1
|
Gheraibia S, Belattar N, Hassan ME, El-Nekeety AA, El-Sawy ER, Abdel-Wahhab MA. Molecular docking of polyphenol compounds and exploring the anticoagulant activity of Costus speciosus extracts in vitro and in vivo. Toxicol Rep 2025; 14:101961. [PMID: 40092046 PMCID: PMC11908605 DOI: 10.1016/j.toxrep.2025.101961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 01/29/2025] [Accepted: 02/10/2025] [Indexed: 03/19/2025] Open
Abstract
Anticoagulants have an important role in the prevention of cardiovascular disorders. Costus speciosus (Costaceae) is a medicinal herb used to treat COVID-19-induced thrombosis. The purpose of this study was to assess the anticoagulant activity of various C. speciosus aqueous (CSAE), ethanol (SCEE), and methanol (CSME) extracts in vivo and in vitro utilizing thrombin time (TT), activated partial thromboplastin time (aPTT), and prothrombin time (PT). Different concentrations of the three extracts were used to evaluate the anticoagulation effects in vitro. In the in vivo assay, male Sprague Dawley rats were used to test the CSME as in vivo anticoagulants. Three groups of rats included the control group and the groups that received CSME daily at a low (200 mg/kg) or high dose (400 mg/kg b.w) for 2 weeks. The molecular docking of the major bioactive constituents of the methanolic extract against the binding site of the thrombin inhibitor complex was evaluated. The HPLC detected 13, 10 and 11 polyphenols in the methanolic, ethanolic and aqueous extracts, respectively. The in vitro results showed that all the studied extracts had anticoagulant activity and increased aPTT, TT, and PT time. The in vivo experiment supported the in vitro results and demonstrated that CSME greatly prolonged the anticoagulant characteristics when compared to the negative control. Both findings suggested that these extracts have significant anticoagulant activity, with CSME being more effective and potentially useful in pharmaceutical applications as a natural anticoagulant medication.
Collapse
Affiliation(s)
- Sara Gheraibia
- Laboratory of Applied Biochemistry, Faculty of Sciences of Nature and Life, Ferhat Abbes University, Setif 1, Algeria
| | - Noureddine Belattar
- Laboratory of Applied Biochemistry, Faculty of Sciences of Nature and Life, Ferhat Abbes University, Setif 1, Algeria
| | - Marwa E. Hassan
- Toxicology Department, Research Institute of Medical Entomology, Giza, Egypt
| | - Aziza A. El-Nekeety
- Food Toxicology & Contaminants Department, National Research Centre, Dokki, Cairo, Egypt
| | - Eslam R. El-Sawy
- Chemistry of Natural Products Department, National Research Centre, Dokki, Cairo, Egypt
| | - Mosaad A. Abdel-Wahhab
- Food Toxicology & Contaminants Department, National Research Centre, Dokki, Cairo, Egypt
| |
Collapse
|
2
|
Pecks U, Bohlmann MK, Andresen K, Büchel J, Bartmann C, Sitter M, Tihon A, Kranke P, Wöckel A, Hollweck R, Dressler-Steinbach I, Gruessner S, Gruber TM, Eichinger T, Manz J, Ruehl IM, Lihs A, Biermann AL, Bauerfeind LM, Oberste KM, Ramsauer B, Russe E, Schrey-Petersen S, Erol FM, Birdir C, Kaup L, Seliger G, Morfeld C, Berghaeuser MA, Richter MF, Jakubowski P, Linnemann B, Rath W. SARS-CoV-2 infection in pregnant women and incidence of thromboembolic disease: an analysis of the Covid-19-Related Obstetric and Neonatal Outcome Study (CRONOS) in Germany. Arch Gynecol Obstet 2025; 311:1667-1682. [PMID: 40131456 PMCID: PMC12055630 DOI: 10.1007/s00404-025-08007-5] [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: 01/27/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025]
Abstract
PURPOSE The aim of the present study was to quantify the rate of thromboembolic events (TE) in pregnant women with SARS-CoV-2 infection and to characterize risk factors to provide a basis for individualized recommendation on prophylactic measures. METHODS CRONOS is a multicenter, prospective observational study conducted in Germany and Austria during the COVID-19 pandemic. Pregnant women with confirmed SARS-CoV-2 infection were enrolled. Data on demographics, medical history, COVID-19-related aspects, and pregnancy and birth outcomes were collected. TE was particularly queried and used as the primary outcome. A combination of "TE," "maternal or fetal death," or "severe postpartum hemorrhage" was defined as a secondary endpoint. Risk analyses were performed using univariate and multivariable logistic regression models. RESULTS Data from 8033 pregnant patients showed 40 TEs (0.5% incidence). TE rates were 10% in ICU patients, 0.2-0.4% in those with moderate-to-mild COVID-19, and < 0.1% in asymptomatic women. Pulmonary embolism occurred in 21 cases, deep vein thrombosis in 12, and 7 had atypical or arterial TE. Risk factors included advanced gestational age, COVID-19 symptoms, hospitalization or ICU admission, premature birth, cesarean section, delivery within 4 weeks of infection, higher weight gain, anemia, and chronic inflammatory bowel disease. COVID-19 vaccination reduced risk. The logistic risk model yielded an AUC of 0.87 (95% CI 0.81-0.94). CONCLUSION The TE rate in pregnant women is largely determined by the severity of the disease. In asymptomatic or mild cases, other factors outweigh TE risk, while severe COVID-19 requiring ICU admission poses a high TE risk despite prophylaxis.
Collapse
Affiliation(s)
- Ulrich Pecks
- Department of Obstetrics and Gynecology, Germany AND Institut of Midwifery, University Hospital of Wuerzburg, Würzburg, Germany.
| | - Michael K Bohlmann
- Department of Obstetrics and Gynecology, St. Elisabethen-Krankenhaus Lörrach, Lörrach, Germany
| | - Kristin Andresen
- Department of Obstetrics and Gynecology, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Johanna Büchel
- Department of Obstetrics and Gynecology, University Hospital of Wuerzburg, Würzburg, Germany
| | - Catharina Bartmann
- Department of Obstetrics and Gynecology, University Hospital of Wuerzburg, Würzburg, Germany
| | - Magdalena Sitter
- Department of Anesthesiology, University Hospital of Wuerzburg, Würzburg, Germany
| | - Anastasia Tihon
- Department of Obstetrics and Gynecology, University Hospital of Wuerzburg, Würzburg, Germany
| | - Peter Kranke
- Department of Anesthesiology, University Hospital of Wuerzburg, Würzburg, Germany
| | - Achim Wöckel
- Department of Obstetrics and Gynecology, University Hospital of Wuerzburg, Würzburg, Germany
| | | | | | - Susanne Gruessner
- Department of Gynecology and Obstetrics, Klinikum Wilhelmshaven, Wilhelmshaven, Germany
| | | | - Teresa Eichinger
- Department of Gynecology, Obstetrics and Gynecological Endocrinology, Johannes Kepler University Linz, Linz, Austria
| | - Jula Manz
- Department of Obstetrics and Gynecology, Klinikum Darmstadt GmbH, Darmstadt, Germany
| | - Ina M Ruehl
- Department of Obstetrics, Red Cross Hospital "Taxisstrasse", Munich, Germany
| | - Angela Lihs
- Department of Gynecology and Obstetrics, City Hospital Sindelfingen-Boblingen, Boblingen, Germany
| | - Anna-Lena Biermann
- Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| | - Lea M Bauerfeind
- Department of Obstetrics and Gynecology, LMU University Hospital, Munich, Germany
| | - Kathleen M Oberste
- Department of Obstetrics and Gynecology, University Hospital Muenster, Muenster, Germany
| | - Babett Ramsauer
- Department of Obstetrics, Vivantes-Klinikum Neukölln, Berlin, Germany
| | - Eveline Russe
- Department of Obstetrics and Gynecology, St. Elisabeth-Hospital Lörrach, Lörrach, Germany
| | | | - Filiz Markfeld Erol
- Clinic for Gynaecology and Obstetrics, University Medical Center Freiburg, Freiburg, Germany
| | - Cahit Birdir
- Department of Obstetrics and Gynecology, University Clinic of Dresden, Dresden, Germany
| | - Lisa Kaup
- Department of Obstetrics and Gynecology, Dr Geisenhofer Clinic for Gynecology and Obstetrics, Munich, Germany
| | - Gregor Seliger
- Outpatient centre for women's health, fertility and pregnancy, University Medicine Halle, Halle (Saale), Germany
| | | | | | - Manuela F Richter
- Neonatology, AUF DER BULT-Children's and Youth Hospital, Hannover, Germany
| | - Peter Jakubowski
- Department of Obstetrics, University Hospital of Tuebingen, Tuebingen, Germany
| | - Birgit Linnemann
- Center for Cardiology and Angiology, Johannes-Gutenberg-University, Mainz, Germany
| | - Werner Rath
- Department of Obstetrics and Gynecology, University Hospital of Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
3
|
Hakimi P, Zaboli KA, Golbabapour‐Samakoush M, Azizimohammadi S, Soleimani F, Salmani MH, Teimoori‐Toolabi L. HLA Polymorphisms and COVID-19 Susceptibility and Severity: Insights From an Iranian Patients Cohort. J Cell Mol Med 2025; 29:e70570. [PMID: 40366340 PMCID: PMC12077277 DOI: 10.1111/jcmm.70570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 04/10/2025] [Accepted: 04/17/2025] [Indexed: 05/15/2025] Open
Abstract
The HLA system is a crucial immune response component against infectious agents, including SARS-CoV-2. Certain polymorphisms may impart varying levels of protection or vulnerability to COVID-19. This research aims to understand the possible relationship between HLA polymorphisms and the susceptibility to COVID-19 and its severity. We recruited 290 hospitalised Iranian COVID-19 patients (130 severe and 160 moderate). Using PCR-SSP methods, we conducted a detailed analysis of polymorphisms in HLA class I (HLA-A, HLA-B, and HLA-C) and II (HLA-DRB1 and HLA-DQB1) molecules at low resolution. The study found that certain HLA alleles, including HLA-B*49, HLA-B*52, HLA-C*12, HLA-DRB1*04, and HLA-DQB1*05, were associated with disease susceptibility. Additionally, HLA-A*23, DRB1*10, and DRB1*13 were indicators of disease severity. The study also noted that individuals carrying the HLA-A*23 allele showed a significant decrease in lymphocyte levels and an elevated likelihood of developing thrombosis. We hypothesise that a maladaptive immune response may occur based on these findings. This might be due to the strong affinity of the HLA-A*23 allele group for presenting a wide range of SARS-CoV-2 peptides. Such a presentation possibly leads to a cytokine storm, followed by lymphocyte apoptosis and an increase in platelet count.
