 16:2497907. [PMID: 40310893 PMCID: PMC12051534 DOI: 10.1080/21505594.2025.2497907] [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: 10/22/2024] [Revised: 03/04/2025] [Accepted: 04/21/2025] [Indexed: 05/03/2025] Open
Abstract
Currently, the Omicron variant of the Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to circulate globally. In our multiplex respiratory pathogen detection, we identified numerous instances of co-infection with Echovirus (ECHO) among Coronavirus Disease 2019 (COVID-19) patients, which exacerbated the clinical symptoms of these patients. Such co-infections are likely to impact the subsequent medical treatment. To date, there are no reports on the pathogenic mechanisms related to COVID-19 co-infected with ECHO. Therefore, this study employed the TM Widely-Targeted metabolomics approach to analyze the serum metabolomes of COVID-19 patients with single SARS-CoV-2 infection (COVID-19), COVID-19 patients co-infected with ECHO (COVID-19 + ECHO), and healthy individuals (Control) recruited from routine physical examinations during the same period. Concurrent clinical laboratory tests were performed on the patients to reveal the differences in metabolomic characteristics between the COVID-19 patients and the COVID-19 + ECHO patients, as well as to explore potential metabolic pathways that may exacerbate disease progression. Our findings indicate that both clinical examination indicators and the pathways enriched by differential metabolites confirm that patients with dual infection exhibit higher inflammatory and immune responses compared to those with single COVID-19 infections. This difference is likely reflected through abnormalities in the glycerophospholipid metabolic pathway, with the metabolite Sn-Glycero-3-Phosphocholine playing a crucial role in this process. Finally, we established a diagnostic model based on logistic regression using five differential metabolites, which accurately differentiates between the dual infection population and the single COVID-19 infection population (AUC = 0.828).
Collapse
Affiliation(s)
- Chunhua Wang
- Department of Clinical Laboratory, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, Hubei Province, China
- Department of Central Laboratory, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China
| | - Tingyu Yu
- Department of Central Laboratory, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China
| | - Ying Xia
- Department of Central Laboratory, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China
| | - Feng Tao
- The Key Laboratory for Precision Diagnosis and Treatment of Thrombotic Diseases in Xiangyang City, Zaoyang First People’s Hospital, Zaoyang, Hubei Province, China
| | - Jiali Sun
- Department of Central Laboratory, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China
| | - Jianzhong Zhao
- Department of Clinical Laboratory, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, Hubei Province, China
| | - Xiaogang Mao
- Department of Central Laboratory, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China
| | - Mengjun Tang
- Department of Central Laboratory, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China
| | - Lijuan Yin
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Yang Yang
- Shenzhen Key Laboratory of Pathogen and Immunity, National Clinical Research Center for infectious Disease, State Key Discipline of Infectious Disease, Shenzhen Third People’s Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
| | - Wenjie Tan
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Liang Shen
- Department of Central Laboratory, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China
| | - Shuaijie Zhang
- Department of Central Laboratory, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China
| |
Collapse
|
2
|
Maqsood K, Ahmad S, Saeed A, Roohi N. Plasma protein biomarkers for long COVID-19: Predictors of symptom severity and mortality risk. Clin Chim Acta 2025; 575:120350. [PMID: 40379198 DOI: 10.1016/j.cca.2025.120350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/16/2025] [Accepted: 05/05/2025] [Indexed: 05/19/2025]
Abstract
INTRODUCTION Long COVID-19 is marked by persistent symptoms beyond the acute phase, necessitating a deeper understanding of mortality risk factors for effective management. Plasma proteins hold promise as prognostic markers, offering insights into disease progression and aiding in mortality risk assessment. METHOD A total of 240 patients and 89 healthy controls were enrolled for this study. Two months post-COVID follow-up, patients were categorized as survivors (n = 212) and non-survivors (n = 28). Plasma samples underwent 2-dimensional Gel Electrophoresis (2DE) for protein separation, followed by LC-MS/MS for protein identification, and validation was performed using ELISA. Proteomic data analysis was conducted using Mascot version 2.3.02 and Samespots software 4.5.1. Statistical analyses were carried out using GraphPad Prism and IBM SPSS. RESULTS LC-MS/MS identified fibrinogen (Spot 14; 52.15 kDa/5.32 pI; protein score 2293) and haptoglobin (Spot 08; 45.86 kDa/6.56 pI; protein score 453) as prospective predictive biomarkers. ELISA results confirmed increased plasma levels of fibrinogen and haptoglobin in severe cases (P < 0.001 for both) and non-survivors (P < 0.01 and P < 0.0086, respectively). ROC analysis showed haptoglobin had moderate predictive power for mortality (AUC = 0.68; P = 0.0025), surpassing fibrinogen (AUC = 0.62; P = 0.0374). CONCLUSION Plasma fibrinogen and haptoglobin are promising biomarkers for assessing disease severity and mortality risk in long COVID. These findings highlight their potential for prognostic applications, warranting further research into their mechanistic roles in COVID-19 and broader clinical implications.
Collapse
Affiliation(s)
- Kaleem Maqsood
- Institute of Zoology, University of the Punjab, Lahore, Punjab, Pakistan
| | - Shaaf Ahmad
- King Edward Medical University/Mayo Hospital, Hospital Road, Lahore, Punjab, Pakistan
| | - Azeem Saeed
- Allama Iqbal Medical College, Lahore, Pakistan
| | - Nabila Roohi
- Institute of Zoology, University of the Punjab, Lahore, Punjab, Pakistan.
| |
Collapse
|
3
|
Chen AL, Robbins M, Masters S, Boudiab E, Finn D, Peshel E, Thomas G, Studzinski D, Truscott S, Watterworth C, Novotny N, Ivascu F, Iacco A. Examining the role of thromboelastography in patients with COVID-19. Perfusion 2025:2676591251340967. [PMID: 40366108 DOI: 10.1177/02676591251340967] [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: 05/15/2025]
Abstract
BackgroundCOVID-19 causes a severe respiratory distress syndrome. Systemic inflammation and hypercoagulability are common. These findings are often evaluated with non-specific markers, including CRP, D-dimer, and fibrinogen. We sought to evaluate thromboelastography (TEG) to better understand this complex coagulopathy.MethodsWe conducted a prospective observational study analyzing TEG results in hospitalized patients with COVID-19. TEG was performed on admission and at pre-set intervals. Based on the TEG findings, patients were deemed "hypercoagulable" or "not hypercoagulable." Clinical outcomes were recorded.Results88 patients were evaluated. 78/88 (89%) were hypercoagulable. 10% of the hypercoagulable group (8/78) died compared to none in the non-hypercoagulable group (0/10), with thrombotic events occurring in 9% (8/88), a higher requirement for O2 support in 19% (17/88), and prolonged length of stay exceeding 4 days for 74% (65/88). No statistical significant differences were observed between the groups for any of the four adverse events. Patients with complete fibrinolysis shutdown (Ly30 = 0) had more thrombotic events than those with Ly30 > 0 (30% vs 0%, p = .03).ConclusionPatients with COVID-19 are often hypercoagulable based upon specific TEG parameters. While many TEG parameters are not associated with adverse outcomes, complete fibrinolysis shutdown is associated with an increased risk of thrombotic events. Further studies are warranted to assess the utility of TEG in this population.
Collapse
Affiliation(s)
- Alexander L Chen
- Department of General Surgery, Corewell Health William Beaumont University Hospital, Royal Oak, MI, USA
| | - Matthew Robbins
- Department of General Surgery, Corewell Health William Beaumont University Hospital, Royal Oak, MI, USA
| | - Sean Masters
- Department of General Surgery, Corewell Health William Beaumont University Hospital, Royal Oak, MI, USA
| | - Elizabeth Boudiab
- Department of General Surgery, Corewell Health William Beaumont University Hospital, Royal Oak, MI, USA
| | - Daniel Finn
- Department of General Surgery, Corewell Health William Beaumont University Hospital, Royal Oak, MI, USA
| | - Emanuela Peshel
- Department of General Surgery, Corewell Health William Beaumont University Hospital, Royal Oak, MI, USA
| | - Gregory Thomas
- Department of General Surgery, Corewell Health William Beaumont University Hospital, Royal Oak, MI, USA
| | - Diane Studzinski
- Department of General Surgery, Corewell Health William Beaumont University Hospital, Royal Oak, MI, USA
| | - Steven Truscott
- Special Chemistry and Toxicology Laboratories, Corewell Health William Beaumont University Hospital, Royal Oak, MI, USA
| | - Courtney Watterworth
- Beaumont Research Institute, Corewell Health William Beaumont University Hospital, Royal Oak, MI, USA
| | - Nathan Novotny
- Department of General Surgery, Corewell Health William Beaumont University Hospital, Royal Oak, MI, USA
| | - Felicia Ivascu
- Department of General Surgery, Corewell Health William Beaumont University Hospital, Royal Oak, MI, USA
| | - Anthony Iacco
- Department of General Surgery, Corewell Health William Beaumont University Hospital, Royal Oak, MI, USA
| |
Collapse
|
4
|
Zdravković R, Vicković S, Preveden M, Drobnjak V, Lukić-Šarkanović M, Miljević IB, Tatić M, Tubić T, Videnović N, Mladenović N, Komazec N, Dračina N, Jerković M, Djoković A, Jakovljević A, Kostić A, Mehmedi E, Redžek A. Effect of Continuous Intraoperative Dexmedetomidine on Interleukin-6 and Other Inflammatory Markers After Coronary Artery Bypass Graft Surgery: A Randomized Controlled Trial. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:787. [PMID: 40428745 PMCID: PMC12113577 DOI: 10.3390/medicina61050787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/07/2025] [Accepted: 04/13/2025] [Indexed: 05/29/2025]
Abstract
Background and Objectives: Coronary artery bypass graft (CABG) surgery is the most common cardiac surgery. One of the main causes of postoperative complications and increased mortality after CABG is the inflammatory response. The aim of this study was to investigate whether continuous intraoperative dexmedetomidine can reduce the increase of IL-6 and other inflammatory markers after CABG surgery. Materials and Methods: The study is registered with ClinicalTrials.gov, NCT06378827, accessed on 23 April 2024. This prospective experimental study was conducted from April to December 2024 and included 100 patients undergoing CABG surgery. Patients in the experimental group (50 patients) received a continuous infusion of dexmedetomidine (0.5 μg/kg/h) from anesthesia induction until the end of surgery, while the patients in the control group (50 patients) received the same volume of saline. The primary outcomes were the changes in the values of interleukin-6 (IL-6), C-reactive protein (CRP), white blood cells (WBC), and fibrinogen on the first postoperative day (POD1) compared to the basal, preoperative values. Results: The patients in the control group were on average 65.26 years old, and the patients in the experimental group were 66.28 years old (p = 0.555). From the control group, 40 (80%) patients were male compared to 37 (74%) patients from the experimental group (p = 0.635). Median IL-6 before surgery was 2.0 pg/mL, while on POD 1 it was 76.2 pg/mL (p < 0.001). Median CRP before surgery was 2.5 mg/dL, while the POD1 value was 45.5 mg/dL (p < 0.001). Median WBC values were 6.7 × 109/L before surgery and 13.6 × 109/L on POD1 (p < 0.001). The average value of fibrinogen was 3.19 g/L before surgery, while on POD1 it was 3.37 g/L (p = 0.024). The increase in IL-6 on POD1 (ΔIL-6) was 72.4 pg/mL in the control group and 73.0 pg/mL in the experimental group (p = 0.427). ΔCRP was 41.2 mg/mL (control group) and 38.0 mg/mL (experimental group) (p = 0.725). ΔWBC was 7.45 × 109/L (control group) and 6.81 × 109/L (experimental group) (p = 0.407). Δfibrinogen was 0.16 g/L (control group) and 0.2 g/L (experimental group) (p = 0.771). Conclusions: Intraoperative administration of dexmedetodine during CABG surgery at a dose of 0.5 µg/kg/h without a loading dose does not lead to a decrease in the intensity of the inflammatory response after surgery.
Collapse
Affiliation(s)
- Ranko Zdravković
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
- Institute of Cardiovascular Diseases of Vojvodina, 21208 Sremska Kamenica, Serbia
| | - Sanja Vicković
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
- Clinical Center of Vojvodina, 21000 Novi Sad, Serbia
| | - Mihaela Preveden
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
- Institute of Cardiovascular Diseases of Vojvodina, 21208 Sremska Kamenica, Serbia
| | - Vanja Drobnjak
- Institute of Cardiovascular Diseases of Vojvodina, 21208 Sremska Kamenica, Serbia
| | - Mirka Lukić-Šarkanović
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
- Clinical Center of Vojvodina, 21000 Novi Sad, Serbia
| | - Iva Bosić Miljević
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
- Institute of Cardiovascular Diseases of Vojvodina, 21208 Sremska Kamenica, Serbia
| | - Milanka Tatić
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
- Institute of Oncology of Vojvodina, 21204 Sremska Kamenica, Serbia
| | - Teodora Tubić
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
- Clinical Center of Vojvodina, 21000 Novi Sad, Serbia
| | - Nebojša Videnović
- Faculty of Medicine, University of Pristina, 38220 Kosovska Mitrovica, Serbia
| | - Nikola Mladenović
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
- Institute of Cardiovascular Diseases of Vojvodina, 21208 Sremska Kamenica, Serbia
| | - Nikola Komazec
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
- Institute of Cardiovascular Diseases of Vojvodina, 21208 Sremska Kamenica, Serbia
| | - Nina Dračina
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
- Institute of Cardiovascular Diseases of Vojvodina, 21208 Sremska Kamenica, Serbia
| | - Milica Jerković
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
- Clinical Center of Vojvodina, 21000 Novi Sad, Serbia
| | - Aleksandra Djoković
- Clinic of Internal Medicine, Clinical Hospital Center Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia
| | | | - Arijeta Kostić
- Faculty of Medicine, University of Pristina, 38220 Kosovska Mitrovica, Serbia
| | - Erdin Mehmedi
- Faculty of Medicine, University of Pristina, 38220 Kosovska Mitrovica, Serbia
| | - Aleksandar Redžek
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
- Institute of Cardiovascular Diseases of Vojvodina, 21208 Sremska Kamenica, Serbia
| |
Collapse
|
5
|
Mara G, Nini G, Cotoraci C. Chronic Obstructive Pulmonary Disease and COVID-19: The Impact of Hematological Biomarkers on Disease Severity and Outcomes. J Clin Med 2025; 14:2765. [PMID: 40283596 PMCID: PMC12027599 DOI: 10.3390/jcm14082765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Chronic obstructive pulmonary disease (COPD) patients are at heightened risk of severe COVID-19 due to underlying respiratory impairment, systemic inflammation, and immune dysregulation. This review explores the hematological changes that occur in COPD patients with COVID-19 and their implications for disease progression, prognosis, and clinical management. Methods: We conducted a comprehensive analysis of recent peer-reviewed studies from medical databases including Clarivate Analytics, PubMed, and Google Scholar. Results: Hematological alterations, such as lymphopenia, elevated neutrophil-to-lymphocyte ratio (NLR), increased D-dimer and fibrinogen levels, inflammatory anemia, and erythrocyte dysfunction, are commonly observed in COPD patients with COVID-19. These changes are linked to immune suppression, hyperinflammation, oxidative stress, and thromboembolic complications. Conclusions: Hematological biomarkers are valuable tools for early risk assessments and guiding treatment strategies in this high-risk population. The regular monitoring of D-dimer, fibrinogen, and NLR is advisable. Prophylactic anticoagulation and immunomodulatory therapies, such as corticosteroids and IL-6 and IL-1 inhibitors, may improve clinical outcomes. Further clinical studies are needed to validate personalized approaches and explore antioxidant-based interventions.
Collapse
Affiliation(s)
- Gabriela Mara
- Multidisciplinary Doctoral School, Vasile Goldis Western University of Arad, 310414 Arad, Romania;
- Pneumology Department, Vasile Goldis Western University of Arad, 310414 Arad, Romania;
| | - Gheorghe Nini
- Pneumology Department, Vasile Goldis Western University of Arad, 310414 Arad, Romania;
| | - Coralia Cotoraci
- Clinical Hematology Department, Vasile Goldis Western University of Arad, 310025 Arad, Romania
| |
Collapse
|
6
|
Desheva YA, Shvedova TN, Kopteva OS, Guzenkov DS, Kudar PA, Kotomina TS, Petrachkova DS, Grigorieva EP, Lerner AA, Ponkratov SV. The Clinical and Laboratory Landscape of COVID-19 During the Initial Period of the Pandemic and at the Beginning of the Omicron Era. Viruses 2025; 17:481. [PMID: 40284924 PMCID: PMC12031490 DOI: 10.3390/v17040481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 03/19/2025] [Accepted: 03/23/2025] [Indexed: 04/29/2025] Open
Abstract
INTRODUCTION Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) underwent significant mutations, resulting in the Omicron variant. METHODS In this study, we analyzed blood samples from 98 patients with acute coronavirus disease 19 (COVID-19) hospitalized during the initial SARS-CoV-2 wave and the onset of Omicron in 2021. High-resolution melting (HRM) analysis of PCR products was used to analyze RNA extracted from clinical samples collected in July and November 2021 from patients infected with SARS-CoV-2. RESULTS HRM analysis revealed a characteristic deletion in the N protein RNA of the virus isolated in November 2021, associated with the Omicron variant. Elevated levels of inflammatory markers and interleukin-6 (IL-6) were observed in both waves of COVID-19. Complement levels and IgG and IgM antibodies to SARS-CoV-2 were detected more often during the second wave. An increase in hemagglutinin-inhibiting (HI) antibodies against influenza viruses was observed in paired blood specimens from moderate to severe COVID-19 patients during both outbreaks. CONCLUSIONS Patients admitted during both waves of COVID-19 showed a significant rise in inflammatory markers, suggesting that Omicron triggers inflammatory responses. The rapid formation of IgM and IgG in Omicron may indicate a faster immune response. Seasonal flu may negatively impact the clinical course of coronavirus infections.
Collapse
Affiliation(s)
- Yulia A. Desheva
- Federal State Budgetary Scientific Institution ‘Institute of Experimental Medicine’, 12, Acad. Pavlov Street, 197022 Saint-Petersburg, Russia; (O.S.K.); (D.S.G.); (P.A.K.); (T.S.K.); (D.S.P.); (E.P.G.)