Collapse
Affiliation(s)
- Pooria Hakimi
- Molecular Medicine Department, Biotechnology Research CenterPasteur Institute of IranTehranIran
| | - Kasra Arbabi Zaboli
- Molecular Medicine Department, Biotechnology Research CenterPasteur Institute of IranTehranIran
| | | | | | - Fatemeh Soleimani
- Molecular Medicine Department, Biotechnology Research CenterPasteur Institute of IranTehranIran
| | | | - Ladan Teimoori‐Toolabi
- Molecular Medicine Department, Biotechnology Research CenterPasteur Institute of IranTehranIran
| |
Collapse
|
4
|
Maier CL, Nakahara H, Barker NA, Auld SC, Truong AD, Friend S, Caridi-Scheible M, Connor M, Gaddh M, Cobb J, Polly DM, Guarner J, Powell C, Kempton CL, Daniels L, Wynn AT, Sniecinski R, Duncan A, Roback J, Masud T, Conlon S, Wade J, Wong A, Verkerke H, Zerra PE, Butler H, Sullivan HC, Easley KA, Josephson CD, Stowell SR. Therapeutic plasma exchange for fibrinogen-associated hyperviscosity: results of the COVID-19 PLasma EXchange (COPLEX) randomized controlled trial. J Thromb Haemost 2025; 23:1393-1400. [PMID: 39746400 DOI: 10.1016/j.jtha.2024.12.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 11/01/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Therapeutic plasma exchange (TPE) is the primary intervention for treating symptomatic hyperviscosity from hypergammaglobulinemia, yet its efficacy for treating hyperviscosity related to hyperfibrinogenemia is unclear. OBJECTIVES Define the safety and efficacy of TPE for critically ill COVID-19 patients with elevated blood viscosity from hyperfibrinogenemia. METHODS We performed a prospective randomized controlled trial in critically ill COVID-19 patients in a single US healthcare system. Patients with hyperfibrinogenemia (>800 mg/dL) or elevated plasma viscosity (2.3-3.5 centipoise [cP]) were randomized to receive TPE on 2 consecutive days or continued standard of care (SOC). RESULTS Twenty participants were enrolled, with 10 receiving TPE and 10 receiving SOC alone. Mean (±SEM) plasma viscosity decreased significantly from 2.35 cP (±0.12) to 1.61 cP (±0.03) in the TPE group and was unchanged in the SOC group (2.47 cP [±0.11] to 2.47 cP [±0.15]). Mean fibrinogen decreased from 934.0 mg/dL (±25.1) to 359.1 mg/dL (±22.5) after TPE vs from 859.6 mg/dL (±57.6) to 807.3 mg/dL (±63.1) in SOC. There was no significant difference in 28-day all-cause mortality between groups, with 2 deaths in the TPE cohort and 5 deaths in the SOC cohort (P = .13). No serious safety events related to TPE were reported. TPE significantly decreased biomarkers of inflammation (erythrocyte sedimentation rate and C-reactive protein) and endothelial activation (von Willebrand factor and factor VIII) but not hemostatic activation (prothrombin fragment 1.2, thrombin-antithrombin complex, and fibrin monomer) or immunoglobulin (IgG and IgM) levels. CONCLUSION TPE is safe and effective for normalizing elevated blood viscosity from hyperfibrinogenemia in COVID-19 patients. Additional studies are needed to determine the impact of TPE on overall patient outcomes, including in those with non-COVID-19 conditions associated with hyperfibrinogenemia.
Collapse
Affiliation(s)
- Cheryl L Maier
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, Georgia, USA.
| | - Hirotomo Nakahara
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, Georgia, USA; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Nicholas A Barker
- Department of Pharmaceutical Services, Emory Healthcare, Atlanta, Georgia, USA
| | - Sara C Auld
- Department of Medicine, Emory School of Medicine, Atlanta, Georgia, USA; Departments of Epidemiology and Global Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | | | - Sarah Friend
- Department of Hematology and Medical Oncology, Emory School of Medicine, Atlanta, Georgia, USA
| | | | - Michael Connor
- Department of Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Manila Gaddh
- Department of Hematology and Medical Oncology, Emory School of Medicine, Atlanta, Georgia, USA
| | - Jason Cobb
- Department of Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Derek M Polly
- Department of Pharmaceutical Services, Emory Healthcare, Atlanta, Georgia, USA
| | - Jeannette Guarner
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Cindy Powell
- Department of Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Christine L Kempton
- Department of Hematology and Medical Oncology, Emory School of Medicine, Atlanta, Georgia, USA
| | - Lisa Daniels
- Department of Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - A Thanushi Wynn
- Department of Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Roman Sniecinski
- Department of Anesthesiology, Emory School of Medicine, Atlanta, Georgia, USA
| | - Alexander Duncan
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - John Roback
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Tahsun Masud
- Department of Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Shaun Conlon
- Department of Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Jenna Wade
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Andrew Wong
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Hans Verkerke
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Patricia E Zerra
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Hailly Butler
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - H Cliff Sullivan
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Kirk A Easley
- Department of Biostatistics and Bioinformatics, Emory Rollins School of Public Health, Atlanta, Georgia, USA
| | - Cassandra D Josephson
- Cancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St. Petersburg, Florida, USA
| | - Sean R Stowell
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Vasković I, Marković M, Udovičić I, Arsenović L, Stojić M, Ignjatović A, Jovanović D, Nešković V. Effectiveness of different anticoagulation regimens in critically ill patients - experience from COVID 19 patients. Blood Coagul Fibrinolysis 2025; 36:82-89. [PMID: 40053316 DOI: 10.1097/mbc.0000000000001354] [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: 12/16/2024] [Accepted: 02/23/2025] [Indexed: 04/05/2025]
Abstract
This study compared the efficacy of therapeutic anticoagulation guided by anti-Xa levels vs. a D-dimer-based protocol in ICU patients with COVID-19. Given the heightened risk of thrombosis despite anticoagulation therapy in some cases, we hypothesised that anti-Xa measurement improves anticoagulation effectiveness and clinical outcomes in this population. We retrospectively analysed data from ICU patients at COVID Hospital Karaburma between April 2020 and December 2021. The primary outcome was the incidence of failed noninvasive ventilation necessitating intubation. Secondary endpoints included mortality rates, thromboembolic and bleeding complications, and anticoagulation effectiveness assessed by antifactor Xa activity. The analysis included 395 patients - 137 in the anti-Xa group and 258 in the D-dimer group. The D-dimer group showed a higher rate of failed noninvasive ventilation requiring intubation (65.7% vs. 50%, P = 0.009). The overall mortality was 48.3%, significantly higher in the D-dimer group (52.7%) compared to the anti-Xa group (40.1%, P = 0.02). Thromboembolic complications were lower in the anti-Xa group (2.9%) than in the D-dimer group (9.7%, P = 0.014), with no significant difference in bleeding. Following the first LMWH administration, 70.8% of patients had anti-Xa levels below the therapeutic and 11.7% below the prophylactic range. Anti-Xa-guided anticoagulation improves survival and reduces thromboembolic complications compared to D-dimer-based treatment without increasing bleeding risk. This study highlights the potential of the anti-Xa assay in managing anticoagulation in critically ill COVID-19 patients. Our findings provide a foundation for future research on using anti-Xa measurements as a guiding tool to optimise anticoagulation therapy in other critically ill populations.
Collapse
Affiliation(s)
- Igor Vasković
- Clinic for Anesthesiology and Intensive Care, Military Medical Academy
| | - Marija Marković
- Clinic for Anesthesiology and Intensive Care, Military Medical Academy
| | - Ivo Udovičić
- Clinic for Anesthesiology and Intensive Care, Military Medical Academy
| | | | - Mihailo Stojić
- Clinic for Anesthesiology and Intensive Care, Military Medical Academy
| | | | - Dragana Jovanović
- Clinic for Anesthesiology and Intensive Care, Military Medical Academy
| | | |
Collapse
|
6
|
Suades R, Greco MF, Padró T, de Santisteban V, Domingo P, Benincasa G, Napoli C, Greco S, Madè A, Ranucci M, Devaux Y, Martelli F, Badimon L. Blood CD45 +/CD3 + lymphocyte-released extracellular vesicles and mortality in hospitalized patients with coronavirus disease 2019. Eur J Clin Invest 2025; 55:e14354. [PMID: 39548690 DOI: 10.1111/eci.14354] [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: 07/14/2024] [Accepted: 11/05/2024] [Indexed: 11/18/2024]
Abstract
BACKGROUND The global pandemic of coronavirus disease 2019 (COVID-19) represented a major public health concern. Growing evidence shows that plasma of COVID-19 patients contains large numbers of circulating extracellular vesicles (cEVs) that correlate with disease severity and recovery. In this study, we sought to characterize the longitudinal cEV signature in critically ill COVID-19 patients during hospitalization and its relation to mortality risk. METHODS cEVs were quantitatively and phenotypically analysed in hospitalized non-surviving COVID-19 patients at baseline (n = 42) and before exitus (n = 40) and in 40 healthy volunteers as a reference group by high sensitivity nano flow cytometry using specific markers for parental cell sources and activation. RESULTS Levels of cEV subtypes differed between patients with severe COVID-19 and healthy subjects, specifically those from platelets and endothelial, inflammatory and viral infected cells, which associate to high mortality risk. In the longitudinal analysis from baseline to the time point immediately preceding death, no changes were found for platelet, pan-leukocyte, and lung epithelial cell-shed cEVs, while endothelial cell releases of EVs (eEVs) significantly differed. Vascular endothelial growth factor receptor 2-positive eEVs were significantly increased before death compared to admission whereas endoglin and E-selectin-containing eEVs did not change. Conversely, lymphocyte (ℓEV), monocyte, macrophage, pericyte and progenitor cell-derived cEVs displayed significant reductions before exitus. Noteworthy, levels of CD45+/CD3+-ℓEVs were significantly associated to the patient's survival time. CONCLUSIONS An evolving cEV profile able to discriminate prompt risk of death during hospitalization has been defined suggesting a role for circulating and vascular cell-derived EVs in COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Rosa Suades
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Maria F Greco
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, Milan, Italy
| | - Teresa Padró
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos III (ISCIII), Madrid, Spain
| | | | - Pere Domingo
- Infectious Diseases Unit, Department of Internal Medicine, Hospital de la Santa Creu i Sant Pau-IR SANT PAU, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Giuditta Benincasa
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Claudio Napoli
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, Italy
- Department of Internal Medicine and Specialistics, Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology (SIMT), Azienda Universitaria Policlinico (AOU), Naples, Italy
| | - Simona Greco
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Milan, Italy
| | - Alisia Madè
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Milan, Italy
| | - Marco Ranucci
- Department of Cardiovascular Anesthesia and Intensive Care, IRCCS Policlinico San Donato, Milan, Italy
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Milan, Italy
| | - Lina Badimon
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos III (ISCIII), Madrid, Spain
- Cardiovascular Research Chair, UAB, Barcelona, Spain
| |
Collapse
|
7
|
Ashique S, Mishra N, Garg A, Garg S, Farid A, Rai S, Gupta G, Dua K, Paudel KR, Taghizadeh-Hesary F. A Critical Review on the Long-Term COVID-19 Impacts on Patients With Diabetes. Am J Med 2025; 138:308-329. [PMID: 38485111 DOI: 10.1016/j.amjmed.2024.02.029] [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: 11/12/2023] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 04/30/2024]
Abstract
BACKGROUND The world is currently grappling with the potentially life-threatening coronavirus disease 2019 (COVID-19), marking it as the most severe health crisis in the modern era. COVID-19 has led to a pandemic, with the World Health Organization (WHO) predicting that individuals with diabetes are at a higher risk of contracting the virus compared to the general population. This review aims to provide a practical summary of the long-term impacts of COVID-19 on patients with diabetes. Specifically, it focuses on the effects of SARS-CoV-2 on different types of diabetic patients, the associated mortality rate, the underlying mechanisms, related complications, and the role of vitamin D and zinc in therapeutic and preventive approaches. METHODS Relevant literature was identified through searches on PubMed, Web of Science, and Science Direct in English, up to April 2023. RESULTS COVID-19 can lead to distressing symptoms and pose a significant challenge for individuals living with diabetes. Older individuals and those with pre-existing conditions such as diabetes, coronary illness, and asthma are more susceptible to COVID-19 infection. Managing COVID-19 in individuals with diabetes presents challenges, as it not only complicates the fight against the infection but also potentially prolongs the recovery time. Moreover, the virus may thrive in individuals with high blood glucose levels. Various therapeutic approaches, including antidiabetic drugs, are available to help prevent COVID-19 in diabetic patients. CONCLUSIONS Diabetes increases the morbidity and mortality risk for patients with COVID-19. Efforts are globally underway to explore therapeutic interventions aimed at reducing the impact of diabetes on COVID-19.
Collapse
Affiliation(s)
- Sumel Ashique
- Department of Pharmaceutical Sciences, Bengal College of Pharmaceutical Sciences & Research, Durgapur, West Bengal, India
| | - Neeraj Mishra
- Amity Institute of Pharmacy, Amity University Madhya Pradesh (AUMP), Gwalior, Madhya Pradesh, India
| | - Ashish Garg
- Drug Delivery and Nanotechnology Laboratories, Department of Pharmaceutics, Guru Ramdas Khalsa Institute of Science and Technology (Pharmacy), Kukrikheda, Barela, Jabalpur, Madhya Pradesh, India
| | - Sweta Garg
- Guru Ramdas Khalsa Institute of Science and Technology, Pharmacy, Jabalpur, Madhya Pradesh, India
| | - Arshad Farid
- Gomal Center of Biochemistry and Biotechnology, Gomal University, Dera Ismail Khan, Pakistan
| | - Shweta Rai
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Gyan Vihar Marg, Jagatpura, Jaipur, Rajasthan 302017, India
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW, Australia
| | - Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, Australia
| | - Farzad Taghizadeh-Hesary
- ENT and Head and Neck Research Center, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Pivina L, Batenova G, Omarov N, Ygiyeva D, Messova A, Alibayeva G, Jamedinova U, Kurumbayev R, Pivin M. Peculiarities of in-Stent Thrombosis and Restenosis in Coronary Arteries Post-COVID-19: A Systematic Review of Clinical Cases and Case Series. Open Access Emerg Med 2025; 17:15-30. [PMID: 39872756 PMCID: PMC11769847 DOI: 10.2147/oaem.s470523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 11/20/2024] [Indexed: 01/30/2025] Open
Abstract
Background One of the most serious complications of coronary artery stenting is restenosis and in-stent thrombosis; their prevalence can reach 20-25%. Stent thrombosis can be acute (up to 24 hours), subacute (24 hours to 30 days), late (30 days to 1 year), and very late (> 1 year after previous stenting). In the patients with COVID-19 in intensive care units, the proportion of those with elevated troponin levels reached 25%. Objective Evaluation of the association between COVID-19 and the development of in-stent thrombosis and restenosis of the coronary arteries based on the analysis of clinical cases and case series. Materials and Methods We searched the PubMed and Scopus databases for relevant case reports and case series of stent restenosis and in-stent thrombosis associated with coronavirus infection (CVI) published between 2020 and the present. Thirty-eight full-text publications were screened and manually checked for analysis. We found 10 publications describing cases of thrombosis and restenosis of stents associated with coronavirus infection, of which only 2 were case series. In total, we analyzed 22 cases. Results In the structure of in-stent restenosis and thrombosis, 59.1% were very late, 9.1% were late; 18.2% were considered subacute events, and 13.6% were acute events. All cases were angiographically confirmed. The main location of restenosis or thrombosis was the left coronary artery (LAD) (51.1%), thrombosis of the right coronary artery (RCA) occurred in 27.3%, and location in circumflex artery was in 22.7%. All patients had COVID-19 confirmed by a PCR test or the presence of immunoglobulins G and M. In fourteen patients (54.5%), an X-ray examination showed the presence of bilateral polysegmental infiltration. Conclusion Analysis of publications demonstrates the association between restenosis and in-stent thrombosis in patients with coronary arteries disease (CAD) and coronavirus infection.
Collapse
Affiliation(s)
- Lyudmila Pivina
- Department of Emergency Medicine, Semey Medical University, Semey, Abay Region, Kazakhstan
| | - Gulnara Batenova
- Department of Emergency Medicine, Semey Medical University, Semey, Abay Region, Kazakhstan
| | - Nazarbek Omarov
- Department of Emergency Medicine, Semey Medical University, Semey, Abay Region, Kazakhstan
| | - Diana Ygiyeva
- Department of Emergency Medicine, Semey Medical University, Semey, Abay Region, Kazakhstan
| | - Assylzhan Messova
- Department of Emergency Medicine, Semey Medical University, Semey, Abay Region, Kazakhstan
| | | | - Ulzhan Jamedinova
- Department of Emergency Medicine, Semey Medical University, Semey, Abay Region, Kazakhstan
| | - Ruslan Kurumbayev
- Department of Emergency Medicine, Semey Medical University, Semey, Abay Region, Kazakhstan
| | - Maksim Pivin
- Nuclear Medicine Department, Center of Nuclear Medicine and Oncology, Semey, Abay Region, Kazakhstan
| |
Collapse
|
9
|
Cambon A, Guervilly C, Delteil C, Potere N, Bachelier R, Tellier E, Abdili E, Leprince M, Giani M, Polidoro I, Albanese V, Ferrante P, Coffin L, Schiffrin M, Arnaud L, Lacroix R, Roque S, Forel JM, Hraiech S, Daniel L, Papazian L, Dignat-George F, Kaplanski G. Caspase-1 activation, IL-1/IL-6 signature and IFNγ-induced chemokines in lungs of COVID-19 patients. Front Immunol 2025; 15:1493306. [PMID: 39882243 PMCID: PMC11774885 DOI: 10.3389/fimmu.2024.1493306] [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: 09/08/2024] [Accepted: 12/11/2024] [Indexed: 01/31/2025] Open
Abstract
Rationale COVID-19-associated acute-respiratory distress syndrome (C-ARDS) results from a direct viral injury associated with host excessive innate immune response mainly affecting the lungs. However, cytokine profile in the lung compartment of C-ARDS patients has not been widely studied, nor compared to non-COVID related ARDS (NC-ARDS). Objectives To evaluate caspase-1 activation, IL-1 signature, and other inflammatory cytokine pathways associated with tissue damage using post-mortem lung tissues, bronchoalveolar lavage fluids (BALF), and serum across the spectrum of COVID-19 severity. Methods Histological features were described and activated-caspase-1 labeling was performed in 40 post-mortem biopsies. Inflammatory cytokines were quantified in BALF and serum from 19 steroid-treated-C-ARDSand compared to 19 NC-ARDS. Cytokine concentrations were also measured in serum from 128 COVID-19 patients at different severity stages. Measurements and main results Typical "diffuse alveolar damage" in lung biopsies were associated with activated caspase-1 expression and vascular lesions. Soluble Caspase-1p20, IL-1β, IL-1Ra, IL-6 and at lower level IFNγ and CXCL-10, were highly elevated in BALF from steroid-treated-C-ARDS as well as in NC-ARDS. IL-1β appeared concentrated in BALF, whereas circulating IL-6 and IL-1Ra concentrations were comparable to those in BALF and correlated with severity. TNFα, TNFR1 and CXCL8 however, were significantly higher in NC-ARDS compared to C-ARDS, treated by steroid. Conclusions In the lungs of C-ARDS, both caspase-1 activation with a predominant IL-1β/IL-6 signature and IFNγ -associated chemokines are elevated despite steroid treatment. These pathways may be specifically targeted in ARDS to improve response to treatment and to limit alveolar and vascular lung damage.