- Medical Institute, St Petersburg University, 21st Line, bldg. 8a, 199034 Saint-Petersburg, Russia
| | - Tamara N. Shvedova
- Vsevolozhsk Clinical Interdistrict Hospital, 20, Koltushskoe Highway, Leningrad Region, 188643 Arkhangelsk, Russia; (T.N.S.); (S.V.P.)
| | - Olga S. Kopteva
- Federal State Budgetary Scientific Institution ‘Institute of Experimental Medicine’, 12, Acad. Pavlov Street, 197022 Saint-Petersburg, Russia; (O.S.K.); (D.S.G.); (P.A.K.); (T.S.K.); (D.S.P.); (E.P.G.)
| | - Danila S. Guzenkov
- Federal State Budgetary Scientific Institution ‘Institute of Experimental Medicine’, 12, Acad. Pavlov Street, 197022 Saint-Petersburg, Russia; (O.S.K.); (D.S.G.); (P.A.K.); (T.S.K.); (D.S.P.); (E.P.G.)
| | - Polina A. Kudar
- Federal State Budgetary Scientific Institution ‘Institute of Experimental Medicine’, 12, Acad. Pavlov Street, 197022 Saint-Petersburg, Russia; (O.S.K.); (D.S.G.); (P.A.K.); (T.S.K.); (D.S.P.); (E.P.G.)
| | - Tatiana S. Kotomina
- Federal State Budgetary Scientific Institution ‘Institute of Experimental Medicine’, 12, Acad. Pavlov Street, 197022 Saint-Petersburg, Russia; (O.S.K.); (D.S.G.); (P.A.K.); (T.S.K.); (D.S.P.); (E.P.G.)
| | - Daria S. Petrachkova
- Federal State Budgetary Scientific Institution ‘Institute of Experimental Medicine’, 12, Acad. Pavlov Street, 197022 Saint-Petersburg, Russia; (O.S.K.); (D.S.G.); (P.A.K.); (T.S.K.); (D.S.P.); (E.P.G.)
| | - Elena P. Grigorieva
- Federal State Budgetary Scientific Institution ‘Institute of Experimental Medicine’, 12, Acad. Pavlov Street, 197022 Saint-Petersburg, Russia; (O.S.K.); (D.S.G.); (P.A.K.); (T.S.K.); (D.S.P.); (E.P.G.)
| | - Anna A. Lerner
- Department of Clinical Laboratory Diagnostics, Biological and General Chemistry, Federal State Budgetary Educational Institution of Higher Education North-West State Medical University, 191015 Saint-Petersburg, Russia;
| | - Stanislav V. Ponkratov
- Vsevolozhsk Clinical Interdistrict Hospital, 20, Koltushskoe Highway, Leningrad Region, 188643 Arkhangelsk, Russia; (T.N.S.); (S.V.P.)
| |
Collapse
|
7
|
Starikova EA, Mammedova JT, Rubinstein AA, Sokolov AV, Kudryavtsev IV. Activation of the Coagulation Cascade as a Universal Danger Sign. Curr Issues Mol Biol 2025; 47:108. [PMID: 39996829 PMCID: PMC11854423 DOI: 10.3390/cimb47020108] [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: 12/28/2024] [Revised: 01/29/2025] [Accepted: 02/02/2025] [Indexed: 02/26/2025] Open
Abstract
Hemostasis is a mechanism that stops bleeding from an injured vessel, involves multiple interlinked steps, culminating in the formation of a "clot" sealing the damaged area. Moreover, it has long been recognized that inflammation also provokes the activation of the coagulation system. However, there has been an increasing amount of evidence revealing the immune function of the hemostasis system. This review collects and analyzes the results of the experimental studies and data from clinical observations confirming the inflammatory function of hemostasis. Here, we summarize the latest knowledge of the pathways in immune system activation under the influence of coagulation factors. The data analyzed allow us to consider the components of hemostasis as receptors recognizing «foreign» or damaged «self» or/and as «self» damage signals that initiate and reinforce inflammation and affect the direction of the adaptive immune response. To sum up, the findings collected in the review allow us to classify the coagulation factors, such as Damage-Associated Molecular Patterns that break down the conventional concepts of the coagulation system.
Collapse
Affiliation(s)
- Eleonora A. Starikova
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia (I.V.K.)
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L’va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
- Department of Microbiology and Virology, Institute of Medical Education Almazov National Medical Research Centre, 2 Akkuratova Street, 197341 Saint Petersburg, Russia
| | - Jennet T. Mammedova
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia (I.V.K.)
- Department of Molecular Biotechnology, Chemical and Biotechnology Faculty, Saint Petersburg State Institute of Technology, Moskovski Ave., 26, 190013 Saint Petersburg, Russia
| | - Artem A. Rubinstein
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia (I.V.K.)
| | - Alexey V. Sokolov
- Laboratory of Systemic Virology, Department of Molecular Biology of Viruses, Smorodintsev Research Institute of Influenza, 15/17, Prof. Popova Str., 197376 Saint Petersburg, Russia;
| | - Igor V. Kudryavtsev
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia (I.V.K.)
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L’va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
| |
Collapse
|
8
|
García AMG, Arias Arias AJ, Muñoz FL, García-Rico E. Allostatic Load as a Short-Term Prognostic and Predictive Marker. Stress Health 2025; 41:e3527. [PMID: 39789760 DOI: 10.1002/smi.3527] [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: 07/10/2024] [Revised: 10/22/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025]
Abstract
It would be highly valuable to possess a tool for evaluating disease progression and identifying patients at risk of experiencing a more severe clinical course and potentially worse outcomes. The concept of allostatic load, which represents the overall strain on the body from repeated stress responses, has been recognized as a precursor to the development of chronic illnesses. It functions as a cumulative measure of the body's capacity to adapt to stress. Numerous studies have demonstrated that elevated allostatic load levels are associated with various negative health outcomes, both physical and mental, and are more predictive of mortality than individual biomarkers. Leveraging the unique circumstances presented by the COVID-19 pandemic, we evaluated different clinical and laboratory parameters in hospitalised COVID-19 patients to assess allostatic load. Our results indicated that allostatic load acts as a strong predictor of prolonged hospitalisation, increased ICU days, and mortality. This highlights its efficacy as a precise gauge of biological dysregulation linked to the response to COVID-19 during disease progression. Allostatic load is easily obtainable and provides an early, cost-effective indication of disease prognosis. Additionally, it has the potential to forecast the necessity for ICU admission. As a result, this parameter, indicative of the comprehensive physiological disruption in response to stress, emerges as a promising prognostic marker for hospitalised patients, extending beyond COVID-19.
Collapse
Affiliation(s)
- Ana M Gómez García
- Internal Medicine Unit, Hospital Universitario HM Madrid, Madrid, Spain
- Facultad HM de Ciencias de la Salud de la Universidad Camilo José Cela, Villafranca del Castillo, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
| | - Angel Jesús Arias Arias
- Facultad HM de Ciencias de la Salud de la Universidad Camilo José Cela, Villafranca del Castillo, Spain
| | - Francisco López Muñoz
- Facultad HM de Ciencias de la Salud de la Universidad Camilo José Cela, Villafranca del Castillo, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
| | - E García-Rico
- Facultad HM de Ciencias de la Salud de la Universidad Camilo José Cela, Villafranca del Castillo, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
- Medical Oncology Unit, Hospital Universitario HM Torrelodones, Madrid, Spain
| |
Collapse
|
9
|
Luka N, South K, Jones R, Unsworth AJ, Coutts G, Mosneag I, Younas M, Bradley A, Wong SY, Collins E, Quigley C, Knight SB, McColl BW, McCulloch L, Grainger JR, Smith CJ, Allan SM. The Role of the VWF/ADAMTS13 Axis in the Thromboinflammatory Response in Ischemic Stroke After SARS-CoV2 Infection. Brain Behav 2025; 15:e70348. [PMID: 39972966 PMCID: PMC11839761 DOI: 10.1002/brb3.70348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 10/22/2024] [Accepted: 02/02/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND SARS-CoV2 infections increase the risk of ischemic stroke (IS), potentially through a thromboinflammatory cascade driven by an imbalance in the ratio of Von Willebrand Factor (VWF) and a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13 (ADAMTS13), leading to the formation of ultra-large VWF (UL-VWF). However, the SARS-CoV2 infection's contribution to any VWF/ADAMTS13 axis imbalance and the subsequent thromboinflammatory response post-stroke remain poorly understood. METHODS We performed a detailed thromboinflammatory profile of the plasma samples from three experimental cohorts matched by age, sex, and stroke severity: non-stroke controls (n = 23), SARS-CoV2 negative IS (n = 22), and SARS-CoV2 positive IS (n = 24). SARS-CoV2 positive IS patients presented varying degrees of infection severity. RESULTS We observed an increase in VWF and UL-VWF and a decrease in ADAMTS13 in the SARS-CoV2 positive IS cohort, suggesting a VWF/ADAMTS13 axis imbalance. Interleukin-6 (IL-6) levels were positively correlated with VWF and negatively correlated with ADAMTS13, suggesting that IL-6 may drive this imbalance. Fibrinogen and D-Dimers were elevated in SARS-CoV2 negative IS cohort and SARS-CoV2 positive IS cohort, but D-Dimers were within the normal range, indicating no disseminated intravascular coagulation. Factor IX (FIX) was elevated in the SARS-CoV2 negative IS cohort. Tissue plasminogen activator (tPA) was elevated in the SARS-CoV2 positive IS cohort, suggesting no fibrinolysis defects. Matrix Metalloproteinase-2 (MMP-2) and soluble Intracellular Adhesion Molecule-1 (sICAM-1) were elevated in the SARS-CoV2 negative IS cohort. CONCLUSIONS We show that SARS-CoV2 infections drive a VWF/ADAMTS13 axis imbalance, inducing an increase in tPA while decreasing FIX, MMP-2, and sICAM-1 post-stroke.
Collapse
Affiliation(s)
- Nadim Luka
- Geoffrey Jefferson Brain Research Centre, School of Biological Sciences, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Division of NeuroscienceThe University of ManchesterManchesterUK
| | - Kieron South
- Geoffrey Jefferson Brain Research Centre, School of Biological Sciences, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Division of NeuroscienceThe University of ManchesterManchesterUK
| | - Rachel Jones
- Geoffrey Jefferson Brain Research Centre, School of Biological Sciences, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Division of Cardiovascular SciencesThe University of ManchesterManchesterUK
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and HealthManchester Academic Health Science Centre, The University of ManchesterManchesterUK
| | - Amanda J. Unsworth
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Graham Coutts
- Geoffrey Jefferson Brain Research Centre, School of Biological Sciences, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Division of NeuroscienceThe University of ManchesterManchesterUK
| | - Ioana‐Emilia Mosneag
- Geoffrey Jefferson Brain Research Centre, School of Biological Sciences, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Division of NeuroscienceThe University of ManchesterManchesterUK
| | - Mehwish Younas
- Geoffrey Jefferson Brain Research Centre, School of Biological Sciences, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Division of NeuroscienceThe University of ManchesterManchesterUK
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and HealthManchester Academic Health Science Centre, The University of ManchesterManchesterUK
| | - Amy Bradley
- Geoffrey Jefferson Brain Research Centre, School of Biological Sciences, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
| | - Siew Yan Wong
- Geoffrey Jefferson Brain Research Centre, School of Biological Sciences, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
| | - Ellen Collins
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
| | - Chloe Quigley
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
| | - Sean B. Knight
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
- Division of Immunology, Immunity to Infection and Respiratory MedicineThe University of ManchesterManchesterUK
| | - Barry W. McColl
- UK Dementia Research Institute, Centre for Discovery Brain SciencesThe University of EdinburghEdinburghUK
| | - Laura McCulloch
- Centre for Inflammation Research, Institute for Regeneration and RepairThe University of EdinburghEdinburghUK
| | - John R. Grainger
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
- Division of Immunology, Immunity to Infection and Respiratory MedicineThe University of ManchesterManchesterUK
| | - Craig J. Smith
- Geoffrey Jefferson Brain Research Centre, School of Biological Sciences, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Division of Cardiovascular SciencesThe University of ManchesterManchesterUK
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and HealthManchester Academic Health Science Centre, The University of ManchesterManchesterUK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
| | - Stuart M. Allan
- Geoffrey Jefferson Brain Research Centre, School of Biological Sciences, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Division of NeuroscienceThe University of ManchesterManchesterUK
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and HealthManchester Academic Health Science Centre, The University of ManchesterManchesterUK
| |
Collapse
|
10
|
Melidoro P, Sultan ARA, Qureshi A, Yacoub MH, Elkhodary KL, Lip GYH, Montarello N, Lahoti N, Rajani R, Klis M, Williams SE, Aslanidi O, De Vecchi A. Enhancing stroke risk stratification in atrial fibrillation through non-Newtonian blood modelling and Gaussian process emulation. J Physiol 2024. [PMID: 39689233 DOI: 10.1113/jp287283] [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: 07/15/2024] [Accepted: 11/23/2024] [Indexed: 12/19/2024] Open
Abstract
Atrial fibrillation (AF) is the most common heart arrhythmia, linked to a five-fold increase in stroke risk. The left atrial appendage (LAA), prone to blood stasis, is a common thrombus formation site in AF patients. The LAA can be classified into four morphologies: broccoli, cactus, chicken wing and windsock. Stroke risk prediction in AF typically relies on demographic characteristics and comorbidities, often overlooking blood flow dynamics. We developed patient-specific non-Newtonian models of blood flow, dependent on fibrinogen and haematocrit, to predict changes in LAA viscosity, aiming to predict stroke in AF patients. We conducted 480 computational fluid dynamics (CFD) simulations using the non-Newtonian model across the four LAA morphologies for four virtual patient cohorts: AF + Covid-19, AF + pathological fibrinogen, AF + normal fibrinogen, and healthy controls. Gaussian process emulators (GPEs) were trained on this in silico cohort to predict average LAA viscosity at near-zero computational cost. GPEs demonstrated high accuracy in AF cohorts but lower accuracy when the chicken wing GPE was applied to other morphologies. Global sensitivity analysis showed fibrinogen significantly influenced blood viscosity in all AF cohorts. The chicken wing morphology exhibited the highest viscosity, while the AF + Covid-19 cohort had the highest viscosity. Our non-Newtonian model in CFD simulations confirmed fibrinogen's substantial impact on blood viscosity at low shear rates in the LAA, suggesting that combining blood values and geometric parameters of the LAA into GPE training could enhance stroke risk stratification accuracy. KEY POINTS: Fibrinogen has a significant effect on blood viscosity in the left atrial appendage (LAA) at low shear rates. Gaussian process emulators (GPEs) can predict the viscosity of blood in the LAA at near-zero computational cost. Out of all LAA morphologies, the chicken wing morphology exhibited the highest average blood viscosity. High average blood viscosity in the LAA of atrial fibrilation + Covid-19 patients was observed due to high fibrinogen levels in this cohort. Combining blood values and geometric parameters of the LAA into GPE training could enhance stroke risk stratification accuracy.
Collapse
Affiliation(s)
- Paolo Melidoro
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Abdel Rahman Amr Sultan
- Department of Mechanical Engineering, The American University in Cairo, New Cairo, Egypt
- Aswan Heart Research Centre, Magdi Yacoub Foundation, Aswan, Egypt
| | - Ahmed Qureshi
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Magdi H Yacoub
- Aswan Heart Research Centre, Magdi Yacoub Foundation, Aswan, Egypt
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Khalil L Elkhodary
- Department of Mechanical Engineering, The American University in Cairo, New Cairo, Egypt
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moore's University and Liverpool Heart & Chest Hospital, Liverpool, UK
- Danish Center for Health Services Research, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Natalie Montarello
- Cardiovascular Directorate, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Nishant Lahoti
- Cardiovascular Directorate, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Ronak Rajani
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Magdalena Klis
- Cardiovascular Directorate, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Steven E Williams
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
- Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| | - Oleg Aslanidi
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Adelaide De Vecchi
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| |
Collapse
|
11
|
Chen L, Jiang YH, Li MY, Huang B, Yuan L, Wan JH, Qin TY, Zeng TT, Chen QG. The Value of Common Laboratory Markers in Predicting the Severity of COVID-19 Patients. Infect Drug Resist 2024; 17:5037-5047. [PMID: 39559342 PMCID: PMC11572049 DOI: 10.2147/idr.s478798] [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: 07/26/2024] [Accepted: 09/26/2024] [Indexed: 11/20/2024] Open
Abstract
Purpose The aim of the present study was to identify more effective laboratory markers to assess the severity of corona virus disease 2019 and predict the progression of the disease by collecting more laboratory markers and variables. Patients and Methods In this study, most risk factors, including epidemiological characteristics, blood cell counts, cytokines, and infection markers, were collected from 126 patients with COVID-19 to assess their predictive value. Results The area under curve (AUC) of Albumin (Alb) to fibrinogen (Fib) ratio (AFR) (0.791), Lactate dehydrogenase (LDH) (0.792), myoglobin (MYO) (0.795), C-reactive protein (CRP) (0.801) and lymphocyte count (0.859) were higher than other markers to distinguish severe from non-severe patients in receiver operating characteristic (ROC) analysis. In the univariate logistic regression analysis, thirty-six out of 46 risk factors, including 34 laboratory markers, were significantly associated with increased odds of severe patients. Multivariate logistic regression analysis showed that the CD19+ lymphocyte count, MYO, LDH, and AFR were associated with increased odds of severe disease. Moreover, Lymphocyte count and AFR levels increased, LDH and CRP levels decreased during hospitalization in recovered severe patients, whereas severe lymphocytopenia and continuously increasing LDH levels were observed in deteriorated patients. AFR level increased and CRP level decreased before the disease worsened in the deteriorated patients; however, when the patients deteriorated, AFR decreased and CRP increased significantly. Conclusion CD19+ lymphocyte count, MYO, LDH, and AFR are independent biomarkers for early identification of severe COVID-19. Lymphocyte count, AFR, LDH, and CRP levels were helpful in predicting the clinical progression of the disease.\.
Collapse
Affiliation(s)
- Lian Chen
- Department of Ultrasound Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Yu-Huan Jiang
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Mei-Yong Li
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Bo Huang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Lei Yuan
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Jin-Hua Wan
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Ting-Yu Qin
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Ting-Ting Zeng
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Qing-Gen Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| |
Collapse
|
12
|
Lian H, Cai H, Zhang H, Zhang Y, Wang X. Inflammation, immunity and biomarkers in procoagulant responses of critically ill patients. Am J Transl Res 2024; 16:5797-5812. [PMID: 39544782 PMCID: PMC11558399 DOI: 10.62347/edar9565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/26/2024] [Indexed: 11/17/2024]
Abstract
Understanding the pathobiology of critical illness is essential for patients' prognosis. Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection. As part of the host response, procoagulant responses, one of the most primitive reactions in biology, start at the very beginning of diseases and can be monitored throughout the process. Currently, we can achieve near-complete monitoring of the coagulation process, and procoagulant responses serve as indicators of the severity of host response in critically ill patients. However, the rapid interpretation of the complex results of various biomarkers remains a challenge for many clinicians. The indicators commonly used for coagulation assessment are complex, typically divided into three categories for clarity: process index, functional index, and outcome index. Monitoring and understanding these indicators can help manage procoagulant responses. The intervention of procoagulant response should be part of the bundle therapy, alongside the treatment for primary disease, management for hemodynamics, and controlling for host response. Early intervention for procoagulant response mainly includes anti-inflammation, antiplatelet and anticoagulant therapy, as well as management of primary disease. In this review, we systemically introduce the onset, assessment and intervention of procoagulant response.