Collapse
Affiliation(s)
- Audrey Cambon
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
| | - Christophe Guervilly
- Centre d’Etudes et de Recherches sur les Services de Santé et qualité de vie EA 3279, Aix-Marseille Université, Marseille, France
- Service de Médecine Intensive Réanimation, Hôpital Nord, Assistance Publique- Hôpitaux de Marseille, Chemin des Bourrely, Marseille, France
| | - Clémence Delteil
- Département de Médecine légale, Hôpital de la Timone, Assistance Publique-Hôpitaux de Marseille, Marseille University, Marseille, France
| | - Nicola Potere
- School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | | | - Edwige Tellier
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
| | - Evelyne Abdili
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
- Service d’Hématologie et de Biologie vasculaire, CHU La Timone, APHM, Marseille, France
| | - Marine Leprince
- Service de Médecine interne et d’Immunologie clinique, Assistance Publique - Hôpitaux de Marseille, Hôpital La Conception, Marseille, France
| | - Marco Giani
- School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Ildo Polidoro
- Unit of Legal Medicine, “Santo Spirito” Hospital, Local Health Authority of Pescara, Pescara, Italy
| | - Valentina Albanese
- Unit of Legal Medicine, “Santo Spirito” Hospital, Local Health Authority of Pescara, Pescara, Italy
| | - Paolo Ferrante
- Unit of Legal Medicine, “Santo Spirito” Hospital, Local Health Authority of Pescara, Pescara, Italy
| | | | | | - Laurent Arnaud
- Service d’Hématologie et de Biologie vasculaire, CHU La Timone, APHM, Marseille, France
| | - Romaric Lacroix
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
- Service d’Hématologie et de Biologie vasculaire, CHU La Timone, APHM, Marseille, France
| | - Sandrine Roque
- Service de Médecine interne et d’Immunologie clinique, Assistance Publique - Hôpitaux de Marseille, Hôpital La Conception, Marseille, France
| | - Jean-Marie Forel
- Centre d’Etudes et de Recherches sur les Services de Santé et qualité de vie EA 3279, Aix-Marseille Université, Marseille, France
- Service de Médecine Intensive Réanimation, Hôpital Nord, Assistance Publique- Hôpitaux de Marseille, Chemin des Bourrely, Marseille, France
| | - Sami Hraiech
- Centre d’Etudes et de Recherches sur les Services de Santé et qualité de vie EA 3279, Aix-Marseille Université, Marseille, France
- Service de Médecine Intensive Réanimation, Hôpital Nord, Assistance Publique- Hôpitaux de Marseille, Chemin des Bourrely, Marseille, France
| | - Laurent Daniel
- Service d’Anatomopathologie, APHM, Aix Marseille University, Marseille, France
| | - Laurent Papazian
- Service de Réanimation, Centre Hospitalier de Bastia, Bastia, France
| | - Françoise Dignat-George
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
- Service d’Hématologie et de Biologie vasculaire, CHU La Timone, APHM, Marseille, France
| | - Gilles Kaplanski
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
- Service de Médecine interne et d’Immunologie clinique, Assistance Publique - Hôpitaux de Marseille, Hôpital La Conception, Marseille, France
| |
Collapse
|
10
|
Ma L, Cai L, Pan J, Cheng Z, Lv Y, Zheng J, Xu P, Zhang H, Chen X, Huang Y, Luo X, Zhao J, Xu L. The immunopathology of coronary microembolization and the underlying inflammopathophysiological mechanisms. Allergol Immunopathol (Madr) 2024; 52:137-146. [PMID: 39515808 DOI: 10.15586/aei.v52i6.1170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 08/22/2024] [Indexed: 11/16/2024]
Abstract
In coronary microembolization, inflammatory cell infiltration, patchy necrosis, and extensive intra-myocardial hemorrhage are dominant, which induce myocardial dysfunction with clinical symptoms of chronic ischemic cardiomyopathy. Microembolization can lead to obstruction of the coronary microvessels and result in the micro-infarction of the heart. The inflammation and elevated expression of the tumor necrosis factor in cardiomyocytes and the activation of extracellular ERK are involved in initiating the inflammatory response mechanism. The PI3K/Akt signaling pathway is the enriched pathway, and for controlling, inhibition of PI3K/Akt is necessary. Furthermore, the release of cytokines and the activation of inflammasomes contribute to the enhancement of vascular permeability, which results in edema within the myocardium. The immune response and inflammation represent the primary triggers in this process. The ability to control immune response and inflammation reactions may lead to the development of new therapies for microembolization.
Collapse
Affiliation(s)
- Li Ma
- Department of Cardiovascular Medicine, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Liping Cai
- Health Management Center, Wuhan Third Hospital, Wuhan, China
| | - Jiayue Pan
- Xiangtao College of Medicine, Xiangtao College Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Zimin Cheng
- Department of Cardiovascular Medicine, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Yuanyuan Lv
- Department of Cardiovascular Medicine, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Jie Zheng
- Department of Cardiovascular Medicine, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Peicheng Xu
- Department of Cardiovascular Medicine, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Hong Zhang
- Department of Cardiovascular Medicine, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Xinyu Chen
- Department of Cardiovascular Medicine, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Yimeng Huang
- Department of Cardiovascular Medicine, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Xiaolei Luo
- Department of Cardiovascular Medicine, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Jinhe Zhao
- Department of Cardiovascular Medicine, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China;
| | - Liang Xu
- Department of ICU, Wuhan Wuchang Hospital, Wuhan, China;
| |
Collapse
|
11
|
Salas JR, Jacob C, Ibekwe E, Zakeri AS, Nimjee SM, Strohm T. ROTEM and von Willebrand Factor in COVID patients presenting with acute ischemic stroke: A case series: ROTEM and von Willebrand Factor in COVID-19 Related Stroke. J Stroke Cerebrovasc Dis 2024; 33:107894. [PMID: 39106921 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/09/2024] Open
Abstract
OBJECTIVES SARS-CoV-2 (COVID) induces systemic thrombotic complications including acute ischemic stroke. In this case series, we report markers of inflammation, coagulation factors including von Willebrand factor antigen, and rotational thromboelastometry (ROTEM) data. MATERIALS AND METHODS Retrospective case series of COVID patients seen at a single comprehensive stroke center between 2020-2022. For patients undergoing mechanical thrombectomy (MT), ROTEM data was collected during the procedure and analyzed on ROTEM delta system. RESULTS Fifteen patients (33.3% female) median age 65-years-old presented with COVID and acute ischemic stroke. Thirteen had LVO. The mean NIHSS was 15 (range 0-35) on admission and 18 (0-42) at discharge. Most were cryptogenic (N=7, 46.7%), followed by cardioembolic (N=6, 40%) and large artery-to-artery embolization (N=2, 13.3%). mRS was < 3 in 8 (53%) patients at discharge. None of the patients were on anticoagulation, and five were on antiplatelet therapy pre-hospitalization. Seven received thrombolytics with alteplase (tPA), and 10 had MT. Baseline platelet count was 102 K/uL (range 102-291 K/uL). vWF was measured in 12 patients, all elevated, with seven having levels >400 (180%). ROTEM data was collected in six patients. Three who received tPA had abnormal EXTEM and FIBTEM data (CT extem > 85secs, A10 EXTEM < 45mm, and A10 FIBTEM < 10mm). Notably, INTEM (CT INTEM >208secs) was abnormal in five of the six patients, two of whom did not receive tPA. CONCLUSIONS Elevated vWF antigen levels with abnormal ROTEM data suggests that COVID induces changes in the clotting cascade. More robust research is needed to investigate these findings. Thrombolytics, MT, and antiplatelet agents should be utilized to treat COVID-related ischemic stroke based on current clinical guidelines.
Collapse
Affiliation(s)
- Jesus R Salas
- Department of Neurology, The Ohio State University, Columbus, OH, USA
| | - Connor Jacob
- The Ohio State University College of Medicine, The Ohio State University, Columbus, OH, USA
| | | | - Amanda S Zakeri
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | - Shahid M Nimjee
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | - Tamara Strohm
- Department of Neurology, Division of Neurocritical Care, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
12
|
Babkina AS, Pisarev MV, Grechko AV, Golubev AM. Arterial Thrombosis in Acute Respiratory Infections: An Underestimated but Clinically Relevant Problem. J Clin Med 2024; 13:6007. [PMID: 39408067 PMCID: PMC11477565 DOI: 10.3390/jcm13196007] [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: 09/02/2024] [Revised: 10/07/2024] [Accepted: 10/07/2024] [Indexed: 10/20/2024] Open
Abstract
During the COVID-19 pandemic, there was increased interest in the issue of thrombotic complications of acute respiratory infections. Clinical reports and pathological studies have revealed that thrombus formation in COVID-19 may involve the venous and arterial vasculature. As thrombotic complications of infectious respiratory diseases are increasingly considered in the context of COVID-19, the fact that thrombosis in lung diseases of viral and bacterial etiology was described long before the pandemic is overlooked. Pre-pandemic studies show that bacterial and viral respiratory infections are associated with an increased risk of thrombotic complications such as myocardial infarction, ischemic stroke, pulmonary embolism, and other critical illnesses caused by arterial and venous thrombosis. This narrative review article aims to summarize the current evidence regarding thrombotic complications and their pathogenesis in acute lower respiratory tract infections.
Collapse
Affiliation(s)
- Anastasiya S. Babkina
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia; (M.V.P.); (A.V.G.); (A.M.G.)
| | | | | | | |
Collapse
|
13
|
Iqbal K, Banga A, Arif TB, Rathore SS, Bhurwal A, Naqvi SKB, Mehdi M, Kumar P, Salklan MM, Iqbal A, Ahmed J, Sharma N, Lal A, Kashyap R, Bansal V, Domecq JP. Anticoagulant use before COVID-19 diagnosis prevent COVID-19 associated acute venous thromboembolism or not: A systematic review and meta-analysis. World J Methodol 2024; 14:92983. [PMID: 39310244 PMCID: PMC11230074 DOI: 10.5662/wjm.v14.i3.92983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/24/2024] [Accepted: 05/11/2024] [Indexed: 06/25/2024] Open
Abstract
BACKGROUND Coagulopathy and thromboembolic events are associated with poor outcomes in coronavirus disease 2019 (COVID-19) patients. There is conflicting evidence on the effects of chronic anticoagulation on mortality and severity of COVID-19 disease. AIM To summarize the body of evidence on the effects of pre-hospital anticoagulation on outcomes in COVID-19 patients. METHODS A Literature search was performed on LitCovid PubMed, WHO, and Scopus databases from inception (December 2019) till June 2023 for original studies reporting an association between prior use of anticoagulants and patient outcomes in adults with COVID-19. The primary outcome was the risk of thromboembolic events in COVID-19 patients taking anticoagulants. Secondary outcomes included COVID-19 disease severity, in terms of intensive care unit admission or invasive mechanical ventilation/intubation requirement in patients hospitalized with COVID-19 infection, and mortality. The random effects models were used to calculate crude and adjusted odds ratios (aORs) with 95% confidence intervals (95%CIs). RESULTS Forty-six observational studies met our inclusion criteria. The unadjusted analysis found no association between prior anticoagulation and thromboembolic event risk [n = 43851, 9 studies, odds ratio (OR)= 0.67 (0.22, 2.07); P = 0.49; I 2 = 95%]. The association between prior anticoagulation and disease severity was non-significant [n = 186782; 22 studies, OR = 1.08 (0.78, 1.49); P = 0.64; I 2 = 89%]. However, pre-hospital anticoagulation significantly increased all-cause mortality risk [n = 207292; 35 studies, OR = 1.72 (1.37, 2.17); P < 0.00001; I 2 = 93%]. Pooling adjusted estimates revealed a statistically non-significant association between pre-hospital anticoagulation and thromboembolic event risk [aOR = 0.87 (0.42, 1.80); P = 0.71], mortality [aOR = 0.94 (0.84, 1.05); P = 0.31], and disease severity [aOR = 0.96 (0.72, 1.26); P = 0.76]. CONCLUSION Prehospital anticoagulation was not significantly associated with reduced risk of thromboembolic events, improved survival, and lower disease severity in COVID-19 patients.
Collapse
Affiliation(s)
- Kinza Iqbal
- Department of Internal Medicine, Dow Medical College, Karachi 74200, Pakistan
| | - Akshat Banga
- Department of Internal Medicine, Sawai Man Singh Medical College, Jaipur 302004, India
| | - Taha Bin Arif
- Department of Internal Medicine, Dow Medical College, Karachi 74200, Pakistan
| | - Sawai Singh Rathore
- Department of Internal Medicine, Dr. Sampurnanand Medical College, Jodhpur 342003, Rajasthan, India
| | - Abhishek Bhurwal
- Department of Gastroenterology and Hepatology, Rutgers Robert Wood Johnson School of Medicine, New Brunswick, NJ 08901, United States
| | | | - Muhammad Mehdi
- Department of Internal Medicine, Dow Medical College, Karachi 74200, Pakistan
| | - Pankaj Kumar
- Department of Internal Medicine, Dow Medical College, Karachi 74200, Pakistan
| | - Mitali Madhu Salklan
- Department of Internal Medicine, Pandit Bhagwat Dayal Sharma Post Graduate Institute of Medical Sciences, Rohtak 124001, Haryana, India
| | - Ayman Iqbal
- Department of Internal Medicine, Dow Medical College, Karachi 74200, Pakistan
| | - Jawad Ahmed
- Department of Internal Medicine, Dow Medical College, Karachi 74200, Pakistan
| | - Nikhil Sharma
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, United States
| | - Amos Lal
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN 55905, United States
| | - Rahul Kashyap
- Department of Research, Wellspan Health, York, PA 17403, United States
| | - Vikas Bansal
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, United States
| | - Juan Pablo Domecq
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, United States
| |
Collapse
|
14
|
Karim ZA, Reese RA, Smith AN, Blackadar ME, Arora V, Moore NM, Johnson EA. Positive impact of nutrition in the prevention of peripheral vascular disease and severe acute respiratory syndrome coronavirus 2: review. Front Nutr 2024; 11:1418028. [PMID: 39364158 PMCID: PMC11448360 DOI: 10.3389/fnut.2024.1418028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 08/27/2024] [Indexed: 10/05/2024] Open
Abstract
Recent research has shown that there is a link between the trend of cardiovascular disease (CVD), chronic symptoms of SARS-CoV-2 (COVID-19), and medical nutrition therapy. Making positive changes to an individual's lifestyle can help to reduce the symptoms that follow exposure to CVD and COVID-19. Sustainable nutrition and lifestyle changes can positively impact an individual's health. Studies have considered the risk factors associated with the disease, medical history, the link between nutrition and peripheral vascular disease (PVD), symptom management, and the interrelationship between nutrition, COVID-19, and PVD. One study has demonstrated that Western Dietary intake can boost the innate immune system while suppressing humoral response, causing chronic inflammation and poor host defense against viruses. However, further investigation is needed to confirm. Patients with PVD and COVID-19 have experienced a reduction in side effects when prescribed a regimen of medical nutrition therapy, heart-healthy diets, and adequate physical activity before and after symptoms of both diseases appear. This approach has proven to be a protective factor during the combination of both illnesses. Our findings indicate that balanced diet and lifestyle are essential in supporting an optimal immune system that can reduce the risk of virus load in individuals at risk of infection and symptoms from COVID-19 and PVD.
Collapse
Affiliation(s)
- Zubair A Karim
- Department of Nutrition and Dietetics, College of Allied Health Sciences, Augusta University, Augusta, GA, United States
| | - Rebecca A Reese
- Department of Nutrition and Dietetics, College of Allied Health Sciences, Augusta University, Augusta, GA, United States
| | - Adrianne N Smith
- Department of Nutrition and Dietetics, College of Allied Health Sciences, Augusta University, Augusta, GA, United States
| | - Madeline E Blackadar
- Department of Nutrition and Dietetics, College of Allied Health Sciences, Augusta University, Augusta, GA, United States
| | - Vishal Arora
- Department of Medicine: Cardiology, Wellstar MCG Health, Augusta University, Augusta, GA, United States
| | - Nicole M Moore
- Department of Nutrition and Dietetics, College of Allied Health Sciences, Augusta University, Augusta, GA, United States
| | - Emily A Johnson
- Department of Nutrition and Dietetics, College of Allied Health Sciences, Augusta University, Augusta, GA, United States
| |
Collapse
|
15
|
Mahroum N, Habra M, Alrifaai MA, Shoenfeld Y. Antiphospholipid syndrome in the era of COVID-19 - Two sides of a coin. Autoimmun Rev 2024; 23:103543. [PMID: 38604461 DOI: 10.1016/j.autrev.2024.103543] [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: 01/31/2024] [Revised: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
In addition to the respiratory symptoms associated with COVID-19, the disease has consistently been linked to many autoimmune diseases such as systemic lupus erythematous and antiphospholipid syndrome (APS). APS in particular was of paramount significance due to its devastating clinical sequela. In fact, the hypercoagulable state seen in patients with acute COVID-19 and the critical role of anticoagulant treatment in affected individuals shed light on the possible relatedness between APS and COVID-19. Moreover, the role of autoimmunity in the assumed association is not less important especially with the accumulated data available regarding the autoimmunity-triggering effect of SARS-CoV-2 infection. This is furtherly strengthened at the time patients with COVID-19 manifested antiphospholipid antibodies of different types following infection. Additionally, the severe form of the APS spectrum, catastrophic APS (CAPS), was shown to have overlapping characteristics with severe COVID-19 such as cytokine storm and multi-organ failure. Interestingly, COVID vaccine-induced autoimmune phenomena described in the medical literature have pointed to an association with APS. Whether the antiphospholipid antibodies were present or de novo, COVID vaccine-induced vascular thrombosis in certain individuals necessitates further investigations regarding the possible mechanisms involved. In our current paper, we aimed to focus on the associations mentioned, their implications, importance, and consequences.
Collapse
Affiliation(s)
- Naim Mahroum
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey.
| | - Mona Habra
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | | | - Yehuda Shoenfeld
- Zabludowicz Center for autoimmune diseases, Sheba Medical Center, Ramat-Gan, Israel; Reichman University, Herzliya, Israel
| |
Collapse
|
16
|
Nathal E, Villanueva-Castro E, Ortiz-Plata A, Serrano-Rubio A, Tena Suck ML. Brain Arteriovenous Malformation Hemorrhage and Pituitary Adenoma in a COVID-19-Positive Patient. Cureus 2024; 16:e67644. [PMID: 39314610 PMCID: PMC11417440 DOI: 10.7759/cureus.67644] [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] [Accepted: 08/23/2024] [Indexed: 09/25/2024] Open
Abstract
Brain arteriovenous malformations (AVMs) are usually asymptomatic. They can cause intense pain or bleeding or lead to other serious medical problems. We present a rare case of a woman who presented with a severe headache and was brought to the emergency service for an intracerebral hemorrhage due to a ruptured AVM. During the surgery, a sellar mass was identified that was also resected. AVM showed vasculitis, endarteritis, endothelial damage, leukocyte plug, and damage to the vessel wall with fragmentation of the collagen and actin filaments. The sellar mass showed a non-functioning pituitary adenoma with hemorrhagic foci and necrosis as well as a proteinaceous vs. lipid material deposition with minimal vascular changes such as endothelial hyperplasia with minimal vasculitis and hyperplasia of reticular stellate cells, with positive glial fibrillary acidic protein (GFAP), which expressed low expression of luteinizing hormone (LH) and follicle-stimulating hormone (FSH), IL6, IL10, IL17, tumor necrosis factor-alpha (TNFa), HIF1a, factor VIII (FVIII), platelet-derived growth factor (PDGF), vascular endothelial growth factor (VEGF), and VEGF receptor 2 (VEGFR2). The patient's polymerase chain reaction COVID-19 test was positive, and she died three days after the surgery procedure. In our knowledge of COVID-19 brain lesions and in the literature review, this was a rare case of a double pathology associated with COVID-19 infection characterized by rupture of the AVM with hemorrhages and brain infarcts associated with endarteritis, vessel wall injuries, and pituitary apoplexy.
Collapse
Affiliation(s)
- Edgar Nathal
- Department of Neurosurgery, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City, MEX
| | - Eliezer Villanueva-Castro
- Department of Neurosurgery, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City, MEX
| | - Alma Ortiz-Plata
- Laboratory of Experimental Neuropathology, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City, MEX
| | - Alejandro Serrano-Rubio
- Department of Neurosurgery, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City, MEX
| | - Martha Lilia Tena Suck
- Department of Neuropathology, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City, MEX
| |
Collapse
|
17
|
Chatterjee B, Modi N, Desai K, Murugan Y, Trivedi A. Alterations in hematologic, coagulation, and inflammatory markers based on fever status in hospitalized COVID-19 patients: A retrospective study. J Family Med Prim Care 2024; 13:3220-3224. [PMID: 39228600 PMCID: PMC11368334 DOI: 10.4103/jfmpc.jfmpc_226_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 03/15/2024] [Accepted: 04/01/2024] [Indexed: 09/05/2024] Open
Abstract
Background Laboratory markers like lymphopenia, thrombocytopenia, elevated D-dimer, and C-reactive protein (CRP) predict worse outcomes in coronavirus disease 2019 (COVID-19). However, a comprehensive analysis of hematologic and coagulation parameter alterations based on fever status is lacking. Methods This retrospective study analyzed 300 COVID-19 patients hospitalized from March to December 2020. Demographic, clinical, and laboratory data were extracted from electronic medical records. Patients were stratified into fever (n = 200) and no fever (n = 100) groups. Hematologic, coagulation, and inflammatory markers were compared between groups using appropriate statistical tests. Multivariate regression identified independent predictors of fever. Results Fever was associated with leukocytosis, neutrophilia, lymphopenia, thrombocytopenia, elevated CRP, D-dimer, procalcitonin, interleukin-6, neutrophil to lymphocyte ratio (NLR), and ferritin compared to no fever (all P < 0.05). D-dimer (r = 0.42), CRP (r = 0.52), NLR (r = 0.48), and interleukin-6 (r = 0.46) demonstrated the strongest correlation with fever (P < 0.001). High D-dimer >1000 ng/mL (adjusted odds ratio 2.7), CRP >100 mg/L (3.1), lymphopenia <1.0 × 109/L (2.8), NLR >4 (2.9), and thrombocytopenia <150 × 109/L (2.7) were significant independent predictors of fever status (P < 0.005). These parameters had moderate sensitivity (40-60%) and high specificity (74-88%) for discriminating febrile patients with AUC of 0.85. Conclusions Marked alterations in hematologic, coagulation, and inflammatory markers occur in COVID-19 based on fever. Routine laboratory parameters can facilitate diagnosis and risk stratification.