Collapse
Affiliation(s)
- Hui Lian
- Department of Health Care, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100730, China
| | - Huacong Cai
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100730, China
| | - Hongmin Zhang
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100730, China
| | - Yan Zhang
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100730, China
| | - Xiaoting Wang
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100730, China
| |
Collapse
|
13
|
Harutyunyan T, Sargsyan A, Kalashyan L, Stepanyan N, Aroutiounian R, Liehr T, Hovhannisyan G. DNA Damage in Moderate and Severe COVID-19 Cases: Relation to Demographic, Clinical, and Laboratory Parameters. Int J Mol Sci 2024; 25:10293. [PMID: 39408623 PMCID: PMC11476890 DOI: 10.3390/ijms251910293] [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: 08/19/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
The ability of the SARS-CoV-2 virus to cause DNA damage in infected humans requires its study as a potential indicator of COVID-19 progression. DNA damage was studied in leukocytes of 65 COVID-19 patients stratified by sex, age, and disease severity in relation to demographic, clinical, and laboratory parameters. In a combined group of COVID-19 patients, DNA damage was shown to be elevated compared to controls (12.44% vs. 5.09%, p < 0.05). Severe cases showed higher DNA damage than moderate cases (14.66% vs. 10.65%, p < 0.05), and males displayed more damage than females (13.45% vs. 8.15%, p < 0.05). DNA damage is also correlated with international normalized ratio (INR) (r = 0.471, p < 0.001) and creatinine (r = 0.326, p < 0.05). In addition to DNA damage, severe COVID-19 is associated with age, C-reactive protein (CRP), and creatinine. Receiver operating characteristic analysis identified age, INR, creatinine, DNA damage, and CRP as significant predictors of disease severity, with cut-off values of 72.50 years, 1.46 s, 78.0 µmol/L, 9.72%, and 50.0 mg/L, respectively. The results show that DNA damage correlates with commonly accepted COVID-19 risk factors. These findings underscore the potential of DNA damage as a biomarker for COVID-19 severity, suggesting its inclusion in prognostic assessments to facilitate early intervention and improve patient outcomes.
Collapse
Affiliation(s)
- Tigran Harutyunyan
- Laboratory of General and Molecular Genetics, Research Institute of Biology, Yerevan State University, Alex Manoogian 1, Yerevan 0025, Armenia; (T.H.); (A.S.); (L.K.); (R.A.); (G.H.)
- Department of Genetics and Cytology, Yerevan State University, Alex Manoogian 1, Yerevan 0025, Armenia
| | - Anzhela Sargsyan
- Laboratory of General and Molecular Genetics, Research Institute of Biology, Yerevan State University, Alex Manoogian 1, Yerevan 0025, Armenia; (T.H.); (A.S.); (L.K.); (R.A.); (G.H.)
- Department of Genetics and Cytology, Yerevan State University, Alex Manoogian 1, Yerevan 0025, Armenia
| | - Lily Kalashyan
- Laboratory of General and Molecular Genetics, Research Institute of Biology, Yerevan State University, Alex Manoogian 1, Yerevan 0025, Armenia; (T.H.); (A.S.); (L.K.); (R.A.); (G.H.)
| | - Naira Stepanyan
- National Center for Infectious Diseases, Arno Babajanyan 21, Yerevan 0064, Armenia;
| | - Rouben Aroutiounian
- Laboratory of General and Molecular Genetics, Research Institute of Biology, Yerevan State University, Alex Manoogian 1, Yerevan 0025, Armenia; (T.H.); (A.S.); (L.K.); (R.A.); (G.H.)
- Department of Genetics and Cytology, Yerevan State University, Alex Manoogian 1, Yerevan 0025, Armenia
| | - Thomas Liehr
- Jena University Hospital, Institute of Human Genetics, Friedrich Schiller University, Am Klinikum 1, D-07747 Jena, Germany
| | - Galina Hovhannisyan
- Laboratory of General and Molecular Genetics, Research Institute of Biology, Yerevan State University, Alex Manoogian 1, Yerevan 0025, Armenia; (T.H.); (A.S.); (L.K.); (R.A.); (G.H.)
- Department of Genetics and Cytology, Yerevan State University, Alex Manoogian 1, Yerevan 0025, Armenia
| |
Collapse
|
14
|
Antar SA, Ashour NA, Hamouda AO, Noreddin AM, Al-Karmalawy AA. Recent advances in COVID-19-induced liver injury: causes, diagnosis, and management. Inflammopharmacology 2024:10.1007/s10787-024-01535-7. [PMID: 39126569 DOI: 10.1007/s10787-024-01535-7] [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: 10/14/2023] [Accepted: 03/29/2024] [Indexed: 08/12/2024]
Abstract
Since the start of the pandemic, considerable advancements have been made in our understanding of the effects of SARS-CoV-2 infection and the associated COVID-19 on the hepatic system. There is a broad range of clinical symptoms for COVID-19. It affects multiple systems and has a dominant lung illness depending on complications. The progression of COVID-19 in people with pre-existing chronic liver disease (CLD) has also been studied in large multinational groups. Notably, SARS-CoV-2 infection is associated with a higher risk of hepatic decompensation and death in patients with cirrhosis. In this review, the source, composition, mechanisms, transmission characteristics, clinical characteristics, therapy, and prevention of SARS-CoV-2 were clarified and discussed, as well as the evolution and variations of the virus. This review briefly discusses the causes and effects of SARS-CoV-2 infection in patients with CLD. As part of COVID-19, In addition, we assess the potential of liver biochemistry as a diagnostic tool examine the data on direct viral infection of liver cells, and investigate potential pathways driving SARS-CoV-2-related liver damage. Finally, we explore how the pandemic has had a significant impact on patient behaviors and hepatology services, which may increase the prevalence and severity of liver disease in the future. The topics encompassed in this review encompass the intricate relationships between SARS-CoV-2, liver health, and broader health management strategies, providing valuable insights for both current clinical practice and future research directions.
Collapse
Affiliation(s)
- Samar A Antar
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA, 24016, USA
- Department of Pharmacology, Faculty of Pharmacy, Horus University-Egypt, New Damietta, 34518, Egypt
| | - Nada A Ashour
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| | - Amir O Hamouda
- Department of Biochemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta, 34518, Egypt
| | - Ayman M Noreddin
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ahram Canadian University, 6Th of October City, Giza, 12566, Egypt
- Department of Internal Medicine, School of Medicine, University of California -Irvine, Irvine, USA
| | - Ahmed A Al-Karmalawy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta, New Damietta, 34518, Egypt.
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ahram Canadian University, 6Th of October City, Giza, 12566, Egypt.
| |
Collapse
|
15
|
Li X, Du H, Song Z, Meiqi, Zhang G, Yuan S, Yuanfeng, Wang H. Association between fibrinogen levels and stroke-associated pneumonia in acute ischemic stroke patients. BMC Neurol 2024; 24:256. [PMID: 39048948 PMCID: PMC11267856 DOI: 10.1186/s12883-024-03752-7] [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: 01/25/2024] [Accepted: 07/04/2024] [Indexed: 07/27/2024] Open
Abstract
PURPOSE Prior research had indicated a relationship between fibrinogen and stroke-associated pneumonia (SAP), yet the nature of this relationship had not been thoroughly investigated. Therefore, this study was designed to elucidate the prognostic value of fibrinogen levels in forecasting the occurrence of SAP among patients with acute ischemic stroke (AIS). PATIENTS AND METHODS In this retrospective cross-sectional analysis, we included 1092 patients who had experienced AIS and were admitted to our facility within 72 h of the onset of their symptoms. Based on the SAP diagnostic criteria, patients were classified into two groups: SAP and non-SAP. The correlation between serum fibrinogen concentration and SAP was examined using univariate analysis. Curve fitting and multivariable logistic regression model were utilized for statistical evaluation. RESULTS Out of the ischemic stroke patients included in the study, SAP was identified in 112 (10.26%) patients. A direct correlation was observed between fibrinogen levels and the incidence of SAP. An increase in fibrinogen levels corresponded with a heightened incidence of SAP. Multivariable logistic regression revealed a significant positive association between fibrinogen levels and SAP incidence (OR = 1.53, 95% confidence interval [CI]: 1.18, 1.99)). CONCLUSION A linear relationship between serum fibrinogen levels and the incidence of SAP in ischemic stroke patients was shown. The serum fibrinogen levels were positively and linearly correlated to SAP risk.
Collapse
Affiliation(s)
- Xiaoqiang Li
- Department of Neurology, Xiaolan People's Hospital of Zhongshan, No. 65, Jucheng Rd. Xiaolan Dist, Zhongshan, Guangdong Prov, 528415, P.R. China
| | - Hui Du
- Department of Blood Transfusion, Xiaolan People's Hospital of Zhongshan, Zhongshan, Guangdong, China
| | - Zhibin Song
- Department of Neurology, Xiaolan People's Hospital of Zhongshan, No. 65, Jucheng Rd. Xiaolan Dist, Zhongshan, Guangdong Prov, 528415, P.R. China
| | - Meiqi
- Department of Neurology, Xiaolan People's Hospital of Zhongshan, No. 65, Jucheng Rd. Xiaolan Dist, Zhongshan, Guangdong Prov, 528415, P.R. China
| | - Guifeng Zhang
- Department of Neurology, Xiaolan People's Hospital of Zhongshan, No. 65, Jucheng Rd. Xiaolan Dist, Zhongshan, Guangdong Prov, 528415, P.R. China
| | - Suhua Yuan
- Medical Records Room, Xiaolan People's Hospital of Zhongshan, Zhongshan, Guangdong, China
| | - Yuanfeng
- Southern Medical University, Guangzhou, Guangdong, China
| | - Hui Wang
- Department of Neurology, Xiaolan People's Hospital of Zhongshan, No. 65, Jucheng Rd. Xiaolan Dist, Zhongshan, Guangdong Prov, 528415, P.R. China.
| |
Collapse
|
16
|
Giancola ML, Fontana A, Panebianco C, Mazzarelli A, Beccacece A, De Marco P, Cocomazzi G, De Giuli C, Grassi G, Fontana C, Baldini GM, Contu V, Copetti M, Perri F, Nicastri E, Pazienza V. Efficacy of a Multistrain Synbiotic Treatment in Acute and Post-Acute COVID-19 Patients: A Double-Blind, Placebo-Controlled Randomized Trial. Microorganisms 2024; 12:1443. [PMID: 39065211 PMCID: PMC11279369 DOI: 10.3390/microorganisms12071443] [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: 06/17/2024] [Revised: 07/03/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND AND AIMS Several studies reported the effect of COVID-19 on inducing gut dysbiosis, which is also correlated with disease severity. This study aims to investigate the effect of a nutraceutical formula on the shift of microbiota profiles and, secondly, on the clinical-pathological parameters of acute and post-acute COVID-19 patients. METHODS In this randomised, double-blind, placebo-controlled trial conducted at National Institute for Infectious diseases (INMI) Lazzaro Spallanzani (Italy), 52 patients were randomly assigned (1:1) to receive a multistrain synbiotic formula (Kebirah®) or placebo orally for 35 days at COVID-19 diagnosis. Health professionals, investigators, and patients were masked to group assignments. The V3-V4 hypervariable region of 16S rRNA gene sequencing was employed to study the gut microbiota composition in the two groups. RESULTS Supplementation with Kebirah® prevented the decrease in the Shannon diversity index of gut microbiota, which was instead observed in patients receiving the placebo. In addition, decreases in lymphocyte count and haemoglobin levels were observed only in the placebo group and not in the treated group, which was also characterised by an amelioration of the gut microbial profile, with an enrichment in beneficial bacteria and a preservation of species diversity. CONCLUSIONS Our data suggest that modulating the gut microbiota in acute disease through administration of a specific symbiotic formula could be a useful strategy in the frame of SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Maria Letizia Giancola
- National Institute for Infectious Diseases, INMI “Lazzaro Spallanzani”, IRCCS, 00149 Rome, Italy; (M.L.G.); (A.M.); (A.B.); (P.D.M.); (C.D.G.); (G.G.); (C.F.)
| | - Andrea Fontana
- Biostatistic Unit, Fondazione-IRCCS “Casa Sollievo della Sofferenza” Hospital, 71013 San Giovanni Rotondo, FG, Italy; (A.F.); (M.C.)
| | - Concetta Panebianco
- Gastroenterology Unit, Fondazione-IRCCS “Casa Sollievo della Sofferenza” Hospital, Opera di San Pio da Pietrelcina, 71013 San Giovanni Rotondo, FG, Italy; (C.P.); (G.C.)
| | - Antonio Mazzarelli
- National Institute for Infectious Diseases, INMI “Lazzaro Spallanzani”, IRCCS, 00149 Rome, Italy; (M.L.G.); (A.M.); (A.B.); (P.D.M.); (C.D.G.); (G.G.); (C.F.)
| | - Alessia Beccacece
- National Institute for Infectious Diseases, INMI “Lazzaro Spallanzani”, IRCCS, 00149 Rome, Italy; (M.L.G.); (A.M.); (A.B.); (P.D.M.); (C.D.G.); (G.G.); (C.F.)
| | - Patrizia De Marco
- National Institute for Infectious Diseases, INMI “Lazzaro Spallanzani”, IRCCS, 00149 Rome, Italy; (M.L.G.); (A.M.); (A.B.); (P.D.M.); (C.D.G.); (G.G.); (C.F.)
| | - Giovanna Cocomazzi
- Gastroenterology Unit, Fondazione-IRCCS “Casa Sollievo della Sofferenza” Hospital, Opera di San Pio da Pietrelcina, 71013 San Giovanni Rotondo, FG, Italy; (C.P.); (G.C.)
| | - Chiara De Giuli
- National Institute for Infectious Diseases, INMI “Lazzaro Spallanzani”, IRCCS, 00149 Rome, Italy; (M.L.G.); (A.M.); (A.B.); (P.D.M.); (C.D.G.); (G.G.); (C.F.)
| | - Germana Grassi
- National Institute for Infectious Diseases, INMI “Lazzaro Spallanzani”, IRCCS, 00149 Rome, Italy; (M.L.G.); (A.M.); (A.B.); (P.D.M.); (C.D.G.); (G.G.); (C.F.)
| | - Carla Fontana
- National Institute for Infectious Diseases, INMI “Lazzaro Spallanzani”, IRCCS, 00149 Rome, Italy; (M.L.G.); (A.M.); (A.B.); (P.D.M.); (C.D.G.); (G.G.); (C.F.)
| | - Giorgio Maria Baldini
- AO Consorziale Policlinico di Bari, Università Aldo Moro di Bari, 70121 Bari, BA, Italy;
| | - Viviana Contu
- Integrative Medicine Unit, Humanitas Gradenigo, Corso Regina Margherita 8/10, 10153 Torino, TO, Italy;
| | - Massimiliano Copetti
- Biostatistic Unit, Fondazione-IRCCS “Casa Sollievo della Sofferenza” Hospital, 71013 San Giovanni Rotondo, FG, Italy; (A.F.); (M.C.)
| | - Francesco Perri
- Gastroenterology Unit, Fondazione-IRCCS “Casa Sollievo della Sofferenza” Hospital, Opera di San Pio da Pietrelcina, 71013 San Giovanni Rotondo, FG, Italy; (C.P.); (G.C.)
| | - Emanuele Nicastri
- National Institute for Infectious Diseases, INMI “Lazzaro Spallanzani”, IRCCS, 00149 Rome, Italy; (M.L.G.); (A.M.); (A.B.); (P.D.M.); (C.D.G.); (G.G.); (C.F.)
| | - Valerio Pazienza
- Gastroenterology Unit, Fondazione-IRCCS “Casa Sollievo della Sofferenza” Hospital, Opera di San Pio da Pietrelcina, 71013 San Giovanni Rotondo, FG, Italy; (C.P.); (G.C.)
| |
Collapse
|
17
|
Paes Leme AF, Yokoo S, Normando AGC, Ormonde JVS, Domingues RR, Cruz FF, Silva PL, Souza BSF, Dos Santos CC, Castro-Faria-Neto H, Martins CM, Lopes-Pacheco M, Rocco PRM. Proteomics of serum-derived extracellular vesicles are associated with the severity and different clinical profiles of patients with COVID-19: An exploratory secondary analysis. Cytotherapy 2024; 26:444-455. [PMID: 38363248 DOI: 10.1016/j.jcyt.2024.02.001] [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: 11/12/2023] [Revised: 02/01/2024] [Accepted: 02/01/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND AIMS Coronavirus disease 2019 (COVID-19) is characterized by a broad spectrum of clinical manifestations with the potential to progress to multiple organ dysfunction in severe cases. Extracellular vesicles (EVs) carry a range of biological cargoes, which may be used as biomarkers of disease state. METHODS An exploratory secondary analysis of the SARITA-2 and SARITA-1 datasets (randomized clinical trials on patients with mild and moderate/severe COVID-19) was performed. Serum-derived EVs were used for proteomic analysis to identify enriched biological processes and key proteins, thus providing insights into differences in disease severity. Serum-derived EVs were separated from patients with COVID-19 by size exclusion chromatography and nanoparticle tracking analysis was used to determine particle concentration and diameter. Liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) was performed to identify and quantify protein signatures. Bioinformatics and multivariate statistical analysis were applied to distinguish candidate proteins associated with disease severity (mild versus moderate/severe COVID-19). RESULTS No differences were observed in terms of the concentration and diameter of enriched EVs between mild (n = 14) and moderate/severe (n = 30) COVID-19. A total of 414 proteins were found to be present in EVs, of which 360 were shared while 48 were uniquely present in severe/moderate compared to mild COVID-19. The main biological signatures in moderate/severe COVID-19 were associated with platelet degranulation, exocytosis, complement activation, immune effector activation, and humoral immune response. Von Willebrand factor, serum amyloid A-2 protein, histone H4 and H2A type 2-C, and fibrinogen β-chain were the most differentially expressed proteins between severity groups. CONCLUSION Exploratory proteomic analysis of serum-derived EVs from patients with COVID-19 detected key proteins related to immune response and activation of coagulation and complement pathways, which are associated with disease severity. Our data suggest that EV proteins may be relevant biomarkers of disease state and prognosis.
Collapse
Affiliation(s)
- Adriana F Paes Leme
- Laboratório Nacional de Biociências - LNBio, Centro Nacional de Pesquisa em Energia e Materiais - CNPEM, Campinas, São Paulo, Brazil
| | - Sami Yokoo
- Laboratório Nacional de Biociências - LNBio, Centro Nacional de Pesquisa em Energia e Materiais - CNPEM, Campinas, São Paulo, Brazil
| | - Ana Gabriela C Normando
- Laboratório Nacional de Biociências - LNBio, Centro Nacional de Pesquisa em Energia e Materiais - CNPEM, Campinas, São Paulo, Brazil
| | - João Vitor S Ormonde
- Laboratório Nacional de Biociências - LNBio, Centro Nacional de Pesquisa em Energia e Materiais - CNPEM, Campinas, São Paulo, Brazil
| | - Romenia Ramos Domingues
- Laboratório Nacional de Biociências - LNBio, Centro Nacional de Pesquisa em Energia e Materiais - CNPEM, Campinas, São Paulo, Brazil
| | - Fernanda F Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSaúde, Research Support Foundation of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro L Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSaúde, Research Support Foundation of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruno S F Souza
- Goncalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, Brazil; D'Or Institute for Research and Education (IDOR), Salvador, Bahia, Brazil; Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Bahia, Brazil
| | - Claudia C Dos Santos
- The Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada; Institute of Medical Sciences and Interdepartmental Division of Critical Care, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | | | - Miquéias Lopes-Pacheco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSaúde, Research Support Foundation of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
18
|
Gygi JP, Maguire C, Patel RK, Shinde P, Konstorum A, Shannon CP, Xu L, Hoch A, Jayavelu ND, Haddad EK, IMPACC Network, Reed EF, Kraft M, McComsey GA, Metcalf JP, Ozonoff A, Esserman D, Cairns CB, Rouphael N, Bosinger SE, Kim-Schulze S, Krammer F, Rosen LB, van Bakel H, Wilson M, Eckalbar WL, Maecker HT, Langelier CR, Steen H, Altman MC, Montgomery RR, Levy O, Melamed E, Pulendran B, Diray-Arce J, Smolen KK, Fragiadakis GK, Becker PM, Sekaly RP, Ehrlich LI, Fourati S, Peters B, Kleinstein SH, Guan L. Integrated longitudinal multiomics study identifies immune programs associated with acute COVID-19 severity and mortality. J Clin Invest 2024; 134:e176640. [PMID: 38690733 PMCID: PMC11060740 DOI: 10.1172/jci176640] [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: 10/26/2023] [Accepted: 03/12/2024] [Indexed: 05/03/2024] Open
Abstract
BACKGROUNDPatients hospitalized for COVID-19 exhibit diverse clinical outcomes, with outcomes for some individuals diverging over time even though their initial disease severity appears similar to that of other patients. A systematic evaluation of molecular and cellular profiles over the full disease course can link immune programs and their coordination with progression heterogeneity.METHODSWe performed deep immunophenotyping and conducted longitudinal multiomics modeling, integrating 10 assays for 1,152 Immunophenotyping Assessment in a COVID-19 Cohort (IMPACC) study participants and identifying several immune cascades that were significant drivers of differential clinical outcomes.RESULTSIncreasing disease severity was driven by a temporal pattern that began with the early upregulation of immunosuppressive metabolites and then elevated levels of inflammatory cytokines, signatures of coagulation, formation of neutrophil extracellular traps, and T cell functional dysregulation. A second immune cascade, predictive of 28-day mortality among critically ill patients, was characterized by reduced total plasma Igs and B cells and dysregulated IFN responsiveness. We demonstrated that the balance disruption between IFN-stimulated genes and IFN inhibitors is a crucial biomarker of COVID-19 mortality, potentially contributing to failure of viral clearance in patients with fatal illness.CONCLUSIONOur longitudinal multiomics profiling study revealed temporal coordination across diverse omics that potentially explain the disease progression, providing insights that can inform the targeted development of therapies for patients hospitalized with COVID-19, especially those who are critically ill.TRIAL REGISTRATIONClinicalTrials.gov NCT04378777.FUNDINGNIH (5R01AI135803-03, 5U19AI118608-04, 5U19AI128910-04, 4U19AI090023-11, 4U19AI118610-06, R01AI145835-01A1S1, 5U19AI062629-17, 5U19AI057229-17, 5U19AI125357-05, 5U19AI128913-03, 3U19AI077439-13, 5U54AI142766-03, 5R01AI104870-07, 3U19AI089992-09, 3U19AI128913-03, and 5T32DA018926-18); NIAID, NIH (3U19AI1289130, U19AI128913-04S1, and R01AI122220); and National Science Foundation (DMS2310836).
Collapse
Affiliation(s)
| | - Cole Maguire
- The University of Texas at Austin, Austin, Texas, USA
| | | | - Pramod Shinde
- La Jolla Institute for Immunology, La Jolla, California, USA
| | | | - Casey P. Shannon
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada
- Prevention of Organ Failure (PROOF) Centre of Excellence, Providence Research, Vancouver, British Columbia, Canada
| | - Leqi Xu
- Yale School of Public Health, New Haven, Connecticut, USA
| | - Annmarie Hoch
- Clinical and Data Coordinating Center (CDCC) and
- Precision Vaccines Program, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Elias K. Haddad
- Drexel University, Tower Health Hospital, Philadelphia, Pennsylvania, USA
| | - IMPACC Network
- The Immunophenotyping Assessment in a COVID-19 Cohort (IMPACC) Network is detailed in Supplemental Acknowledgments
| | - Elaine F. Reed
- David Geffen School of Medicine at the UCLA, Los Angeles, California, USA
| | - Monica Kraft
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Grace A. McComsey
- Case Western Reserve University and University Hospitals of Cleveland, Cleveland, Ohio, USA
| | - Jordan P. Metcalf
- Oklahoma University Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Al Ozonoff
- Clinical and Data Coordinating Center (CDCC) and
- Precision Vaccines Program, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Charles B. Cairns
- Drexel University, Tower Health Hospital, Philadelphia, Pennsylvania, USA
| | | | | | | | - Florian Krammer
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria
| | - Lindsey B. Rosen
- National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Harm van Bakel
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | | | | | | - Hanno Steen
- Precision Vaccines Program, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | | | - Ofer Levy
- Precision Vaccines Program, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Bali Pulendran
- Stanford University School of Medicine, Palo Alto, California, USA
| | - Joann Diray-Arce
- Clinical and Data Coordinating Center (CDCC) and
- Precision Vaccines Program, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kinga K. Smolen
- Precision Vaccines Program, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Patrice M. Becker
- National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Rafick P. Sekaly
- Case Western Reserve University and University Hospitals of Cleveland, Cleveland, Ohio, USA
| | | | - Slim Fourati
- Case Western Reserve University and University Hospitals of Cleveland, Cleveland, Ohio, USA
| | - Bjoern Peters
- La Jolla Institute for Immunology, La Jolla, California, USA
- Department of Medicine, UCSD, La Jolla, California, USA
| | | | - Leying Guan
- Yale School of Public Health, New Haven, Connecticut, USA
| |
Collapse
|
19
|
Iqbal Q, Mudatsir M, Harapan H, Nurjannah N, Maulana T. Hemostatic and liver function parameters as COVID-19 severity markers. NARRA J 2024; 4:e178. [PMID: 38798852 PMCID: PMC11125416 DOI: 10.52225/narra.v4i1.178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 10/04/2023] [Indexed: 05/29/2024]
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a disease newly discovered in December 2019 which affects coagulation cascade and liver functions. The aim of this study was to investigate the potential of hemostatic and liver function parameters as severity markers in COVID-19 patients. This study was an observational analytic with cohort retrospective design using total sampling method. Data were retrieved from medical record of COVID-19 patients admitted to provincial hospital in Banda Aceh, Indonesia from March 2020 to March 2022. There were 1208 data eligible for the study after applying certain criteria. Mann-Whitney, logistic regression, and receiving operating characteristic (ROC) analyses were used to analysis the data. Thrombocyte count (p<0.001), prothrombin time (p<0.001), activated partial thromboplastin time (p<0.001), D-dimer (p<0.001), fibrinogen (p<0.001), aspartate aminotransferase (p<0.001), and alanine transaminase (p<0.001) significantly increased in severe compared to mild COVID-19 patients. After being adjusted, age (odds ratio (OR); 1.026 (95% confidence interval (CI): 1.016-1.037) was the most significant factor in predicting COVID-19 severity. Fibrinogen (cut-off 526.5 mg/L) was the best parameter associated with COVID-19 severity with 70% sensitivity and 66.4% specificity. Meanwhile, D-dimer (cut-off 805 ng/mL) had a sensitivity of 72.3% and specificity of 66.4%. Combining the parameters resulted in improved sensitivity to 82.0% with a slight decline of specificity to 65.5%. In conclusion, fibrinogen and D-dimer level on admission could be used as biomarkers in predicting COVID-19 prognosis. Routine monitoring and evaluation of laboratory testing especially D-dimer and fibrinogen could be implemented in order to reduce morbidity and mortality rate of COVID-19.
Collapse
Affiliation(s)
- Qanita Iqbal
- Master Program of Public Health, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Mudatsir Mudatsir
- Medical Research Unit, School of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
- Tropical Disease Centre, School of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Harapan Harapan
- Medical Research Unit, School of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
- Tropical Disease Centre, School of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
- Department of Microbiology, School of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
- Tsunami and Disaster Mitigation Research Center, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Nurjannah Nurjannah
- Department of Public Health, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Teuku Maulana
- Department of Public Health, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| |
Collapse
|
20
|
Jalili S, Hashemi SMA, Sarvari J. SARS-COV-2 ORF9b Dysregulate Fibrinogen and Albumin Genes in a Liver Cell Line. Rep Biochem Mol Biol 2024; 13:51-58. [PMID: 39582820 PMCID: PMC11580134 DOI: 10.61186/rbmb.13.1.51] [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: 03/08/2024] [Accepted: 06/02/2024] [Indexed: 11/26/2024]
Abstract
Background Individuals experiencing severe cases of Coronavirus Disease 2019 (COVID-19) exhibited elevated fibrinogen levels and decreased albumin levels, potentially linked to the presence of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) proteins. Consequently, our study endeavors to examine the impact of SARS-CoV-2 ORF9b on the expression of fibrinogen and albumin genes within the Hep-G2 cell line. Methods In this study, the Hep-G2 liver cell line was utilized alongside the plasmid pcDNA3.1 hyg+ containing ORF9b from the SARS-CoV-2 strain originating in Wuhan. Transfection procedures were executed, and the transfected cells were selected utilizing hygromycin B. Validation of ORF9b expression was conducted through SYBR green-based real-time PCR, and the expression of the Fibrinogen α (FGA), Fibrinogen β (FGB), Fibrinogen γ (FGG), and Albumin (ALB) genes was quantified using the same method. Results The real-time PCR analysis revealed a significant upregulation of fibrinogen genes-α (P=0.03), β (P=0.02), and γ (P=0.029) in Hep-G2 cells containing ORF9b compared to control cells. Furthermore, the findings indicated a markedly lower expression level of albumin in Hep-G2 cells harboring ORF9b compared to the control cells (P=0.028). Conclusions The findings suggest that SARS-CoV-2 ORF9b could potentially influence the course of SARS-CoV-2 infection by triggering the expression of α, β, and γ fibrinogen gene chains while suppressing the albumin gene. Further investigations are warranted to validate these observations across various SARS-CoV-2 strains exhibiting differing levels of pathogenicity.
Collapse
Affiliation(s)
- Shirin Jalili
- Institute of police equipment and technologies, policing sciences and social studies research institute, Tehran, Iran.
| | - Seyed Mohammad Ali Hashemi
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Jamal Sarvari
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
21
|
Özmerdiven GE, İrvem A, Şahin AS. Risk analysis of candidemia and its effect on mortality in COVID 19 and non COVID 19 patients. IRANIAN JOURNAL OF MICROBIOLOGY 2024; 16:236-242. [PMID: 38854981 PMCID: PMC11162174 DOI: 10.18502/ijm.v16i2.15357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Background and Objectives Candidemia is the most common serious fungal infection in critically ill patients in intensive care units (ICU). It series fourth among bloodstream infectious agents. In this study, candidemia risk analysis was examined in COVID 19 and non-COVID 19 patients during the pandemic period. Materials and Methods COVID 19 and non-COVID 19 cases who were followed up with candidemia in the ICU of our hospital were retrospectively screened. Demographic data, intubation, central venous catheter (CVC), medications, and total parenteral nutrition (TPN) status were evaluated in terms of risk between the two groups. Isolated Candida species and susceptibilty were evaluated. Results When age, gender, medication, intubation, TPN and CVC were evaluated, no difference was seen in terms of risk. Differences were detected in terms of comorbidities. While the most frequently identified Candida species was C. albicans, the most frequently detected species in the COVID19 patient group was C. parapsilosis. Conclusion There was no difference in candidemia incidence and risk factors between the two groups. Since candidemias were evaluated in terms of comorbidities, it was determined that Diabetes Mellitus (DM) and chronic obstructive pulmoner disease (COPD) were more common in patients with COVID 19 and less common in coronary artery disease (CAD) and malignancy.
Collapse
Affiliation(s)
- Gülşah Ece Özmerdiven
- Department of Microbiology, Kanuni Sultan Suleyman Research and Training Hospital, Istanbul, Turkey
| | - Arzu İrvem
- Department of Microbiology, Kanuni Sultan Suleyman Research and Training Hospital, Istanbul, Turkey
| | - Ayça Sultan Şahin
- Department of Anesthesia and Reanimation, Kanuni Sultan Suleyman Research and Training Hospital, Istanbul, Turkey
| |
Collapse
|
22
|
Constantin L, Ungurianu A, Streinu-Cercel A, Săndulescu O, Aramă V, Margină D, Țârcomnicu I. Investigation of Serum Endocan Levels in SARS-CoV-2 Patients. Int J Mol Sci 2024; 25:3042. [PMID: 38474287 PMCID: PMC10932032 DOI: 10.3390/ijms25053042] [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: 02/15/2024] [Revised: 02/27/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024] Open
Abstract
Endocan is an endothelial-cell-specific proteoglycan (ESM-1) and has emerged as an endothelial dysfunction and inflammatory marker in recent years. Endocan can be used as a marker of inflammatory endothelial dysfunction in endothelium-dependent disease: cardiovascular disease, sepsis, lung and kidney disease and malignancies. Recent data suggest that endothelial dysfunction is a key mechanism in COVID-19 pathogenesis. Endotheliitis and thrombo-inflammation are associated with severe forms of SARS-CoV-2 infection, and endocan is currently under investigation as a potential diagnostic and prognostic marker. The aim of this study was to determine serum endocan levels in patients with COVID-19 to evaluate the correlation between endocan levels and clinical disease diagnosis and prognosis. This study enrolled 56 patients, divided into three groups depending on disease severity: mild (15), moderate (25) and severe (16). The biochemical, demographic, clinical and imagistic data were collected and evaluated in correlation with the endocan levels. Serum endocan levels were significantly higher in the COVID-19 patients compared to the control group; also, endocan concentration correlated with vaccination status. The results revealed significantly elevated serum endocan levels in COVID-19 patients compared to the control group, with a correlation observed between endocan concentration and vaccination status. These findings suggest that endocan may serve as a novel biomarker for detecting inflammation and endothelial dysfunction risk in COVID-19 patients. There was no significant relationship between serum endocan levels and disease severity or the presence of cardiovascular diseases. Endocan can be considered a novel biomarker for the detection of inflammation and endothelial dysfunction risk in COVID-19 patients.
Collapse
Affiliation(s)
- Laura Constantin
- National Institute of Infectious Diseases “Prof. Dr. Matei Bals”, 021105 Bucharest, Romania; (L.C.); (A.S.-C.); (V.A.); (I.Ț.)
- Department of Biochemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 020956 Bucharest, Romania;
| | - Anca Ungurianu
- Department of Biochemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 020956 Bucharest, Romania;
| | - Anca Streinu-Cercel
- National Institute of Infectious Diseases “Prof. Dr. Matei Bals”, 021105 Bucharest, Romania; (L.C.); (A.S.-C.); (V.A.); (I.Ț.)
- Department of Biochemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 020956 Bucharest, Romania;
| | - Oana Săndulescu
- National Institute of Infectious Diseases “Prof. Dr. Matei Bals”, 021105 Bucharest, Romania; (L.C.); (A.S.-C.); (V.A.); (I.Ț.)
- Department of Biochemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 020956 Bucharest, Romania;
| | - Victoria Aramă
- National Institute of Infectious Diseases “Prof. Dr. Matei Bals”, 021105 Bucharest, Romania; (L.C.); (A.S.-C.); (V.A.); (I.Ț.)
- Department of Biochemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 020956 Bucharest, Romania;
| | - Denisa Margină
- Department of Biochemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 020956 Bucharest, Romania;
| | - Isabela Țârcomnicu
- National Institute of Infectious Diseases “Prof. Dr. Matei Bals”, 021105 Bucharest, Romania; (L.C.); (A.S.-C.); (V.A.); (I.Ț.)
| |
Collapse
|
23
|
El-Molla A, Fetouh FA, Bawazir S, Ali Y, Alwahby Y, Bahadeg M, Gotah Y, Badahdah FA, Alsaeed AH, Basseet A. Role of epinephrine in attenuating cytokine storm, decreasing ferritin, and inhibiting ferroptosis in SARS-CoV-2. Egypt Heart J 2024; 76:22. [PMID: 38376738 PMCID: PMC10879067 DOI: 10.1186/s43044-024-00455-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 02/16/2024] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the virus responsible for coronavirus disease 2019. It presents one of the most threatening pandemics in the history of humanity. The mortality and morbidity represent an unprecedented challenge to the modern medical era. SARS-CoV-2 results in acute respiratory distress syndrome, high concentrations of proinflammatory mediators, cytokine storm (CS) due to massive release of cytokines, hypercoagulation, and hemoglobin disintegration. Dysregulation of iron homeostasis, iron overload as indicated by high ferritin level, and ferroptosis are major factors in the pathogenesis of the disease. We report a case of SARS-CoV-2 in which the use of epinephrine (Epi) resulted in an unexpected attenuation of CS, decreasing ferritin level and inhibiting ferroptosis. CASE PRESENTATION A 64-year-old male patient with a history of multiple medical comorbidities had been diagnosed with SARS-CoV-2. Further evaluation showed marked increase in inflammatory markers, severe hyperferritinemia, and lymphopenia in laboratory blood tests. The characteristic score of CS was strongly positive, and in addition to regular treatment, the patient received Epi due to development of acute generalized skin rash, severe itching, and edema of lips and tongue. Epi may have successfully terminated not only the acute cutaneous condition, but also have attenuated CS, decreased ferritin level, and other inflammatory markers in addition to complete patient's recovery. CONCLUSION Epinephrine may attenuate CS and inhibit ferroptosis which is an iron-dependent, non-apoptotic mode of cell death. Epi interacts with ferric and/or ferrous iron and built a stable complex that impedes activation of beta-adrenergic receptors. Epi may cause marked decrease of ferritin and other inflammatory markers. Epi may be used to decrease iron overload which is associated with many medical diseases like type 2 diabetes mellitus and cardiometabolic diseases such as coronary heart disease and cerebrovascular disease. As a new clinical indication extensive studies are required for further assessment and possible therapeutic uses.