Collapse
Affiliation(s)
- Bijoya Chatterjee
- Department of Biochemistry, Shri MP Shah Government Medical College, Jamnagar, Gujarat, India
| | - Nikunj Modi
- Department of Biochemistry, Shri MP Shah Government Medical College, Jamnagar, Gujarat, India
| | - Khushi Desai
- Department of Internal Medicine, Trinity Health Livonia Hospital, Michigan, USA
| | - Yogesh Murugan
- Department of Community Medicine, Shri MP Shah Government Medical College, Jamnagar, Gujarat, India
| | - Ami Trivedi
- Department of Medicine, Shri MP Shah Government Medical College, Jamnagar, Gujarat, India
| |
Collapse
|
18
|
Negaresh M, Ghobadi H, Hoseininia S, Samadi Takaldani AH, Javanshir N, Iranijam E, Aslani MR. Evaluation of the Efficacy of Therapeutic and Prophylactic Anticoagulation in COVID-19 Patients With Venous Catheter and Its Correlation With Clinical Outcomes. INFECTIOUS DISEASES IN CLINICAL PRACTICE 2024; 32. [DOI: 10.1097/ipc.0000000000001382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
IntroductionThe COVID-19 disease was first detected in December 2019, and since then, various treatments have been used to manage it. One such treatment method is therapeutic plasma exchange. This method involves implanting a venous catheter, which increases the risk of venous thromboembolism (VTE). Other risk factors for VTE include infections like COVID-19, inflammation, or malignancy.Materials and MethodsIn this retrospective study, patients with acute respiratory syndrome caused by COVID-19 who were hospitalized and had venous catheters inserted for therapeutic plasma exchange were enrolled. The prophylactic anticoagulant dose was started for all patients, and after the diagnosis of VTE, it was changed to the therapeutic dose. Patients' information, including demographic data, clinical information, and laboratory findings, was extracted from patients' records and recorded in a checklist designed for each patient.ResultsFrom a total of 168 patients, 26 were diagnosed with VTE (pulmonary embolism in 5 patients and deep vein thrombosis in 21 patients). The prevalence of VTE in COVID-19 patients with the venous catheter was 15.4%. The right femoral vein was the most used route for catheterization and had the highest occurrence of venous thromboses. The patients diagnosed with thrombosis showed a lower mortality rate, higher D-dimer and lactate dehydrogenase levels, and lower platelet counts.ConclusionsThis study showed a higher risk of VTE and subclinical thrombosis in COVID-19 patients with venous catheters. Continuous screening, higher doses of anticoagulants, and early removal of venous catheters are critical in preventing VTE and mortality.
Collapse
Affiliation(s)
- Mohammad Negaresh
- Department of Internal Medicine, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hassan Ghobadi
- Department of Internal Medicine (Pulmonology Division), School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Saeed Hoseininia
- Department of Internal Medicine (Pulmonology Division), School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ali Hossein Samadi Takaldani
- Department of Internal Medicine (Pulmonology Division), School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Nima Javanshir
- Faculty of Medicine, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Effat Iranijam
- Department of Internal Medicine (Hematology Division), School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohammad Reza Aslani
- Lung Diseases Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
19
|
Padín JF, Pérez-Ortiz JM, Redondo-Calvo FJ. Aprotinin (II): Inhalational Administration for the Treatment of COVID-19 and Other Viral Conditions. Int J Mol Sci 2024; 25:7209. [PMID: 39000315 PMCID: PMC11241800 DOI: 10.3390/ijms25137209] [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: 05/27/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
Aprotinin is a broad-spectrum inhibitor of human proteases that has been approved for the treatment of bleeding in single coronary artery bypass surgery because of its potent antifibrinolytic actions. Following the outbreak of the COVID-19 pandemic, there was an urgent need to find new antiviral drugs. Aprotinin is a good candidate for therapeutic repositioning as a broad-spectrum antiviral drug and for treating the symptomatic processes that characterise viral respiratory diseases, including COVID-19. This is due to its strong pharmacological ability to inhibit a plethora of host proteases used by respiratory viruses in their infective mechanisms. The proteases allow the cleavage and conformational change of proteins that make up their viral capsid, and thus enable them to anchor themselves by recognition of their target in the epithelial cell. In addition, the activation of these proteases initiates the inflammatory process that triggers the infection. The attraction of the drug is not only its pharmacodynamic characteristics but also the possibility of administration by the inhalation route, avoiding unwanted systemic effects. This, together with the low cost of treatment (≈2 Euro/dose), makes it a good candidate to reach countries with lower economic means. In this article, we will discuss the pharmacodynamic, pharmacokinetic, and toxicological characteristics of aprotinin administered by the inhalation route; analyse the main advances in our knowledge of this medication; and the future directions that should be taken in research in order to reposition this medication in therapeutics.
Collapse
Affiliation(s)
- Juan-Fernando Padín
- Department of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, 13971 Ciudad Real, Spain
| | - José Manuel Pérez-Ortiz
- Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, 28692 Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, 28015 Madrid, Spain
| | - Francisco Javier Redondo-Calvo
- Department of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, 13971 Ciudad Real, Spain
- Department of Anaesthesiology and Critical Care Medicine, University General Hospital, 13005 Ciudad Real, Spain
- Translational Research Unit, University General Hospital and Research Institute of Castilla-La Mancha (IDISCAM), 13005 Ciudad Real, Spain
| |
Collapse
|
20
|
Li K, Feng KC, Simon M, Fu Y, Galanakis D, Mueller S, Rafailovich MH. Molecular Basis for Surface-Initiated Non-Thrombin-Generated Clot Formation Following Viral Infection. ACS APPLIED MATERIALS & INTERFACES 2024; 16:30703-30714. [PMID: 38848451 DOI: 10.1021/acsami.4c02918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
In this paper, we propose a model that connects two standard inflammatory responses to viral infection, namely, elevation of fibrinogen and the lipid drop shower, to the initiation of non-thrombin-generated clot formation. In order to understand the molecular basis for the formation of non-thrombin-generated clots following viral infection, human epithelial and Madin-Darby Canine Kidney (MDCK, epithelial) cells were infected with H1N1, OC43, and adenovirus, and conditioned media was collected, which was later used to treat human umbilical vein endothelial cells and human lung microvascular endothelial cells. After direct infection or after exposure to conditioned media from infected cells, tissue surfaces of both epithelial and endothelial cells, exposed to 8 mg/mL fibrinogen, were observed to initiate fibrillogenesis in the absence of thrombin. No fibers were observed after direct viral exposure of the endothelium or when the epithelium cells were exposed to SARS-CoV-2 isolated spike proteins. Heating the conditioned media to 60 °C had no effect on fibrillogenesis, indicating that the effect was not enzymatic but rather associated with relatively thermally stable inflammatory factors released soon after viral infection. Spontaneous fibrillogenesis had previously been reported and interpreted as being due to the release of the alpha C domains due to strong interactions of the interior of the fibrinogen molecule in contact with hydrophobic material surfaces rather than cleavage of the fibrinopeptides. Contact angle goniometry and immunohistochemistry were used to demonstrate that the lipids produced within the epithelium and released in the conditioned media, probably after the death of infected epithelial cells, formed a hydrophobic residue responsible for fibrillogenesis. Hence, the standard inflammatory response constitutes the ideal conditions for surface-initiated clot formation.
Collapse
Affiliation(s)
- Kao Li
- School of Biomedicine and Nursing, Shandong Institute of Petroleum and Chemical Technology, Dongying 257061, Shandong, China
- Department of Materials Science and Chemical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| | - Kuan-Che Feng
- Department of Materials Science and Chemical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| | - Marcia Simon
- Department of Oral Biology and Pathology, Stony Brook University Medical Center, Stony Brook, New York 11794, United States
| | - Yuyang Fu
- Dongying Stem Cell Bank Medical Technology Co., Ltd., Dongying 257000, Shandong, China
| | - Dennis Galanakis
- Department of Pathology, Stony Brook University School of Medicine, Stony Brook, New York 11720, United States
| | | | - Miriam H Rafailovich
- Department of Materials Science and Chemical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| |
Collapse
|
21
|
Milentijević M, Katanić N, Joksimović B, Pavlović A, Filimonović J, Anđelković M, Bojović K, Elek Z, Ristić S, Vasiljević M, Stevanović J, Radomirović D, Elez-Burnjaković N, Lalović N, Kulić M, Kulić J, Milić M. The Impact of Cytokines on Coagulation Profile in COVID-19 Patients: Controlled for Socio-Demographic, Clinical, and Laboratory Parameters. Biomedicines 2024; 12:1281. [PMID: 38927488 PMCID: PMC11201770 DOI: 10.3390/biomedicines12061281] [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: 03/21/2024] [Revised: 05/31/2024] [Accepted: 06/02/2024] [Indexed: 06/28/2024] Open
Abstract
Background: Severe coagulation abnormalities are common in patients with COVID-19 infection. We aimed to investigate the relationship between pro-inflammatory cytokines and coagulation parameters concerning socio-demographic, clinical, and laboratory characteristics. Methods: Our study included patients hospitalized during the second wave of COVID-19 in the Republic of Serbia. We collected socio-demographic, clinical, and blood-sample data for all patients. Cytokine levels were measured using flow cytometry. Results: We analyzed data from 113 COVID-19 patients with an average age of 58.15 years, of whom 79 (69.9%) were male. Longer duration of COVID-19 symptoms before hospitalization (B = 69.672; p = 0.002) and use of meropenem (B = 1237.220; p = 0.014) were predictive of higher D-dimer values. Among cytokines, higher IL-5 values significantly predicted higher INR values (B = 0.152; p = 0.040) and longer prothrombin times (B = 0.412; p = 0.043), and higher IL-6 (B = 0.137; p = 0.003) predicted longer prothrombin times. Lower IL-17F concentrations at admission (B = 0.024; p = 0.050) were predictive of higher INR values, and lower IFN-γ values (B = -0.306; p = 0.017) were predictive of higher aPTT values. Conclusions: Our findings indicate a significant correlation between pro-inflammatory cytokines and coagulation-related parameters. Factors such as the patient's level of education, gender, oxygen-therapy use, symptom duration before hospitalization, meropenem use, and serum concentrations of IL-5, IL-6, IL-17F, and IFN-γ were associated with worse coagulation-related parameters.
Collapse
Affiliation(s)
- Milica Milentijević
- Department of Infective Diseases, Faculty of Medicine, University of Pristina Temporarily Settled in Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (M.M.); (N.K.)