Collapse
Affiliation(s)
| | | | - Samir Bawazir
- Department of Otorhinolaryngology, Head and Neck Surgery, Prince Sultan Military Medical City, Riyadh, Kingdom of Saudi Arabia
| | - Yasser Ali
- Prince Sultan Military Medical City, Riyadh, Kingdom of Saudi Arabia
| | - Yehya Alwahby
- King Fahd Medical City, Riyadh, Kingdom of Saudi Arabia
| | - Muhammad Bahadeg
- Prince Sultan Military Medical City, Riyadh, Kingdom of Saudi Arabia
| | - Yousef Gotah
- Prince Sultan Military Medical City, Riyadh, Kingdom of Saudi Arabia
| | | | - Abdullatif H Alsaeed
- King Faisal Specialist Hospital and Research Center (KFSHRC), Jeddah, Kingdom of Saudi Arabia
| | | |
Collapse
|
24
|
Valencia I, Lumpuy-Castillo J, Magalhaes G, Sánchez-Ferrer CF, Lorenzo Ó, Peiró C. Mechanisms of endothelial activation, hypercoagulation and thrombosis in COVID-19: a link with diabetes mellitus. Cardiovasc Diabetol 2024; 23:75. [PMID: 38378550 PMCID: PMC10880237 DOI: 10.1186/s12933-023-02097-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/14/2023] [Indexed: 02/22/2024] Open
Abstract
Early since the onset of the COVID-19 pandemic, the medical and scientific community were aware of extra respiratory actions of SARS-CoV-2 infection. Endothelitis, hypercoagulation, and hypofibrinolysis were identified in COVID-19 patients as subsequent responses of endothelial dysfunction. Activation of the endothelial barrier may increase the severity of the disease and contribute to long-COVID syndrome and post-COVID sequelae. Besides, it may cause alterations in primary, secondary, and tertiary hemostasis. Importantly, these responses have been highly decisive in the evolution of infected patients also diagnosed with diabetes mellitus (DM), who showed previous endothelial dysfunction. In this review, we provide an overview of the potential triggers of endothelial activation related to COVID-19 and COVID-19 under diabetic milieu. Several mechanisms are induced by both the viral particle itself and by the subsequent immune-defensive response (i.e., NF-κB/NLRP3 inflammasome pathway, vasoactive peptides, cytokine storm, NETosis, activation of the complement system). Alterations in coagulation mediators such as factor VIII, fibrin, tissue factor, the von Willebrand factor: ADAMST-13 ratio, and the kallikrein-kinin or plasminogen-plasmin systems have been reported. Moreover, an imbalance of thrombotic and thrombolytic (tPA, PAI-I, fibrinogen) factors favors hypercoagulation and hypofibrinolysis. In the context of DM, these mechanisms can be exacerbated leading to higher loss of hemostasis. However, a series of therapeutic strategies targeting the activated endothelium such as specific antibodies or inhibitors against thrombin, key cytokines, factor X, complement system, the kallikrein-kinin system or NETosis, might represent new opportunities to address this hypercoagulable state present in COVID-19 and DM. Antidiabetics may also ameliorate endothelial dysfunction, inflammation, and platelet aggregation. By improving the microvascular pathology in COVID-19 and post-COVID subjects, the associated comorbidities and the risk of mortality could be reduced.
Collapse
Affiliation(s)
- Inés Valencia
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, IIS Hospital Universitario de La Princesa, 28009, Madrid, Spain.
| | - Jairo Lumpuy-Castillo
- Laboratory of Diabetes and Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040, Madrid, Spain
- Spanish Biomedical Research Centre On Diabetes and Associated Metabolic Disorders (CIBERDEM) Network, Madrid, Spain
| | - Giselle Magalhaes
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, 28029, Madrid, Spain
| | - Carlos F Sánchez-Ferrer
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, 28029, Madrid, Spain
- Vascular Pharmacology and Metabolism (FARMAVASM), IdiPAZ, Madrid, Spain
| | - Óscar Lorenzo
- Laboratory of Diabetes and Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040, Madrid, Spain.
- Spanish Biomedical Research Centre On Diabetes and Associated Metabolic Disorders (CIBERDEM) Network, Madrid, Spain.
| | - Concepción Peiró
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, 28029, Madrid, Spain.
- Vascular Pharmacology and Metabolism (FARMAVASM), IdiPAZ, Madrid, Spain.
| |
Collapse
|
25
|
Risman RA, Belcher HA, Ramanujam RK, Weisel JW, Hudson NE, Tutwiler V. Comprehensive Analysis of the Role of Fibrinogen and Thrombin in Clot Formation and Structure for Plasma and Purified Fibrinogen. Biomolecules 2024; 14:230. [PMID: 38397467 PMCID: PMC10886591 DOI: 10.3390/biom14020230] [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: 01/04/2024] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Altered properties of fibrin clots have been associated with bleeding and thrombotic disorders, including hemophilia or trauma and heart attack or stroke. Clotting factors, such as thrombin and tissue factor, or blood plasma proteins, such as fibrinogen, play critical roles in fibrin network polymerization. The concentrations and combinations of these proteins affect the structure and stability of clots, which can lead to downstream complications. The present work includes clots made from plasma and purified fibrinogen and shows how varying fibrinogen and activation factor concentrations affect the fibrin properties under both conditions. We used a combination of scanning electron microscopy, confocal microscopy, and turbidimetry to analyze clot/fiber structure and polymerization. We quantified the structural and polymerization features and found similar trends with increasing/decreasing fibrinogen and thrombin concentrations for both purified fibrinogen and plasma clots. Using our compiled results, we were able to generate multiple linear regressions that predict structural and polymerization features using various fibrinogen and clotting agent concentrations. This study provides an analysis of structural and polymerization features of clots made with purified fibrinogen or plasma at various fibrinogen and clotting agent concentrations. Our results could be utilized to aid in interpreting results, designing future experiments, or developing relevant mathematical models.
Collapse
Affiliation(s)
- Rebecca A. Risman
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, USA; (R.A.R.); (R.K.R.)
| | - Heather A. Belcher
- Department of Physics, East Carolina University, Greenville, NC 27858, USA; (H.A.B.); (N.E.H.)
| | - Ranjini K. Ramanujam
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, USA; (R.A.R.); (R.K.R.)
| | - John W. Weisel
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Nathan E. Hudson
- Department of Physics, East Carolina University, Greenville, NC 27858, USA; (H.A.B.); (N.E.H.)
| | - Valerie Tutwiler
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, USA; (R.A.R.); (R.K.R.)
| |
Collapse
|
26
|
Lu Z, Wang B, Liu M, Yu D, Li J. Correlation analysis between plasma biomarkers albumin, fibrinogen, and their ratio with postoperative delirium in patients undergoing non-cardiac surgery: a systematic review and meta-analysis. Am J Transl Res 2024; 16:363-373. [PMID: 38463596 PMCID: PMC10918125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/23/2024] [Indexed: 03/12/2024]
Abstract
OBJECTIVES This meta-analysis aimed to investigate the correlation between plasma biomarkers, such as albumin and fibrinogen, and their ratio with postoperative delirium (POD) in patients undergoing non-cardiac surgery. METHODS Relevant observational cohort studies were systematically searched in PubMed, EMBASE, CINAHL, and the Cochrane Library databases as of March 2023. This meta-analysis was conducted using RevMan 5.4.1 and Stata 15.0 software. For continuous variables with non-uniform units, the standardized mean difference (SMD) and 95% confidence intervals (CIs) were used; otherwise, the mean difference (MD) and 95% CIs were employed. The Newcastle-Ottawa Scale (NOS) was applied to assess the quality of included literature. RESULTS Eighteen studies encompassing 7,011 patients were included. The meta-analysis revealed significantly lower albumin levels (sixteen studies, 5,813 patients, SMD = -0.45, 95% CI = -0.64 to -0.26, P < 0.00001, I2 = 80%) and albumin-fibrinogen ratio (AFR) (four studies, 824 patients, MD = -0.62, 95% CI = -0.76 to -0.48, P = 0.56, I2 = 0%) in the delirious group. Conversely, higher fibrinogen concentrations (two studies, 441 patients, MD = 0.13, 95% CI = 0.02 to 0.24, P = 0.69, I2 = 0%) were observed in the delirious group. Due to high heterogeneity in albumin levels (P < 0.00001, I2 = 80%), we conducted a subgroup and sensitivity analysis, and confirmed that the association of albumin levels was not influenced by surgery type, design or delirium evaluation instruments. CONCLUSIONS Preoperative albumin, fibrinogen and AFR levels were associated with POD, potentially aiding in identifying high-risk patients and playing a key role in preventing POD.
Collapse
Affiliation(s)
- Zhenhui Lu
- Department of Anesthesiology, Hebei General HospitalShijiazhuang 050051, Hebei, China
| | - Bei Wang
- Department of Gynaecology, Hebei General HospitalShijiazhuang 050051, Hebei, China
| | - Meinv Liu
- Department of Anesthesiology, Hebei General HospitalShijiazhuang 050051, Hebei, China
| | - Dongdong Yu
- Department of Anesthesiology, Hebei General HospitalShijiazhuang 050051, Hebei, China
| | - Jianli Li
- Department of Anesthesiology, Hebei General HospitalShijiazhuang 050051, Hebei, China
| |
Collapse
|
27
|
Jukic I, Heffernan A, Schelling AF, Kokic Males V, Savicevic NJ, Kovacic V. Association between COVID-19 Infection or Vaccination Outcomes and Methylenetetrahydrofolate Reductase Gene Polymorphism: A Systematic Review of the Literature. J Pers Med 2023; 13:1687. [PMID: 38138914 PMCID: PMC10744904 DOI: 10.3390/jpm13121687] [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: 11/02/2023] [Revised: 11/24/2023] [Accepted: 12/03/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND Thrombosis is a detrimental sequala of COVID-19 infection; thus, prophylactic anti-coagulant therapy has been deemed mandatory in treatment unless serious contraindications are present. Susceptibility to thromboembolic events in COVID-19, or following COVID-19 vaccination, is likely attributable to an interplay of factors, including a patient's baseline clinical status and comorbidities, alongside genetic risk factors. In Europe, 8-20% of the population are homozygous for the MTHFR (methylene tetrahydrofolate reductase) variant, which compromises folate metabolism and elevates homocysteine levels. While heightened homocysteine levels are considered a risk factor for thromboembolic events, the precise clinical significance remains a contentious issue. However, recent research suggests elevated homocysteine levels may predict the course and severity of COVID-19 infection. Given the lack of reliable biomarkers predictive of COVID-19 thrombotic risk existing in practice, and the accessibility of MTHFR screening, we established two main outcomes for this study: (1) to determine the association between hereditary MTHFR mutations and COVID-19 severity and thromboembolic events and (2) to determine the link between MTHFR variants and adverse thrombotic events following COVID-19 vaccination. METHODS The review was conducted in accordance with PRISMA guidelines. Medline, Scopus, and Web of Science databases were searched from pandemic inception (11 March 2020) to 30 October 2023. Eligibility criteria were applied, and data extraction performed. RESULTS From 63 citations identified, a total of 14 articles met the full inclusion criteria (8 of which were cross-sectional or observational studies, and 6 were case studies or reports). Among the eight observational and cross-sectional studies evaluating the relationship between MTHFR variants (C667T; A1298C) and thromboembolic events in COVID-19 infection, four studies established a connection (n = 2200), while the remaining four studies failed to demonstrate any significant association (n = 38). CONCLUSIONS This systematic review demonstrated a possible association between the MTHFR gene variants and COVID-19 severity, thromboembolic events, and adverse events following vaccination. However, the paucity of robust data precluded any firm conclusions being drawn. Further prospective trials are required to determine the connection between the MTHFR gene variant and COVID-19 infection and vaccination outcomes.
Collapse
Affiliation(s)
- Ivana Jukic
- Internal Medicine Department, Gastroenterology Division, University Hospital of Split, 21000 Split, Croatia
- Department of Health Studies, University of Split, 21000 Split, Croatia;
| | - Aisling Heffernan
- School of Medicine, University of Split, 21000 Split, Croatia; (A.H.); (A.F.S.); (N.J.S.); (V.K.)
| | | | - Visnja Kokic Males
- Department of Health Studies, University of Split, 21000 Split, Croatia;
- Internal Medicine Department, Endocrinology Division, University Hospital of Split, 21000 Split, Croatia
| | - Nora Josipa Savicevic
- School of Medicine, University of Split, 21000 Split, Croatia; (A.H.); (A.F.S.); (N.J.S.); (V.K.)
| | - Vedran Kovacic
- School of Medicine, University of Split, 21000 Split, Croatia; (A.H.); (A.F.S.); (N.J.S.); (V.K.)
- Internal Medicine Department, Division of Emergency and Intensive Medicine with Clinical Pharmacology and Toxicology, University Hospital of Split, 21000 Split, Croatia
| |
Collapse
|
28
|
Gygi JP, Maguire C, Patel RK, Shinde P, Konstorum A, Shannon CP, Xu L, Hoch A, Jayavelu ND, Network I, Haddad EK, Reed EF, Kraft M, McComsey GA, Metcalf J, Ozonoff A, Esserman D, Cairns CB, Rouphael N, Bosinger SE, Kim-Schulze S, Krammer F, Rosen LB, van Bakel H, Wilson M, Eckalbar W, Maecker H, Langelier CR, Steen H, Altman MC, Montgomery RR, Levy O, Melamed E, Pulendran B, Diray-Arce J, Smolen KK, Fragiadakis GK, Becker PM, Augustine AD, Sekaly RP, Ehrlich LIR, Fourati S, Peters B, Kleinstein SH, Guan L. Integrated longitudinal multi-omics study identifies immune programs associated with COVID-19 severity and mortality in 1152 hospitalized participants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.03.565292. [PMID: 37986828 PMCID: PMC10659275 DOI: 10.1101/2023.11.03.565292] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Hospitalized COVID-19 patients exhibit diverse clinical outcomes, with some individuals diverging over time even though their initial disease severity appears similar. A systematic evaluation of molecular and cellular profiles over the full disease course can link immune programs and their coordination with progression heterogeneity. In this study, we carried out deep immunophenotyping and conducted longitudinal multi-omics modeling integrating ten distinct assays on a total of 1,152 IMPACC participants and identified several immune cascades that were significant drivers of differential clinical outcomes. Increasing disease severity was driven by a temporal pattern that began with the early upregulation of immunosuppressive metabolites and then elevated levels of inflammatory cytokines, signatures of coagulation, NETosis, and T-cell functional dysregulation. A second immune cascade, predictive of 28-day mortality among critically ill patients, was characterized by reduced total plasma immunoglobulins and B cells, as well as dysregulated IFN responsiveness. We demonstrated that the balance disruption between IFN-stimulated genes and IFN inhibitors is a crucial biomarker of COVID-19 mortality, potentially contributing to the failure of viral clearance in patients with fatal illness. Our longitudinal multi-omics profiling study revealed novel temporal coordination across diverse omics that potentially explain disease progression, providing insights that inform the targeted development of therapies for hospitalized COVID-19 patients, especially those critically ill.
Collapse
|
29
|
Liu J, Lai S, Wu P, Wang J, Wang J, Wang J, Zhang Y. Systematic oxidative stress indices predicts prognosis in patients with urothelial carcinoma of the upper urinary tract after radical nephroureterectomy. Eur J Med Res 2023; 28:469. [PMID: 37898799 PMCID: PMC10612206 DOI: 10.1186/s40001-023-01295-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 08/17/2023] [Indexed: 10/30/2023] Open
Abstract
BACKGROUND Oxidative stress plays an important role in the occurrence and development of malignancy. However, the relationship between oxidative stress and upper urinary tract urothelial carcinoma (UTUC) prognosis remains elusive. This study aimed to evaluate the prognostic value of systematic oxidative stress indices as a predictor of patient outcomes in UTUC after radical nephroureterectomy. METHODS Clinical data for 483 patients with UTUC who underwent radical nephroureterectomy were analyzed. Patients were categorized according to an optimal value of systematic oxidative stress indices (SOSIs), including fibrinogen (Fib), gamma-glutamyl transpeptidase (γ-GGT), creatinine (CRE), lactate dehydrogenase (LDH) and albumin (ALB). Kaplan-Meier analyses were used to investigate associations of SOSIs with overall survival (OS) and progression-free survival (PFS). Moreover, associations between SOSIs and OS and PFS were assessed with univariate and multivariate analyses. RESULTS High values of Fib, γ-GGT, CRE, and LDH, and low values of ALB were associated with reduced OS. SOSIs status correlated with age, tumor site, surgical approach, hydronephrosis, tumor size, T stage, and lymph node status. The Kaplan-Meier survival analysis showed a significant discriminatory ability for death and progression risks in the two groups based on SOSIs. Multivariate Cox proportional hazards models showed that SOSIs were an independent prognostic indicator for OS (p = 0.007) and PFS (p = 0.021). SOSIs and clinical variables were selected to establish a nomogram for OS. The 1-, 3-, and 5-year AUC values were 0.77, 0.78, and 0.81, respectively. Calibration curves of the nomogram showed high consistencies between the predicted and observed survival probability. Decision curve analysis curves showed that the nomogram could well predict the 1-year, 3-year, and 5-year OS. CONCLUSIONS SOSIs are an independent unfavorable predictor of OS and PFS in patients diagnosed with UTUC undergoing RNU. Therefore, incorporating SOSIs into currently available clinical parameters may improve clinical decision-making.
Collapse
Affiliation(s)
- Jianyong Liu
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of the Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Dong Dan, Beijing, 100730, People's Republic of China
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Beijing Hospital Continence Center, Beijing, People's Republic of China
| | - Shicong Lai
- Department of Urology, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Pengjie Wu
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of the Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Dong Dan, Beijing, 100730, People's Republic of China
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Beijing Hospital Continence Center, Beijing, People's Republic of China
| | - Jiawen Wang
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of the Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Dong Dan, Beijing, 100730, People's Republic of China
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Beijing Hospital Continence Center, Beijing, People's Republic of China
| | - Jianye Wang
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of the Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Dong Dan, Beijing, 100730, People's Republic of China.
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China.
- Beijing Hospital Continence Center, Beijing, People's Republic of China.
| | - Jianlong Wang
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of the Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Dong Dan, Beijing, 100730, People's Republic of China.
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China.
- Beijing Hospital Continence Center, Beijing, People's Republic of China.
| | - Yaoguang Zhang
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of the Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Dong Dan, Beijing, 100730, People's Republic of China.
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China.
- Beijing Hospital Continence Center, Beijing, People's Republic of China.
| |
Collapse
|
30
|
VanElzakker MB, Bues HF, Brusaferri L, Kim M, Saadi D, Ratai EM, Dougherty DD, Loggia ML. Neuroinflammation in post-acute sequelae of COVID-19 (PASC) as assessed by [ 11C]PBR28 PET correlates with vascular disease measures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.19.563117. [PMID: 37905031 PMCID: PMC10614970 DOI: 10.1101/2023.10.19.563117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has triggered a consequential public health crisis of post-acute sequelae of COVID-19 (PASC), sometimes referred to as long COVID. The mechanisms of the heterogeneous persistent symptoms and signs that comprise PASC are under investigation, and several studies have pointed to the central nervous and vascular systems as being potential sites of dysfunction. In the current study, we recruited individuals with PASC with diverse symptoms, and examined the relationship between neuroinflammation and circulating markers of vascular dysfunction. We used [11C]PBR28 PET neuroimaging, a marker of neuroinflammation, to compare 12 PASC individuals versus 43 normative healthy controls. We found significantly increased neuroinflammation in PASC versus controls across a wide swath of brain regions including midcingulate and anterior cingulate cortex, corpus callosum, thalamus, basal ganglia, and at the boundaries of ventricles. We also collected and analyzed peripheral blood plasma from the PASC individuals and found significant positive correlations between neuroinflammation and several circulating analytes related to vascular dysfunction. These results suggest that an interaction between neuroinflammation and vascular health may contribute to common symptoms of PASC.
Collapse
Affiliation(s)
- Michael B VanElzakker
- Division of Neurotherapeutics, Department of Psychiatry, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- PolyBio Research Foundation, Medford, MA, USA
| | - Hannah F Bues
- Division of Neurotherapeutics, Department of Psychiatry, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ludovica Brusaferri
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Computer Science And Informatics, School of Engineering, London South Bank University, London, UK
| | - Minhae Kim
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Deena Saadi
- Division of Neurotherapeutics, Department of Psychiatry, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eva-Maria Ratai
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Darin D Dougherty
- Division of Neurotherapeutics, Department of Psychiatry, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marco L Loggia
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
31
|
Guo H, Li W, Wang K, Nie Z, Zhang X, Bai S, Duan N, Li X, Hu B. Analysis of Risk Factors for Revitrectomy in Eyes with Diabetic Vitreous Hemorrhage. Diabetes Metab Syndr Obes 2023; 16:2865-2874. [PMID: 37753483 PMCID: PMC10518247 DOI: 10.2147/dmso.s429938] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 09/14/2023] [Indexed: 09/28/2023] Open
Abstract
Purpose We aimed to investigate the risk factors associated with revitrectomy in eyes with diabetic vitreous hemorrhage and to determine the prognosis of these patients at least one year postoperatively. Patients and Methods This retrospective case-control study had a minimum follow-up period of one year. Patients were divided into single vitrectomy group (control group, n=202) and revitrectomy group (case group, n=36) for analysis. The indications, number, and timing of revitrectomies were documented. And the revitrectomy group was further divided into two vitrectomies group (n=30) and three or more vitrectomies group (n=6). The best-corrected visual acuity (BCVA) at the last follow-up and the occurrence of neovascular glaucoma (NVG) were compared among the single vitrectomy, two vitrectomies and three or more vitrectomies groups. We conducted a thorough collection of patient data and used univariate and binary logistic regression analyses to identify the risk factors associated with revitrectomy. Results A total of 197 patients (238 eyes) were included. Thirty-six eyes (15.1%) required revitrectomy with six eyes (2.5%) undergoing three or more vitrectomies during the follow-up period. The median duration of the second vitrectomy was 3 (2-6) months. The indications for a second vitrectomy included 28 eyes (77.8%) of postoperative vitreous hemorrhage and 7 eyes (22.2%) combined with tractional retinal detachment. Patients undergoing three or more vitrectomies had significantly worse postoperative BCVA and a higher incidence of NVG (P<0.01). Fibrinogen> 4 g/L (P<0.001) and preoperative anti-vascular endothelial growth factor intravitreal injection (P=0.015) were independent risk factors for revitrectomy, and glycated hemoglobin A1c (HbA1c)>10% (P=0.049) showed significant difference only in univariate analysis. Conclusion Patients requiring revitrectomy tended to have higher fibrinogen levels, tightly adhered fibrovascular membranes, higher HbA1c levels, and worse prognoses.
Collapse
Affiliation(s)
- Haoxin Guo
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin University Eye Hospital, Tianjin, People’s Republic of China
| | - Wenbo Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin University Eye Hospital, Tianjin, People’s Republic of China
| | - Kuan Wang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin University Eye Hospital, Tianjin, People’s Republic of China
- Cangzhou Eye Hospital, Cangzhou, People’s Republic of China
| | - Zetong Nie
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin University Eye Hospital, Tianjin, People’s Republic of China
| | - Xiang Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin University Eye Hospital, Tianjin, People’s Republic of China
| | - Siqiong Bai
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin University Eye Hospital, Tianjin, People’s Republic of China
| | - Naxin Duan
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin University Eye Hospital, Tianjin, People’s Republic of China
| | - Xiaorong Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin University Eye Hospital, Tianjin, People’s Republic of China
| | - Bojie Hu
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin University Eye Hospital, Tianjin, People’s Republic of China
| |
Collapse
|
32
|
Jegorović B, Nikolić A, Milinković N, Ignjatović S, Šipetić-Grujičić S. The utility of serum amyloid A and other acute-phase reactants determination in ambulatory care COVID-19 patients. J Med Biochem 2023; 42:492-504. [PMID: 37790210 PMCID: PMC10542288 DOI: 10.5937/jomb0-42799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/13/2023] [Indexed: 10/05/2023] Open
Abstract
Background The unpredictable course of Coronavirus Disease 19 (COVID-19) is making good severity assessment tools crucial. This study aimed to assess the usefulness of serum amyloid A (SAA) and other acute-phase reactants (APRs) in ambulatory care COVID-19 patients and identified relationships between these markers and disease outcomes. Methods From August to November 2020, patients seen in the outpatient department of the Clinic for Infectious and Tropical Diseases (Belgrade, Serbia) with confirmed COVID-19 were included. Patients were classified into mild, moderate, and severe disease groups based on World Health Organization criteria. SAA, C-reactive protein (CRP), interleukin-6 (IL-6), procalcitonin (PCT), ferritin, fibrinogen, D-dimer, albumin, and transferrin were measured. The median values of all APRs were compared between COVID-19 severity groups, hospitalized and non-hospitalized patients, and survivors and non-survivors. The Receiver operator characteristic (ROC) curve analysis was used for the classification characteristics assessment of individual APRs for the severity of illness, hospitalization, and survival.
Collapse
Affiliation(s)
- Boris Jegorović
- University Clinical Center of Serbia, Clinic for Infectious and Tropical Diseases "Prof. Dr. Kosta Todorović", Belgrade
| | - Aleksandra Nikolić
- University of Belgrade, Faculty of Medicine, Institute for Epidemiology, Belgrade
| | - Neda Milinković
- University of Belgrade, Faculty of Pharmacy, Department of Medical Biochemistry, Belgrade
| | - Svetlana Ignjatović
- University of Belgrade, Faculty of Pharmacy, Department of Medical Biochemistry, Belgrade
| | | |
Collapse
|
33
|
Yang J, Li Y, Bhalla A, Maienschein-Cline M, Fukuchi KI. A novel co-culture model for investigation of the effects of LPS-induced macrophage-derived cytokines on brain endothelial cells. PLoS One 2023; 18:e0288497. [PMID: 37440496 PMCID: PMC10343049 DOI: 10.1371/journal.pone.0288497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
In order to study effects of macrophage-derived inflammatory mediators associated with systemic inflammation on brain endothelial cells, we have established a co-culture system consisting of bEnd.3 cells and LPS-activated Raw 264.7 cells and performed its cytokine profiling. The cytokine profile of the co-culture model was compared to that of mice treated with intraperitoneal LPS injection. We found that, among cytokines profiled, eight cytokines/chemokines were similarly upregulated in both in vivo mouse and in vitro co-culture model. In contrast to the co-culture model, the cytokine profile of a common mono-culture system consisting of only LPS-activated bEnd.3 cells had little similarity to that of the in vivo mouse model. These results indicate that the co-culture of bEnd.3 cells with LPS-activated Raw 264.7 cells is a better model than the common mono-culture of LPS-activated bEnd.3 cells to investigate the molecular mechanism in endothelial cells, by which systemic inflammation induces neuroinflammation. Moreover, fibrinogen adherence both to bEnd.3 cells in the co-culture and to brain blood vessels in a LPS-treated animal model of Alzheimer's disease increased. To the best of our knowledge, this is the first to utilize bEnd.3 cells co-cultured with LPS-activated Raw 264.7 cells as an in vitro model to investigate the consequence of macrophage-derived inflammatory mediators on brain endothelial cells.
Collapse
Affiliation(s)
- Junling Yang
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, Illinois, United states of America
| | - Yinchuan Li
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, Jiangsu, People’s Republic of China
| | - Ambuj Bhalla
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, Illinois, United states of America
| | - Mark Maienschein-Cline
- Research Informatics Core, Research Resources Center, University of Illinois Chicago, Chicago, Illinois, United States of America
| | - Ken-ichiro Fukuchi
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, Illinois, United states of America
| |
Collapse
|
34
|
Fernando MC, Hayes T, Besser M, Falter F. Comparison of Blood and Blood Product Transfusion in COVID-19 and Non-COVID-19 Patients Requiring Extracorporeal Membrane Oxygenation for Severe Respiratory Failure. J Clin Med 2023; 12:4667. [PMID: 37510781 PMCID: PMC10381132 DOI: 10.3390/jcm12144667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/08/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
COVID-19 has resulted in an exponential increase in patients with severe respiratory failure requiring extracorporeal membrane oxygenation (ECMO). Patients on ECMO regularly require high volumes of blood and blood products but, so far, there has been no comparison of transfusion requirements between COVID-19 and non-COVID-19. Using electronic patient records at two major UK ECMO centres, Royal Papworth Hospital and University Hospital South Manchester, we reviewed the transfusion requirements of patients requiring ECMO between January 2019 to December 2021. A total of 271 patients, including 168 COVID-19 patients were available for analysis. Since COVID-19 patients spent almost twice as long on ECMO (27.1 vs. 14.16 days, p ≤ 0.0001) we indexed transfusion in both groups to days on ECMO to allow comparison. COVID-19 patients required less red blood cells (RBC) per day (0.408 vs. 0.996, p = 0.0005) but more cryoprecipitate transfusions (0.117 vs. 0.106, p = 0.022) compared to non-COVID-19 patients. COVID-19 patients had more than double the mortality of non-COVID-19 patients (47% vs. 20.4%, p = 0.0001) and those who died during the study period had higher platelet transfusion requirements (p = 0.007) than their non-COVID-19 counterparts. Transfusion requirements and coagulopathy differ between COVID-19 and non-COVID-19 patients. The distinctly different transfusion patterns between the two groups remain difficult to interpret, but further investigations may help explain the haematological aspects of severe COVID-19 infection.
Collapse
Affiliation(s)
- Malindra C Fernando
- Department of Anaesthesia and Intensive Care, Royal Papworth Hospital, Cambridge CB2 0AY, UK
| | - Tim Hayes
- Department of Anaesthesia and Intensive Care, Manchester University Hospitals, Manchester M13 9WL, UK
| | - Martin Besser
- Department of Haematology and Blood Transfusion, Cambridge University Hospitals, Cambridge CB2 0QQ, UK
| | - Florian Falter
- Department of Anaesthesia and Intensive Care, Royal Papworth Hospital, Cambridge CB2 0AY, UK
| |
Collapse
|
35
|
Chen X, Zhao J, You Y, Li Z, Chen S. The Ratio of Fibrinogen to Albumin is Related to the Occurrence of Retinopathy in Type 2 Diabetic Patients. Diabetes Metab Syndr Obes 2023; 16:1859-1867. [PMID: 37384130 PMCID: PMC10295541 DOI: 10.2147/dmso.s407391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 06/06/2023] [Indexed: 06/30/2023] Open
Abstract
Purpose Type 2 diabetic retinopathy is a long-term chronic inflammatory disease. The aim of this study was to investigate the relationship between fibrinogen to albumin ratio (FAR) and retinopathy in type 2 diabetic patients. Methods This was a retrospective study that included 500 patients with type 2 diabetes mellitus (T2DM), and were divided into non-diabetic retinopathy group (NDR, n=297) and diabetic retinopathy group (DR, n=203) according to fundus examination findings, and the DR group was further divided into non-proliferative retinopathy group (NPDR, n=182) and proliferative retinopathy group (PDR, n=21). Baseline data of patients were collected, and the fibrinogen to albumin ratio (FAR) and neutrophil to lymphocyte ratio (NLR) were calculated to analyze the correlation between FAR and NLR and type 2 diabetic retinopathy. Results The FAR and NLR were significantly higher in the DR group compared with the NDR group (both P < 0.001). Spearman correlation analysis showed that FAR was positively correlated with NLR and DR (P < 0.05). As the FAR quartile increased, the prevalence of DR increased (14.8%, 16.7%, 25.1%, and 43.30%, respectively; P < 0.05). Multifactorial logistic regression analysis showed that FAR, diabetic course, systolic blood pressure (SBP) and diabetic peripheral neuropathy (DPN) were risk factors for the development of DR in patients with T2DM. The area under the ROC curve for FAR to predict DR progression was 0.708, with an optimal critical value of 7.04, and the area under the ROC curve for diabetes duration and SBP to predict DR was 0.705 and 0.588, respectively. Conclusion Our findings show for the first time that FAR is an independent risk factor for assessing DR in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Xiaoyi Chen
- Graduate School, Hebei North University, Zhangjiakou, 075000, People’s Republic of China
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, 050051, People’s Republic of China
| | - Jingyu Zhao
- Graduate School, North China University of Science and Technology, Tangshan, 063210, People’s Republic of China
| | - Yanxue You
- Graduate School, Hebei North University, Zhangjiakou, 075000, People’s Republic of China
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, 050051, People’s Republic of China
| | - Zelin Li
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, 050051, People’s Republic of China
| | - Shuchun Chen
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, 050051, People’s Republic of China
| |
Collapse
|
36
|
Valencia-Pérez Rea D, Falfán-Valencia R, Fricke-Galindo I, Buendía-Roldán I, Chávez-Galán L, Nava-Quiroz KJ, Alanis-Ponce J, Pérez-Rubio G. The rs16969968 Tobacco Smoking-Related Single-Nucleotide Variant Is Associated with Clinical Markers in Patients with Severe COVID-19. Int J Mol Sci 2023; 24:9811. [PMID: 37372959 PMCID: PMC10298344 DOI: 10.3390/ijms24129811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 05/26/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Tobacco smoking is the leading risk factor for many respiratory diseases. Several genes are associated with nicotine addiction, such as CHRNA5 and ADAM33. This research aims to evaluate the association of the polymorphisms rs16969968 (CHRNA5) and rs3918396 (ADAM33) in patients who developed severe COVID-19. We included 917 COVID-19 patients hospitalized with critical disease and oxygenation impairment. They were divided into two groups, tobacco-smoking (n = 257) and non-smoker (n = 660) patients. The genotype and allele frequencies of two single nucleotide variants, the rs16969968 (CHRNA5) and rs3918396 (ADAM33), were evaluated. The rs3918396 in ADAM33 does not show a significative association. We analyzed the study population according to the rs16969968 genotype (GA + AA, n = 180, and GG, n = 737). The erythrocyte sedimentation rate (ESR) shows statistical differences; the GA + AA group had higher values than the GG group (p = 0.038, 32 vs. 26 mm/h, respectively). The smoking patients and GA or AA genotype carriers had a high positive correlation (p < 0.001, rho = 0.753) between fibrinogen and C-reactive protein. COVID-19 patients and smokers carriers of one or two copies of the risk allele (rs16969968/A) have high ESR and a positive correlation between fibrinogen and C-reactive protein.
Collapse
Affiliation(s)
- Daniela Valencia-Pérez Rea
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (D.V.-P.R.); (R.F.-V.); (I.F.-G.); (K.J.N.-Q.); (J.A.-P.)
| | - Ramcés Falfán-Valencia
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (D.V.-P.R.); (R.F.-V.); (I.F.-G.); (K.J.N.-Q.); (J.A.-P.)
| | - Ingrid Fricke-Galindo
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (D.V.-P.R.); (R.F.-V.); (I.F.-G.); (K.J.N.-Q.); (J.A.-P.)
| | - Ivette Buendía-Roldán
- Translational Research Laboratory on Aging and Pulmonary Fibrosis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico;
| | - Leslie Chávez-Galán
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico;
| | - Karol J. Nava-Quiroz
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (D.V.-P.R.); (R.F.-V.); (I.F.-G.); (K.J.N.-Q.); (J.A.-P.)
| | - Jesús Alanis-Ponce
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (D.V.-P.R.); (R.F.-V.); (I.F.-G.); (K.J.N.-Q.); (J.A.-P.)
| | - Gloria Pérez-Rubio
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (D.V.-P.R.); (R.F.-V.); (I.F.-G.); (K.J.N.-Q.); (J.A.-P.)
| |
Collapse
|
37
|
Krishnan A, Schneider CV, Schattenberg JM, Alqahtani SA. Risk of severe disease and mortality of COVID-19 in patients with Budd-Chiari syndrome: A population-based matched cohort study. Liver Int 2023; 43:1141-1144. [PMID: 36825357 DOI: 10.1111/liv.15553] [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: 10/18/2022] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 02/25/2023]
Abstract
BACKGROUND & AIMS Budd-Chiari syndrome (BCS) is a rare and potentially life-threatening disorder characterized by obstruction of the hepatic outflow tract. It is unknown whether patients with BCS represent a high risk for severe disease and mortality from coronavirus disease 2019 (COVID-19). Thus, we aimed to assess hospitalization rates, severe disease, all-cause mortality, intensive care unit (ICU) requirement and acute kidney injury (AKI) from COVID-19 diagnoses. METHODS & RESULTS We identified 467 patients with BCS with COVID-19, 96 427 non-chronic liver disease (CLD) and 9652 non-BCS CLD. The BCS and non-CLD cohorts (n = 467 each) and BCS and non-BCS CLD (n = 440 each) were well balanced after propensity matching. When compared to the non-CLD cohort, the BCS group had a higher risk of all-cause mortality (5.1% vs. 2.4%, HR 2.18; 95% CI, 1.08-4.40), severe disease (6.0% vs. 2.4%, HR 2.20; 95% CI, 1.09-4.43), hospitalization (24.6% vs. 13.1%, HR 1.77; 95% CI, 1.30-2.42) and AKI (7.9% vs. 2.8%, HR 2.57; 95% CI, 1.37-4.85), but no significant differences in ICU requirements (2.4% vs. 2.1%, HR 0.75; 95% CI, 0.27-2.08) at 60-days time points. When compared to the non-BCS CLD cohort, the BCS group had a higher risk of all-cause mortality (3.6% vs. 2.5%, HR 3.94; 95% CI, 1.31-11.79), hospitalization (29.8% vs. 21.6%, HR 1.43; 95% CI, 1.09-1.86), but differences in ICU requirements (HR 0.90 (0.38-2.12)), AKI (HR 1.41 (0.86-2.30)) or severe disease (HR 1.92 (0.99-3.71)) did not reach statistical significance at 60-day follow up. CONCLUSION In conclusion, COVID-19 infection in patients with BCS is associated with poor outcomes. Patients with BCS infected with COVID-19 carry a significantly higher risk of hospitalization and all-cause mortality and a possible effect on severe disease and AKI compared with COVID-19 patients without CLD or with non-BCS-CLD.