- Clinical Hospital Center Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (M.A.); (Z.E.); (D.R.)
| | - Nataša Katanić
- Department of Infective Diseases, Faculty of Medicine, University of Pristina Temporarily Settled in Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (M.M.); (N.K.)
| | - Bojan Joksimović
- Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Republic of Srpska, Bosnia and Herzegovina; (K.B.); (S.R.); (M.V.); (N.E.-B.); (N.L.); (M.K.); (J.K.)
| | - Aleksandar Pavlović
- Department of Surgery, Faculty of Medicine, University of Pristina Temporarily Settled in Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia;
| | - Jelena Filimonović
- Department of Epidemiology, Faculty of Medicine, University of Pristina Temporarily Settled in Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (J.F.); (J.S.)
| | - Milena Anđelković
- Clinical Hospital Center Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (M.A.); (Z.E.); (D.R.)
| | - Ksenija Bojović
- Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Republic of Srpska, Bosnia and Herzegovina; (K.B.); (S.R.); (M.V.); (N.E.-B.); (N.L.); (M.K.); (J.K.)
| | - Zlatan Elek
- Clinical Hospital Center Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (M.A.); (Z.E.); (D.R.)
- Department of Surgery, Faculty of Medicine, University of Pristina Temporarily Settled in Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia;
| | - Siniša Ristić
- Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Republic of Srpska, Bosnia and Herzegovina; (K.B.); (S.R.); (M.V.); (N.E.-B.); (N.L.); (M.K.); (J.K.)
| | - Miloš Vasiljević
- Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Republic of Srpska, Bosnia and Herzegovina; (K.B.); (S.R.); (M.V.); (N.E.-B.); (N.L.); (M.K.); (J.K.)
| | - Jasmina Stevanović
- Department of Epidemiology, Faculty of Medicine, University of Pristina Temporarily Settled in Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (J.F.); (J.S.)
| | - Danica Radomirović
- Clinical Hospital Center Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (M.A.); (Z.E.); (D.R.)
| | - Nikolina Elez-Burnjaković
- Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Republic of Srpska, Bosnia and Herzegovina; (K.B.); (S.R.); (M.V.); (N.E.-B.); (N.L.); (M.K.); (J.K.)
| | - Nenad Lalović
- Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Republic of Srpska, Bosnia and Herzegovina; (K.B.); (S.R.); (M.V.); (N.E.-B.); (N.L.); (M.K.); (J.K.)
| | - Milan Kulić
- Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Republic of Srpska, Bosnia and Herzegovina; (K.B.); (S.R.); (M.V.); (N.E.-B.); (N.L.); (M.K.); (J.K.)
| | - Jovan Kulić
- Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Republic of Srpska, Bosnia and Herzegovina; (K.B.); (S.R.); (M.V.); (N.E.-B.); (N.L.); (M.K.); (J.K.)
| | - Marija Milić
- Department of Epidemiology, Faculty of Medicine, University of Pristina Temporarily Settled in Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (J.F.); (J.S.)
| |
Collapse
|
22
|
Mink S, Drexel H, Leiherer A, Frick M, Reimann P, Saely CH, Fraunberger P. Interplay of inflammatory markers and anti-SARS-CoV-2 antibodies in COVID-19 mortality: A prospective cohort study. Int J Infect Dis 2024; 143:107016. [PMID: 38521446 DOI: 10.1016/j.ijid.2024.107016] [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: 12/18/2023] [Revised: 03/11/2024] [Accepted: 03/18/2024] [Indexed: 03/25/2024] Open
Abstract
OBJECTIVES Despite high global vaccination coverage, it remains unclear how vaccination and anti-SARS-CoV-2 antibodies affect immune responses and inflammation levels in patients with COVID-19. It is further unclear whether the inflammatory response differs depending on antibody levels and whether the combination of antibody and inflammation levels in COVID-19 patients affects mortality rates. METHODS We conducted a prospective multicenter cohort study that included 1031 hospitalized COVID-19 patients from five hospitals. Anti-SARS-CoV-2-spike antibodies, interleukin-6 (IL6), and CRP were measured on hospital admission. The prespecified endpoint was all-cause in-hospital mortality. RESULTS We observed significantly lower levels of CRP (P<0.001) and IL6 (P<0.001) in patients with antibody levels above 1200 BAU/ml. After adjusting for potential confounders, patients with high levels of inflammatory markers (CRP>6 mg/dl or IL6>100 pg/ml) combined with low levels of anti-SARS-CoV-2-spike antibodies (<1200 BAU/ml) were approximately 8 times more likely to die than patients with low inflammatory responses and high antibody levels (CRP: aHR 7.973, 95% CI 2.744-23.169, P<0.001; IL6: aHR 8.973, 95% CI 3.549-22.688, P<0.001). CONCLUSION Hospitalized COVID-19 patients presenting with high inflammatory markers and low antibody levels exhibited the highest mortality risks. Higher antibody levels are associated with lower levels of inflammation in hospitalized COVID-19 patients.
Collapse
Affiliation(s)
- Sylvia Mink
- Central Medical Laboratories, Feldkirch, Austria; Private University in the Principality of Liechtenstein, Triesen, Principality of Liechtenstein.
| | - Heinz Drexel
- Private University in the Principality of Liechtenstein, Triesen, Principality of Liechtenstein; VIVIT Institute, Academic Teaching Hospital Feldkirch, Feldkirch, Austria; Drexel University College of Medicine, Philadelphia, PA, USA
| | - Andreas Leiherer
- Central Medical Laboratories, Feldkirch, Austria; VIVIT Institute, Academic Teaching Hospital Feldkirch, Feldkirch, Austria
| | - Matthias Frick
- Department of Internal Medicine, Academic Teaching Hospital Feldkirch, Feldkirch, Austria
| | - Patrick Reimann
- Private University in the Principality of Liechtenstein, Triesen, Principality of Liechtenstein; Department of Internal Medicine, Academic Teaching Hospital Feldkirch, Feldkirch, Austria
| | - Christoph H Saely
- Private University in the Principality of Liechtenstein, Triesen, Principality of Liechtenstein; VIVIT Institute, Academic Teaching Hospital Feldkirch, Feldkirch, Austria
| | - Peter Fraunberger
- Central Medical Laboratories, Feldkirch, Austria; Private University in the Principality of Liechtenstein, Triesen, Principality of Liechtenstein
| |
Collapse
|
23
|
Sourani A, Vahdat N, Bowers CA, Rezvani M, Foroughi M, Sourani A, Mirza R, Baradaran Mahdavi S. SARS-CoV-2 and spinal cord ischemia: a systematic review on clinical presentations, diagnosis, treatment, and outcomes. Spine J 2024; 24:979-988. [PMID: 38365009 DOI: 10.1016/j.spinee.2024.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/16/2023] [Accepted: 02/06/2024] [Indexed: 02/18/2024]
Abstract
BACKGROUND CONTEXT Spinal cord ischemia is a rare but ominous clinical situation with high levels of disability. There are emerging reports on COVID-19 and spinal cord ischemic events. PURPOSE To investigate the cardinal manifestations of SARS-CoV-2 associated spinal cord ischemia, review treatment paradigms, and follow outcomes. STUDY DESIGN A systematic review. METHODS The current study was conducted under Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA) guidelines. The authors searched PubMed, Scopus, Web of Science, and Google Scholar for studies published up to February 12, 2023, on spinal cord ischemia and SARS-CoV-2 infection. Data on patient demographics, study methods, medical records, interventions, and outcomes were extracted from eligible articles. For each data set, the authors performed pooled estimates examining 3 factors of interest, which were (1) predisposing factors (2) treatment regimens, and (3) neurological rehabilitation outcomes. Neurological status was reported as the American Spinal Injury Association (ASIA) impairment scale reported by data sets. RESULTS Six data sets were identified. The mean age of the study population was 50 years old, with 66.6% male predominance. Sixty-six percent of the patients had severe COVID-19. Five data sets reported preexisting coagulopathy. ASIA A and B were the most prevalent primary neurological status (80%). The mean interval between COVID-19 and the first neurological deficit was 13 days. Anterior spinal artery lesions were the most prevalent ischemic pattern. The most common treatment regimens were heparin and steroid therapy. Physical rehabilitation showed poor functional outcomes. CONCLUSIONS SARS-CoV-2 is associated with spinal cord ischemia through multiple neuropathological mechanisms. Proper coagulation profile control and aggressive rehabilitation may play a promising role in the prevention and recovery of spinal cord infarction in SARS-CoV-2 patients.
Collapse
Affiliation(s)
- Arman Sourani
- Department of Neurosurgery, Isfahan University of Medical Sciences, Isfahan, Iran; Environment Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Noushin Vahdat
- Department of Radiology, University of California San Diego (UCSD), San Diego, CA, USA; Department of Radiology, Veterans Administration Healthcare System, San Diego, CA, USA
| | - Christian A Bowers
- Department of Neurosurgery, University of New Mexico Hospital (UNMH), Albuquerque, NM, USA; Bowers Neurosurgical Frailty and Outcomes Data Science Lab, Albuquerque, NM, 87131, USA
| | - Majid Rezvani
- Department of Neurosurgery, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mina Foroughi
- Student Research Committee, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Armin Sourani
- Student Research Committee, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ryan Mirza
- Department of Radiology, University of California San Diego (UCSD), San Diego, CA, USA
| | - Sadegh Baradaran Mahdavi
- Department of Physical Medicine and Rehabilitation, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
24
|
Ebrahimi R, Nasri F, Kalantari T. Coagulation and Inflammation in COVID-19: Reciprocal Relationship between Inflammatory and Coagulation Markers. Ann Hematol 2024; 103:1819-1831. [PMID: 38349409 DOI: 10.1007/s00277-024-05630-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 01/16/2024] [Indexed: 05/14/2024]
Abstract
The coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), formerly known as 2019-nCoV. Numerous cellular and biochemical issues arise after COVID-19 infection. The severe inflammation that is caused by a number of cytokines appears to be one of the key hallmarks of COVID-19. Additionally, people with severe COVID-19 have coagulopathy and fulminant thrombotic events. We briefly reviewed the COVID-19 disease at the beginning of this paper. The inflammation and coagulation markers and their alterations in COVID-19 illness are briefly discussed in the parts that follow. Next, we talked about NETosis, which is a crucial relationship between coagulation and inflammation. In the end, we mentioned the two-way relationship between inflammation and coagulation, as well as the factors involved in it. We suggest that inflammation and coagulation are integrated systems in COVID-19 that act on each other in such a way that not only inflammation can activate coagulation but also coagulation can activate inflammation.