Collapse
Affiliation(s)
- Arunkumar Krishnan
- Section of Gastroenterology and Hepatology, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| | - Carolin V Schneider
- Department of Medicine III, Gastroenterology, Metabolic diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Jörn M Schattenberg
- Metabolic Liver Research Program, Department of Medicine I, University Medical Center Mainz, Mainz, Germany
| | - Saleh A Alqahtani
- Liver Transplant Center, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
38
|
Porosnicu TM, Sirbu IO, Oancea C, Sandesc D, Bratosin F, Rosca O, Jipa D, Boeriu E, Bandi SSS, Pricop M. The Impact of Therapeutic Plasma Exchange on Inflammatory Markers and Acute Phase Reactants in Patients with Severe SARS-CoV-2 Infection. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59050867. [PMID: 37241099 DOI: 10.3390/medicina59050867] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/21/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023]
Abstract
Background and Objectives: Due to the poor prognosis and the very high mortality rate associated with severe SARS-CoV-2 infections, various regimens have been tried to stop the evolution of the inflammatory cascade, such as immunomodulatory therapy and plasma clearance of the acute phase reactants involved. Therefore, the objective of this review was to analyze the effects of using therapeutic plasma exchange (TPE), also known as plasmapheresis, on the inflammatory markers of critically ill COVID-19 patients admitted to the intensive care unit (ICU). Materials and Methods: A thorough scientific database search was performed, and it included a review of articles published on PubMed, Cochrane Database, Scopus, and Web of Science from the beginning of the COVID-19 pandemic in March 2020 until September 2022 that focused on the treatment of SARS-CoV-2 infections using plasma exchange for patients admitted to the ICU. The current study included original articles, reviews, editorials, and short or special communications regarding the topic of interest. Results: A total of 13 articles were selected after satisfying the inclusion criterion of three or more patients enrolled with clinically severe COVID-19 that were eligible for TPE. From the included articles, it was observed that TPE was used as a last-resort salvage therapy that can be regarded as an alternative treatment method when the standard management for these patients fails. TPE significantly decreased the inflammatory status as measured by Interleukin-6 (IL-6), C-reactive protein (CRP), lymphocyte count, and D-dimers, as well as improving the clinical status measured with PaO2/FiO2 and duration of hospitalization. The pooled mortality risk reduction after TPE was 20%. Conclusions: There are sufficient studies and evidence to show that TPE reduces inflammatory mediators and improves coagulation function and the clinical/paraclinical status. Nevertheless, although it was shown that TPE decreases the severe inflammatory status without significant complications, the improvement of survival rate remains unclear.
Collapse
Affiliation(s)
- Tamara Mirela Porosnicu
- Doctoral School, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
- Intensive Care Unit, "Pius Brinzeu" Emergency Clinical County Hospital, 300723 Timisoara, Romania
| | - Ioan Ovidiu Sirbu
- Center for Complex Network Sciences, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Cristian Oancea
- Center for Research and Innovation in Precision Medicine of Respiratory Disease, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Dorel Sandesc
- Department of Anesthesia and Intensive Care, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Felix Bratosin
- Department XIII, Discipline of Infectious Disease, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Ovidiu Rosca
- Department XIII, Discipline of Infectious Disease, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Daniel Jipa
- Intensive Care Unit, "Pius Brinzeu" Emergency Clinical County Hospital, 300723 Timisoara, Romania
| | - Estera Boeriu
- Department of Pediatrics, Discipline of Pediatric Oncology and Hematology, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Satya Sai Sri Bandi
- Malla Reddy Institute of Medical Sciences, Suraram Main Road 138, Hyderabad 500055, India
| | - Marius Pricop
- Discipline of Oral and Maxillo-Facial Surgery, Faculty of Dental Medicine, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| |
Collapse
|
39
|
Tartibian B, Azadpour N, Eslami R, Khayat SMA. Home-based exercise alters pulmonary function and cellular stress markers in overweight middle-aged men during covid-19 Home quarantine. BMC Sports Sci Med Rehabil 2023; 15:61. [PMID: 37081518 PMCID: PMC10116448 DOI: 10.1186/s13102-023-00673-9] [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: 10/23/2022] [Accepted: 04/13/2023] [Indexed: 04/22/2023]
Abstract
BACKGROUND This study aimed to investigate the effects of a combined home-based exercise program on potential indicators of severe coronavirus disease 2019 (COVID-19) in overweight middle-aged men during home quarantine caused by COVID-19. METHODS Forty men (aged 45-64 years) were assigned to the exercise (EXE, n = 20) or control (CON, n = 20) groups. A 6-week combined program was carried out three days/week, starting at 20 min per session at 50% maximal heart rate (HRmax) and advancing to 45 min at 70% HRmax. Pulmonary functional and cellular stress biomarkers were measured before and after the training program. Analysis of the covariance (ANCOVA) was used for comparison between the two groups considering the baseline values. RESULTS Thirty-six participants (EXE, n = 17; CON, n = 19) completed the research protocol. The EXE group showed post-training improvements in forced vital capacity (FVC), forced expiratory volume in 1 s (FEV1), FEV1/FVC, Vital capacity (VC), and Forced expiratory flow at 25-75% (FEF25-75) compared to the CON group (P < 0. 05). Further, the plasma levels of fibrinogen, Interleukin (IL)-6, Interleukin (IL)-1β, D-dimer, and angiotensin (Ang II) decreased in the EXE group compared to the CON group (P < 0. 05). After six weeks of the training program, leukocyte counts increased in the EXE group compared to the CON group (P < 0. 05). There was a significant positive correlation between body mass index (BMI) with cardiovascular and inflammatory biomarkers other than white blood cells (WBC) in the EXE group (P < 0.05). CONCLUSIONS The findings suggest that combined home-based exercise during home quarantine improves risk factors for severe COVID-19 in overweight middle-aged men. These improvements were further correlated with changes in BMI. Future research is required to confirm the findings of this study.
Collapse
Affiliation(s)
- Bakhtyar Tartibian
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Allameh Tabataba'i University, Tehran, Iran.
| | - Noushin Azadpour
- Department of Physiotherapy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Rasoul Eslami
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Allameh Tabataba'i University, Tehran, Iran
| | - Sirwan Mohammad Amini Khayat
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Urmia University, Urmia, Iran
| |
Collapse
|
40
|
Rizzi M, D'Onghia D, Tonello S, Minisini R, Colangelo D, Bellan M, Castello LM, Gavelli F, Avanzi GC, Pirisi M, Sainaghi PP. COVID-19 Biomarkers at the Crossroad between Patient Stratification and Targeted Therapy: The Role of Validated and Proposed Parameters. Int J Mol Sci 2023; 24:ijms24087099. [PMID: 37108262 PMCID: PMC10138390 DOI: 10.3390/ijms24087099] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/06/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Clinical knowledge about SARS-CoV-2 infection mechanisms and COVID-19 pathophysiology have enormously increased during the pandemic. Nevertheless, because of the great heterogeneity of disease manifestations, a precise patient stratification at admission is still difficult, thus rendering a rational allocation of limited medical resources as well as a tailored therapeutic approach challenging. To date, many hematologic biomarkers have been validated to support the early triage of SARS-CoV-2-positive patients and to monitor their disease progression. Among them, some indices have proven to be not only predictive parameters, but also direct or indirect pharmacological targets, thus allowing for a more tailored approach to single-patient symptoms, especially in those with severe progressive disease. While many blood test-derived parameters quickly entered routine clinical practice, other circulating biomarkers have been proposed by several researchers who have investigated their reliability in specific patient cohorts. Despite their usefulness in specific contexts as well as their potential interest as therapeutic targets, such experimental markers have not been implemented in routine clinical practice, mainly due to their higher costs and low availability in general hospital settings. This narrative review will present an overview of the most commonly adopted biomarkers in clinical practice and of the most promising ones emerging from specific population studies. Considering that each of the validated markers reflects a specific aspect of COVID-19 evolution, embedding new highly informative markers into routine clinical testing could help not only in early patient stratification, but also in guiding a timely and tailored method of therapeutic intervention.
Collapse
Affiliation(s)
- Manuela Rizzi
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Davide D'Onghia
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Stelvio Tonello
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Rosalba Minisini
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Donato Colangelo
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Mattia Bellan
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Luigi Mario Castello
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Francesco Gavelli
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Gian Carlo Avanzi
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Mario Pirisi
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Pier Paolo Sainaghi
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
| |
Collapse
|
41
|
Garyfallogiannis K, Ramanujam RK, Litvinov RI, Yu T, Nagaswami C, Bassani JL, Weisel JW, Purohit PK, Tutwiler V. Fracture toughness of fibrin gels as a function of protein volume fraction: Mechanical origins. Acta Biomater 2023; 159:49-62. [PMID: 36642339 PMCID: PMC11824895 DOI: 10.1016/j.actbio.2022.12.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 01/15/2023]
Abstract
The mechanical stability of blood clots necessary for their functions is provided by fibrin, a fibrous gel. Rupture of clots leads to life-threatening thrombotic embolization, which is little understood. Here, we combine experiments and simulations to determine the toughness of plasma clots as a function of fibrin content and correlate toughness with fibrin network structure characterized by confocal and scanning electron microscopy. We develop fibrin constitutive laws that scale with fibrin concentration and capture the force-stretch response of cracked clot specimens using only a few material parameters. Toughness is calculated from the path-independent J* integral that includes dissipative effects due to fluid flow and uses only the constitutive model and overall stretch at crack propagation as input. We show that internal fluid motion, which is not directly measurable, contributes significantly to clot toughness, with its effect increasing as fibrin content increases, because the reduced gel porosity at higher density results in greater expense of energy in fluid motion. Increasing fibrin content (1→10mg/mL) results in a significant increase in clot toughness (3→15 N/m) in accordance with a power law relation reminiscent of cellular solids and elastomeric gels. These results provide a basis for understanding and predicting the tendency for thrombotic embolization. STATEMENT OF SIGNIFICANCE: Fibrin, a naturally occurring biomaterial, is the major determinant of the structural and mechanical integrity of blood clots. We determined that increasing the fibrin content in clots, as in some thrombi and fibrin-based anti-bleeding sealants, results in an increase in clot toughness. Toughness corresponds to the ability to resist rupturing in the presence of a defect. We couple bulk mechanical testing, microstructural measurements, and finite element modeling to capture the force-stretch response of fibrin clots and compute toughness. We show that increased fibrin content in clots reduces porosity and limits fluid motion and that fluid motion drastically alters the clot toughness. These results provide a fundamental understanding of blood clot rupture and could help in rational design of fibrin-containing biomaterials.
Collapse
Affiliation(s)
| | - Ranjini K Ramanujam
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Rustem I Litvinov
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Tony Yu
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | | | - John L Bassani
- Department of Mechanical Engineering and Applied Mechanics, University of Pennsylvania, Philadelphia, PA, USA
| | - John W Weisel
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Prashant K Purohit
- Department of Mechanical Engineering and Applied Mechanics, University of Pennsylvania, Philadelphia, PA, USA
| | - Valerie Tutwiler
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
42
|
Guo M, Wu G, Tan Y, Li Y, Jin X, Qi W, Guo X, Zhang C, Zhu Z, Zhao L. Guild-Level Microbiome Signature Associated with COVID-19 Severity and Prognosis. mBio 2023; 14:e0351922. [PMID: 36744910 PMCID: PMC9973266 DOI: 10.1128/mbio.03519-22] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 01/13/2023] [Indexed: 02/07/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) severity has been associated with alterations of the gut microbiota. However, the relationship between gut microbiome alterations and COVID-19 prognosis remains elusive. Here, we performed a genome-resolved metagenomic analysis on fecal samples from 300 in-hospital COVID-19 patients, collected at the time of admission. Among the 2,568 high quality metagenome-assembled genomes (HQMAGs), redundancy analysis identified 33 HQMAGs which showed differential distribution among mild, moderate, and severe/critical severity groups. Co-abundance network analysis determined that the 33 HQMAGs were organized as two competing guilds. Guild 1 harbored more genes for short-chain fatty acid biosynthesis, and fewer genes for virulence and antibiotic resistance, compared with Guild 2. Based on average abundance difference between the two guilds, the guild-level microbiome index (GMI) classified patients from different severity groups (average AUROC [area under the receiver operating curve] = 0.83). Moreover, age-adjusted partial Spearman's correlation showed that GMIs at admission were correlated with 8 clinical parameters, which are predictors for COVID-19 prognosis, on day 7 in hospital. In addition, GMI at admission was associated with death/discharge outcome of the critical patients. We further validated that GMI was able to consistently classify patients with different COVID-19 symptom severities in different countries and differentiated COVID-19 patients from healthy subjects and pneumonia controls in four independent data sets. Thus, this genome-based guild-level signature may facilitate early identification of hospitalized COVID-19 patients with high risk of more severe outcomes at time of admission. IMPORTANCE Previous reports on the associations between COVID-19 and gut microbiome have been constrained by taxonomic-level analysis and overlook the interaction between microbes. By applying a genome-resolved, reference-free, guild-based metagenomic analysis, we demonstrated that the relationship between gut microbiota and COVID-19 is genome-specific instead of taxon-specific or even species-specific. Moreover, the COVID-19-associated genomes were not independent but formed two competing guilds, with Guild 1 potentially beneficial and Guild 2 potentially more detrimental to the host based on comparative genomic analysis. The dominance of Guild 2 over Guild 1 at time of admission was associated with hospitalized COVID-19 patients at high risk for more severe outcomes. Moreover, the guild-level microbiome signature is not only correlated with the symptom severity of COVID-19 patients, but also differentiates COVID-19 patients from pneumonia controls and healthy subjects across different studies. Here, we showed the possibility of using genome-resolved and guild-level microbiome signatures to identify hospitalized COVID-19 patients with a high risk of more severe outcomes at the time of admission.
Collapse
Affiliation(s)
- Mingquan Guo
- Department of Laboratory Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Guojun Wu
- Department of Biochemistry and Microbiology, School of Environmental and Biological Sciences and Center for Microbiome, Nutrition, and Health, New Jersey Institute for Food, Nutrition, and Health, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
- Rutgers-Jiaotong Joint Laboratory for Microbiome and Human Health, New Brunswick, New Jersey, USA
| | - Yun Tan
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai, China
| | - Yan Li
- State Key Laboratory of Microbial Metabolism and Ministry of Education Key Laboratory of Systems Biomedicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xin Jin
- Department of Laboratory Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Weiqiang Qi
- Department of Laboratory Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Xiaokui Guo
- School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenhong Zhang
- State Key Laboratory of Microbial Metabolism and Ministry of Education Key Laboratory of Systems Biomedicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhaoqin Zhu
- Department of Laboratory Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Liping Zhao
- State Key Laboratory of Microbial Metabolism and Ministry of Education Key Laboratory of Systems Biomedicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Department of Biochemistry and Microbiology, School of Environmental and Biological Sciences and Center for Microbiome, Nutrition, and Health, New Jersey Institute for Food, Nutrition, and Health, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
- Rutgers-Jiaotong Joint Laboratory for Microbiome and Human Health, New Brunswick, New Jersey, USA
| |
Collapse
|
43
|
Kim KS, Oh AR, Park J, Ryu JA. Association between Fibrinogen-to-Albumin Ratio and Prognosis in Patients Admitted to an Intensive Care Unit. J Clin Med 2023; 12:jcm12041407. [PMID: 36835941 PMCID: PMC9962887 DOI: 10.3390/jcm12041407] [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: 01/11/2023] [Revised: 02/05/2023] [Accepted: 02/09/2023] [Indexed: 02/12/2023] Open
Abstract
The objective of this study was to investigate the usefulness of fibrinogen-to-albumin ratio (FAR) as a prognostic marker in patients admitted to an intensive care unit (ICU) compared with Sequential Organ Failure Assessment (SOFA) score, a widely used prognostic scoring system. An inverse probability weighting (IPW) was used to control for selection bias and confounding factors. After IPW adjustment, the high FAR group showed significantly higher risk of 1-year compared with low FAR group (36.4% vs. 12.4%, adjust hazard ratio = 1.72; 95% confidence interval (CI): 1.59-1.86; p < 0.001). In the receiver-operating characteristic curve analysis associated with the prediction of 1-year mortality, there was no significant difference between the area under the curve of FAR on ICU admission (C-statistic: 0.684, 95% CI: 0.673-0.694) and that of SOFA score on ICU admission (C-statistic: 0.679, 95% CI: 0.669-0.688) (p = 0.532). In this study, FAR and SOFA score at ICU admission were associated with 1-year mortality in patients admitted to an ICU. Especially, FAR was easier to obtain in critically ill patients than SOFA score. Therefore, FAR is feasible and might help predict long-term mortality in these patients.
Collapse
Affiliation(s)
- Keun-Soo Kim
- Department of Critical Care Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Ah-Ran Oh
- Department of Anesthesiology and Pain Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Jungchan Park
- Department of Anesthesiology and Pain Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
- Correspondence: (J.P.); (J.-A.R.); Tel.: +82-2-3410-6399 (J.-A.R.); Fax: 82-2-2148-7088 (J.-A.R.)
| | - Jeong-Am Ryu
- Department of Critical Care Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
- Correspondence: (J.P.); (J.-A.R.); Tel.: +82-2-3410-6399 (J.-A.R.); Fax: 82-2-2148-7088 (J.-A.R.)
| |
Collapse
|
44
|
Qin R, He L, Yang Z, Jia N, Chen R, Xie J, Fu W, Chen H, Lin X, Huang R, Luo T, Liu Y, Yao S, Jiang M, Li J. Identification of Parameters Representative of Immune Dysfunction in Patients with Severe and Fatal COVID-19 Infection: a Systematic Review and Meta-analysis. Clin Rev Allergy Immunol 2023; 64:33-65. [PMID: 35040086 PMCID: PMC8763427 DOI: 10.1007/s12016-021-08908-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2021] [Indexed: 01/26/2023]
Abstract
Abnormal immunological indicators associated with disease severity and mortality in patients with COVID-19 have been reported in several observational studies. However, there are marked heterogeneities in patient characteristics and research methodologies in these studies. We aimed to provide an updated synthesis of the association between immune-related indicators and COVID-19 prognosis. We conducted an electronic search of PubMed, Scopus, Ovid, Willey, Web of Science, Cochrane library, and CNKI for studies reporting immunological and/or immune-related parameters, including hematological, inflammatory, coagulation, and biochemical variables, tested on hospital admission of COVID-19 patients with different severities and outcomes. A total of 145 studies were included in the current meta-analysis, with 26 immunological, 11 hematological, 5 inflammatory, 4 coagulation, and 10 biochemical variables reported. Of them, levels of cytokines, including IL-1β, IL-1Ra, IL-2R, IL-4, IL-6, IL-8, IL-10, IL-18, TNF-α, IFN-γ, IgA, IgG, and CD4+ T/CD8+ T cell ratio, WBC, neutrophil, platelet, ESR, CRP, ferritin, SAA, D-dimer, FIB, and LDH were significantly increased in severely ill patients or non-survivors. Moreover, non-severely ill patients or survivors presented significantly higher counts of lymphocytes, monocytes, lymphocyte/monocyte ratio, eosinophils, CD3+ T,CD4+T and CD8+T cells, B cells, and NK cells. The currently updated meta-analysis primarily identified a hypercytokinemia profile with the severity and mortality of COVID-19 containing IL-1β, IL-1Ra, IL-2R, IL-4, IL-6, IL-8, IL-10, IL-18, TNF-α, and IFN-γ. Impaired innate and adaptive immune responses, reflected by decreased eosinophils, lymphocytes, monocytes, B cells, NK cells, T cells, and their subtype CD4+ and CD8+ T cells, and augmented inflammation, coagulation dysfunction, and nonpulmonary organ injury, were marked features of patients with poor prognosis. Therefore, parameters of immune response dysfunction combined with inflammatory, coagulated, or nonpulmonary organ injury indicators may be more sensitive to predict severe patients and those non-survivors.
Collapse
Affiliation(s)
- Rundong Qin
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Li He
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhaowei Yang
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Nan Jia
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ruchong Chen
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jiaxing Xie
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wanyi Fu
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Hao Chen
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xinliu Lin
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Renbin Huang
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Tian Luo
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yukai Liu
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Siyang Yao
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Mei Jiang
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Jing Li
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
45
|
Li D, Li J, Zhao C, Liao X, Liu L, Xie L, Shang W. Diagnostic value of procalcitonin, hypersensitive C-reactive protein and neutrophil-to-lymphocyte ratio for bloodstream infections in pediatric tumor patients. Clin Chem Lab Med 2023; 61:366-376. [PMID: 36367370 DOI: 10.1515/cclm-2022-0801] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/24/2022] [Indexed: 11/13/2022]
Abstract
OBJECTIVES Bloodstream infection (BSI) is one of the major causes of death in pediatric tumor patients. Blood samples are relatively easy to obtain and thus provide a ready source of infection-related biological markers for the prompt evaluation of infection risk. METHODS A total of 259 pediatric tumor patients were included from May 2019 to March 2022. Patients were divided into BSI group (n=70) and control group (n=189). Clinical and biological data were collected using electronic medical records. Differences in biological markers between BSI group and control group and differences before and during infection in BSI group were analyzed. RESULTS The infected group showed higher levels of procalcitonin (PCT) and hypersensitive C-reactive-protein (hsCRP), and lower prealbumin (PA) than the uninfected group. Area under the receiver-operating curve (ROC) curves (AUC) of PCT, hsCRP and NLR (absolute neutrophil count to the absolute lymphocyte count) were 0.756, 0.617 and 0.612. The AUC of other biomarkers was ≤0.6. In addition, PCT, hsCRP, NLR and fibrinogen (Fg) were significantly increased during infection, while PA and lymphocyte (LYM) were significantly decreased. Antibiotic resistant of Gram-positive bacteria to CHL, SXT, OXA and PEN was lower than that of Coagulase-negative Staphylococcus. Resistant of Gram-positive bacteria to CHL was lower, while to SXT was higher than that of Gram-negative bacteria. CONCLUSIONS This study explored the utility of biomarkers to assist in diagnosis and found that the PCT had the greatest predictive value for infection in pediatric tumor patients with BSI. Additionally, the PCT, hsCRP, NLR, PA, LYM and Fg were changed by BSI.
Collapse
Affiliation(s)
- Dongmei Li
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, P.R. China
| | - Jie Li
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, P.R. China
| | - Chuanxi Zhao
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, P.R. China
| | - Xianglu Liao
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, P.R. China
| | - Lisheng Liu
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, P.R. China
| | - Li Xie
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, P.R. China
| | - Wenjing Shang
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, P.R. China
| |
Collapse
|
46
|
Krishnan A, Patel RA, Hadi YB, Mukherjee D, Shabih S, Thakkar S, Singh S, Woreta TA, Alqahtani SA. Clinical characteristics and outcomes of COVID-19 in patients with autoimmune hepatitis: A population-based matched cohort study. World J Hepatol 2023; 15:68-78. [PMID: 36744163 PMCID: PMC9896506 DOI: 10.4254/wjh.v15.i1.68] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/25/2022] [Accepted: 11/14/2022] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Patients with autoimmune hepatitis (AIH) require life-long immunosuppressive agents that may increase the risk of poor coronavirus disease 2019 (COVID-19) outcomes. There is a paucity of large data at the population level to assess whether patients with AIH have an increased risk of severe diseases. AIM To evaluate the impact of pre-existing AIH on the clinical outcomes of patients with COVID-19. METHODS We conducted a population-based, multicenter, propensity score-matched cohort study with consecutive adult patients (≥ 18 years) diagnosed with COVID-19 using the TriNeTx research network platform. The outcomes of patients with AIH (main group) were compared to a propensity score-matched cohort of patients: (1) Without chronic liver disease (CLD); and (2) Patients with CLD except AIH (non-AIH CLD) control groups. Each patient in the main group was matched to a patient in the control group using 1:1 propensity score matching to reduce confounding effects. The primary outcome was all-cause mortality, and secondary outcomes were hospitalization rate, need for critical care, severe disease, mechanical ventilation, and acute kidney injury (AKI). For each outcome, the risk ratio (RR) and confidence intervals (CI) were calculated to compare the association of AIH with the outcome. RESULTS We identified 375 patients with AIH, 1647915 patients with non-CLD, and 15790 patients with non-AIH CLD with COVID-19 infection. Compared to non-CLD patients, the AIH cohort had an increased risk of all-cause mortality (RR = 2.22; 95%CI: 1.07-4.61), hospitalization rate (RR = 1.78; 95%CI: 1.17-2.69), and severe disease (RR = 1.98; 95%CI: 1.19-3.26). The AIH cohort had a lower risk of hospitalization rate (RR = 0.72; 95%CI: 0.56-0.92), critical care (RR = 0.50; 95%CI: 0.32-0.79), and AKI (RR = 0.56; 95%CI: 0.35-0.88) compared to the non-AIH CLD patients. CONCLUSION Patients with AIH are associated with increased hospitalization risk, severe disease, and all-cause mortality compared to patients without pre-existing CLD from the diagnosis of COVID-19. However, patients with AIH were not at risk for worse outcomes with COVID-19 than other causes of CLD.
Collapse
Affiliation(s)
- Arunkumar Krishnan
- Section of Gastroenterology and Hepatology, West Virginia University School of Medicine, Morgantown, WV 26505, United States
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States.
| | - Ruhee A Patel
- Section of Gastroenterology and Hepatology, West Virginia University School of Medicine, Morgantown, WV 26505, United States
| | - Yousaf Bashir Hadi
- Section of Gastroenterology and Hepatology, West Virginia University School of Medicine, Morgantown, WV 26505, United States
| | - Diptasree Mukherjee
- Department of Medicine, Apex Institute of Medical Science, Kolkata 700075, West Bengal, India
| | - Sarah Shabih
- Section of Gastroenterology and Hepatology, West Virginia University School of Medicine, Morgantown, WV 26505, United States
| | - Shyam Thakkar
- Section of Gastroenterology and Hepatology, West Virginia University School of Medicine, Morgantown, WV 26505, United States
| | - Shailendra Singh
- Section of Gastroenterology and Hepatology, West Virginia University School of Medicine, Morgantown, WV 26505, United States
| | - Tinsay A Woreta
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Saleh A Alqahtani
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Liver Transplant Center, King Faisal Specialist Hospital and Research Center, Riyadh 12713, Saudi Arabia
| |
Collapse
|
47
|
Stanford S, Roy A, Rea C, Harris B, Ashton A, Mangles S, Everington T, Taher R, Burns D, Arbuthnot E, Cecil T. Pilot study to evaluate hypercoagulation and inflammation using rotational thromboelastometry and calprotectin in COVID-19 patients. PLoS One 2023; 18:e0269738. [PMID: 36607961 PMCID: PMC9821718 DOI: 10.1371/journal.pone.0269738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 12/21/2022] [Indexed: 01/07/2023] Open
Abstract
INTRODUCTION Abnormal coagulation and inflammation are hallmarks of SARs-COV-19. Stratifying affected patients on admission to hospital may help identify those who at are risk of developing severe disease early on. Rotational Thromboelastometry (ROTEM) is a point of care test that can be used to measure abnormal coagulation and calprotectin is a measure of inflammation. AIM Assess if ROTEM can measure hypercoagulability on admission and identify those who will develop severe disease early on. Assess if calprotectin can measure inflammation and if there is a correlation with ROTEM and calprotectin. METHODS COVID-19 patients were recruited on admission and ROTEM testing was undertaken daily for a period of 7 days. Additionally inflammatory marker calprotectin was also tested for the same period. RESULTS 33 patients were recruited to the study out of which 13 were admitted to ITU and 20 were treated on the ward. ROTEM detected a hypercoagulable state on admission but did not stratify between those admitted to a ward or escalated to ITU. Calprotectin levels were raised but there was no statistical difference (p = 0.73) between patients admitted to a ward or escalated to ITU. Significant correlations were observed between FIBA5 (r = 0.62; p<0.00), FIBCFT (r = -0.57; p<0.00), FIBMCF (r = 0.64; p<0.00) and INMCF (r = 0.57; p<0.00) and calprotectin. CONCLUSION COVID-19 patients were hypercoagulable on admission. The correlations between ROTEM and calprotectin underline the interactions between inflammation and coagulation.
Collapse
Affiliation(s)
- Sophia Stanford
- Peritoneal Malignancy Institute, Hampshire Hospitals NHS Foundation Trust, Basingstoke, Hampshire, United Kingdom
| | - Ashok Roy
- Peritoneal Malignancy Institute, Hampshire Hospitals NHS Foundation Trust, Basingstoke, Hampshire, United Kingdom
- * E-mail:
| | - Catherine Rea
- East Sussex Healthcare NHS Foundation Trust, Eastbourne, United Kingdom
| | - Ben Harris
- Anaesthetics and Critical Care, Hampshire Hospitals NHS Foundation Trust, Basingstoke, Hampshire, United Kingdom
| | - Antony Ashton
- Anaesthetics and Critical Care, Hampshire Hospitals NHS Foundation Trust, Basingstoke, Hampshire, United Kingdom
| | - Sarah Mangles
- Haemophilia, Haemostasis and Thrombosis Centre, Hampshire Hospitals NHS Foundation Trust, Basingstoke, Hampshire, United Kingdom
| | - Tamara Everington
- Haemophilia, Haemostasis and Thrombosis Centre, Hampshire Hospitals NHS Foundation Trust, Basingstoke, Hampshire, United Kingdom
| | - Rayan Taher
- Peritoneal Malignancy Institute, Hampshire Hospitals NHS Foundation Trust, Basingstoke, Hampshire, United Kingdom
| | - Daniel Burns
- Peritoneal Malignancy Institute, Hampshire Hospitals NHS Foundation Trust, Basingstoke, Hampshire, United Kingdom
| | - Emily Arbuthnot
- Peritoneal Malignancy Institute, Hampshire Hospitals NHS Foundation Trust, Basingstoke, Hampshire, United Kingdom
| | - Tom Cecil
- Peritoneal Malignancy Institute, Hampshire Hospitals NHS Foundation Trust, Basingstoke, Hampshire, United Kingdom
| |
Collapse
|
48
|
Significance of Catecholamine Biosynthetic/Metabolic Pathway in SARS-CoV-2 Infection and COVID-19 Severity. Cells 2022; 12:cells12010012. [PMID: 36611805 PMCID: PMC9818320 DOI: 10.3390/cells12010012] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
The SARS-CoV-2 infection was previously associated with the expression of the dopamine biosynthetic enzyme L-Dopa decarboxylase (DDC). Specifically, a negative correlation was detected between DDC mRNA and SARS-CoV-2 RNA levels in in vitro infected epithelial cells and the nasopharyngeal tissue of COVID-19 patients with mild/no symptoms. However, DDC, among other genes related to both DDC expression and SARS-CoV-2-infection (ACE2, dACE2, EPO), was upregulated in these patients, possibly attributed to an orchestrated host antiviral response. Herein, by comparing DDC expression in the nasopharyngeal swab samples of severe/critical to mild COVID-19 cases, we showed a 20 mean-fold reduction, highlighting the importance of the expression of this gene as a potential marker of COVID-19 severity. Moreover, we identified an association of SARS-CoV-2 infection with the expression of key catecholamine biosynthesis/metabolism-related genes, in whole blood samples from hospitalized patients and in cultured cells. Specifically, viral infection downregulated the biosynthetic part of the dopamine pathway (reduction in DDC expression up to 7.5 mean-fold), while enhanced the catabolizing part (increase in monoamine oxidases A and B expression up to 15 and 10 mean-fold, respectively) in vivo, irrespectively of the presence of comorbidities. In accordance, dopamine levels in the sera of severe cases were reduced (up to 3.8 mean-fold). Additionally, a moderate positive correlation between DDC and MAOA mRNA levels (r = 0.527, p < 00001) in the blood was identified upon SARS-CoV-2-infection. These observations were consistent to the gene expression data from SARS-CoV-2-infected Vero E6 and A549 epithelial cells. Furthermore, L-Dopa or dopamine treatment of infected cells attenuated the virus-derived cytopathic effect by 55% and 59%, respectively. The SARS-CoV-2 mediated suppression of dopamine biosynthesis in cell culture was, at least in part, attributed to hypoxia-like conditions triggered by viral infection. These findings suggest that L-Dopa/dopamine intake may have a preventive or therapeutic value for COVID-19 patients.
Collapse
|
49
|
Saha L, Vij S, Rawat K. Liver injury induced by COVID 19 treatment - what do we know? World J Gastroenterol 2022; 28:6314-6327. [PMID: 36533104 PMCID: PMC9753058 DOI: 10.3748/wjg.v28.i45.6314] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/07/2022] [Accepted: 11/17/2022] [Indexed: 12/02/2022] Open
Abstract
The severity of coronavirus disease 2019 (COVID-19) may be correlated with the risk of liver injury development. An increasing number of studies indicate that degrees of hepatotoxicity have been associated with using some medications in the management of COVID-19 patients. However, limited studies have systematically investigated the evidence of drug-induced liver injury (DILI) in COVID-19 patients. An increasing number of studies indicate that degrees of hepatotoxicity have been associated with using some of these medications in the management of COVID-19 patients. Significantly, it was relieved after the cessation of these agents. However, to our knowledge, no studies have systematically investigated the evidence of DILI in COVID-19 patients. In this review, we discussed the association between hepatotoxicity in COVID-19 patients and the drugs used in these patients and possible mechanisms of hepatotoxicity. The currently available evidence on the association of different therapeutic agents with hepatotoxicity in COVID-19 patient was systematically reviewed.
Collapse
Affiliation(s)
- Lekha Saha
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Soumya Vij
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Kajal Rawat
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| |
Collapse
|
50
|
Assante G, Tourna A, Carpani R, Ferrari F, Prati D, Peyvandi F, Blasi F, Bandera A, Le Guennec A, Chokshi S, Patel VC, Cox IJ, Valenti L, Youngson NA. Reduced circulating FABP2 in patients with moderate to severe COVID-19 may indicate enterocyte functional change rather than cell death. Sci Rep 2022; 12:18792. [PMID: 36335131 PMCID: PMC9637119 DOI: 10.1038/s41598-022-23282-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 10/25/2022] [Indexed: 11/08/2022] Open
Abstract
The gut is of importance in the pathology of COVID-19 both as a route of infection, and gut dysfunction influencing the severity of disease. Systemic changes caused by SARS-CoV-2 gut infection include alterations in circulating levels of metabolites, nutrients and microbial products which alter immune and inflammatory responses. Circulating plasma markers for gut inflammation and damage such as zonulin, lipopolysaccharide and β-glycan increase in plasma along with severity of disease. However, Intestinal Fatty Acid Binding Protein / Fatty Acid Binding Protein 2 (I-FABP/FABP2), a widely used biomarker for gut cell death, has paradoxically been shown to be reduced in moderate to severe COVID-19. We also found this pattern in a pilot cohort of mild (n = 18) and moderately severe (n = 19) COVID-19 patients in Milan from March to June 2020. These patients were part of the first phase of COVID-19 in Europe and were therefore all unvaccinated. After exclusion of outliers, patients with more severe vs milder disease showed reduced FABP2 levels (median [IQR]) (124 [368] vs. 274 [558] pg/mL, P < 0.01). A reduction in NMR measured plasma relative lipid-CH3 levels approached significance (median [IQR]) (0.081 [0.011] vs. 0.073 [0.024], P = 0.06). Changes in circulating lipid levels are another feature commonly observed in severe COVID-19 and a weak positive correlation was observed in the more severe group between reduced FABP2 and reduced relative lipid-CH3 and lipid-CH2 levels. FABP2 is a key regulator of enterocyte lipid import, a process which is inhibited by gut SARS-CoV-2 infection. We propose that the reduced circulating FABP2 in moderate to severe COVID-19 is a marker of infected enterocyte functional change rather than gut damage, which could also contribute to the development of hypolipidemia in patients with more severe disease.
Collapse
Affiliation(s)
- G Assante
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London, UK
- Faculty of Life Sciences & Medicine, King's College, London, UK
| | - A Tourna
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London, UK
- Faculty of Life Sciences & Medicine, King's College, London, UK
| | - R Carpani
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - F Ferrari
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - D Prati
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - F Peyvandi
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
- Department of Pathophysiology and Transplantation, Università Degli Studi Di Milano, Milan, Italy
| | - F Blasi
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
- Department of Pathophysiology and Transplantation, Università Degli Studi Di Milano, Milan, Italy
| | - A Bandera
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
- Department of Pathophysiology and Transplantation, Università Degli Studi Di Milano, Milan, Italy
| | - A Le Guennec
- Randall Centre for Cell & Molecular Biophysics, King's College, London, UK
| | - S Chokshi
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London, UK
- Faculty of Life Sciences & Medicine, King's College, London, UK
| | - V C Patel
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London, UK
- Faculty of Life Sciences & Medicine, King's College, London, UK
- Institute of Liver Studies, King's College Hospital, London, UK
| | - I J Cox
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London, UK.
- Faculty of Life Sciences & Medicine, King's College, London, UK.
| | - L Valenti
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy.
- Department of Pathophysiology and Transplantation, Università Degli Studi Di Milano, Milan, Italy.
| | - N A Youngson
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London, UK.
- Faculty of Life Sciences & Medicine, King's College, London, UK.
| |
Collapse
|