Collapse
Affiliation(s)
- Rasoul Ebrahimi
- Division of Laboratory Hematology and Blood Banking, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Nasri
- Division of Laboratory Hematology and Blood Banking, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Tahereh Kalantari
- Division of Laboratory Hematology and Blood Banking, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
25
|
Sahin ME, Sahin E, Kirlangic MM, Ak M, Daglıtuncezdi Cam S, Cundubey CR, Col Madendag I, Madendag Y. Fetal Diaphragmatic Excursion Is Decreased in Hospitalized Pregnant Women Infected with COVID-19 during the Second and Third Trimesters. Am J Perinatol 2024; 41:e1384-e1389. [PMID: 36724872 DOI: 10.1055/a-2024-0907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE In the present study, we aimed to evaluate coronavirus disease 2019 (COVID-19) infection effects on fetal diaphragm thickness and diaphragmatic excursion, which together show the quality of diaphragmatic contractions. STUDY DESIGN One hundred and ninety-two pregnant women were included in this prospective case-control study. Patients were divided into four groups according to their COVID-19 infection history in their second or third trimester: hospitalized COVID-19-infected pregnant women group (n = 48), outpatient COVID-19-infected pregnant women group (n = 48), common cold (COVID-19 polymerase chain reaction negative) pregnant women group (n = 48), and noninfected healthy controls (n = 48). The number of patients was determined by power analysis following the pilot study. All participants underwent an ultrasound examination to determine fetal diaphragm parameters at 32 to 37 weeks of gestation. RESULTS Demographic characteristics were similar among the four groups. The gestational age at ultrasound examination and gestational age at delivery were similar among the groups. Neonatal intensive care unit (NICU) admission rate was significantly higher in the hospitalized COVID-19-infected pregnant women group than the other groups. The fetal diaphragm thickness during inspiration and expiration, and fetal costophrenic angles at inspiration and expiration were similar among the groups. Fetal diaphragmatic excursion was significantly decreased in the hospitalized COVID-19-infected pregnant women group compared with the other groups. CONCLUSION Our results indicated that moderate maternal COVID-19 infection decreased fetal diaphragmatic excursion, and ultrasonographic evaluation of fetal diaphragmatic excursion before delivery can provide critical information to predict whether infants will require NICU admission. KEY POINTS · Diaphragm ultrasound as a new technique for characterizing the diaphragm's structure and function.. · Fetal diaphragmatic excursion is decreased in the presence of moderate COVID-19 infection.. · Ultrasonographic evaluation of fetal diaphragmatic excursion provides critical information to predict NICU admission..
Collapse
Affiliation(s)
| | - Erdem Sahin
- Department of Obstetrics and Gynecology, Kayseri City Hospital, Kayseri, Turkey
| | - Mehmet M Kirlangic
- Department of Obstetrics and Gynecology, Kartal Dr. Lutfu Kirdar City Hospital, Istanbul, Turkey
| | - Mehmet Ak
- Department of Obstetrics and Gynecology, Kayseri City Hospital, Kayseri, Turkey
| | | | - Cevat R Cundubey
- Department of Obstetrics and Gynecology, Kayseri City Hospital, Kayseri, Turkey
| | - Ilknur Col Madendag
- Department of Obstetrics and Gynecology, Kayseri City Hospital, Kayseri, Turkey
| | - Yusuf Madendag
- Department of Obstetrics and Gynecology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| |
Collapse
|
26
|
Bruno AM, Allshouse AA, Benson AE, Yost CC, Metz TD, Varner MW, Silver RM, Branch DW. Thrombotic Markers in Pregnant Patients with and without SARS-CoV-2 Infection. Am J Perinatol 2024; 41:e3202-e3209. [PMID: 37967868 PMCID: PMC11427751 DOI: 10.1055/a-2211-5052] [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] [Indexed: 11/17/2023]
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is associated with coagulation abnormalities and increased risk for venous and arterial thrombi. This study aimed to evaluate D-dimer levels and lupus anticoagulant (LAC) positivity in pregnant individuals with and without Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. STUDY DESIGN This was a prospective cohort study of pregnant individuals delivering at a single academic institution from April 2020 to March 2022. Individuals with a positive SARS-CoV-2 result during pregnancy were compared with a convenience sample of those without a positive SARS-CoV-2 result. For individuals with SARS-CoV-2 infection, severity was assessed based on the National Institutes of Health classification system. The primary outcome was D-dimer level measured during delivery admission. The secondary outcomes were LAC positivity and thromboembolic events. Outcomes were compared between individuals with and without a positive SARS-CoV-2 result, and further by disease severity. RESULTS Of 98 participants, 77 (78.6%) were SARS-CoV-2 positive during pregnancy. Among individuals with SARS-CoV-2 infection, severity was asymptomatic in 20 (26.0%), mild in 13 (16.9%), moderate in 4 (5.2%), severe in 38 (49.4%), and critical in 2 (2.6%). The D-dimer concentration at delivery did not significantly differ between those with a SARS-CoV-2 positive result compared with those without (mean 2.03 µg/mL [95% confidence interval {CI} 1.72-2.40] vs. 2.37 µg/mL [95% CI 1.65-3.40]; p = 0.43). Three individuals (4%) with SARS-CoV-2 infection and none (0%) without infection were LAC positive (p = 0.59). There were no clinically apparent thromboses in either group. D-dimer concentrations and LAC positive results did not differ by COVID-19 severity. CONCLUSION Thrombotic markers did not differ in pregnant individuals by SARS-CoV-2 infection; however, high rates of LAC positivity were detected. KEY POINTS · Thrombotic markers did not differ in pregnant individuals by SARS-CoV-2 infection.. · Higher than expected rates of LAC positivity were detected.. · There were no clinically apparent thromboses..
Collapse
Affiliation(s)
- Ann M Bruno
- Department of Obstetrics & Gynecology, University of Utah Health, Salt Lake City, Utah
- Department of Obstetrics & Gynecology, Intermountain Health, Salt Lake City, Utah
| | - Amanda A Allshouse
- Department of Obstetrics & Gynecology, University of Utah Health, Salt Lake City, Utah
| | - Ashley E Benson
- Department of Obstetrics & Gynecology, Oregon Health and Science University, Portland, Oregon
| | - Christian Con Yost
- Department of Obstetrics & Gynecology, University of Utah Health, Salt Lake City, Utah
- Molecular Medicine Program, Molecular Medicine Program, University of Utah, Salt Lake City, Utah
| | - Torri D Metz
- Department of Obstetrics & Gynecology, University of Utah Health, Salt Lake City, Utah
- Department of Obstetrics & Gynecology, Intermountain Health, Salt Lake City, Utah
| | - Michael W Varner
- Department of Obstetrics & Gynecology, University of Utah Health, Salt Lake City, Utah
| | - Robert M Silver
- Department of Obstetrics & Gynecology, University of Utah Health, Salt Lake City, Utah
| | - D Ware Branch
- Department of Obstetrics & Gynecology, University of Utah Health, Salt Lake City, Utah
- Department of Obstetrics & Gynecology, Intermountain Health, Salt Lake City, Utah
| |
Collapse
|
27
|
Riou M, Coste F, Meyer A, Enache I, Talha S, Charloux A, Reboul C, Geny B. Mechanisms of Pulmonary Vasculopathy in Acute and Long-Term COVID-19: A Review. Int J Mol Sci 2024; 25:4941. [PMID: 38732160 PMCID: PMC11084496 DOI: 10.3390/ijms25094941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Despite the end of the pandemic, coronavirus disease 2019 (COVID-19) remains a major public health concern. The first waves of the virus led to a better understanding of its pathogenesis, highlighting the fact that there is a specific pulmonary vascular disorder. Indeed, COVID-19 may predispose patients to thrombotic disease in both venous and arterial circulation, and many cases of severe acute pulmonary embolism have been reported. The demonstrated presence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) within the endothelial cells suggests that direct viral effects, in addition to indirect effects of perivascular inflammation and coagulopathy, may contribute to pulmonary vasculopathy in COVID-19. In this review, we discuss the pathological mechanisms leading to pulmonary vascular damage during acute infection, which appear to be mainly related to thromboembolic events, an impaired coagulation cascade, micro- and macrovascular thrombosis, endotheliitis and hypoxic pulmonary vasoconstriction. As many patients develop post-COVID symptoms, including dyspnea, we also discuss the hypothesis of pulmonary vascular damage and pulmonary hypertension as a sequela of the infection, which may be involved in the pathophysiology of long COVID.
Collapse
Affiliation(s)
- Marianne Riou
- Translational Medicine Federation of Strasbourg (FMTS), University of Strasbourg, CRBS, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg, France; (M.R.); (A.M.); (I.E.); (S.T.); (A.C.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’hôpital, 67091 Strasbourg, France
| | - Florence Coste
- EA4278, Laboratoire de Pharm-Ecologie Cardiovasculaire, UFR Sciences Technologies Santé, Pôle Sport et Recherche, 74 rue Louis Pasteur, 84000 Avignon, France; (F.C.); (C.R.)
| | - Alain Meyer
- Translational Medicine Federation of Strasbourg (FMTS), University of Strasbourg, CRBS, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg, France; (M.R.); (A.M.); (I.E.); (S.T.); (A.C.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’hôpital, 67091 Strasbourg, France
| | - Irina Enache
- Translational Medicine Federation of Strasbourg (FMTS), University of Strasbourg, CRBS, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg, France; (M.R.); (A.M.); (I.E.); (S.T.); (A.C.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’hôpital, 67091 Strasbourg, France
| | - Samy Talha
- Translational Medicine Federation of Strasbourg (FMTS), University of Strasbourg, CRBS, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg, France; (M.R.); (A.M.); (I.E.); (S.T.); (A.C.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’hôpital, 67091 Strasbourg, France
| | - Anne Charloux
- Translational Medicine Federation of Strasbourg (FMTS), University of Strasbourg, CRBS, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg, France; (M.R.); (A.M.); (I.E.); (S.T.); (A.C.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’hôpital, 67091 Strasbourg, France
| | - Cyril Reboul
- EA4278, Laboratoire de Pharm-Ecologie Cardiovasculaire, UFR Sciences Technologies Santé, Pôle Sport et Recherche, 74 rue Louis Pasteur, 84000 Avignon, France; (F.C.); (C.R.)
| | - Bernard Geny
- Translational Medicine Federation of Strasbourg (FMTS), University of Strasbourg, CRBS, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg, France; (M.R.); (A.M.); (I.E.); (S.T.); (A.C.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’hôpital, 67091 Strasbourg, France
| |
Collapse
|
28
|
Jahagirdar P, Vaishnav K, Sarathy NA, Singh H, Kumia K, Banerjee A. Role of C-reactive protein, IL-6, and D-dimers in prediction of severity of coronavirus disease 2019: A pilot study. J Oral Maxillofac Pathol 2024; 28:205-210. [PMID: 39157833 PMCID: PMC11329092 DOI: 10.4103/jomfp.jomfp_28_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/17/2024] [Accepted: 03/29/2024] [Indexed: 08/20/2024] Open
Abstract
Background The global outbreak of coronavirus disease 2019 (COVID-19) presents numerous obstacles for healthcare professionals. The present study aimed to evaluate and compare the role of serum biomarkers like- C-reactive protein (CRP), interleukin-6 (IL-6), and D-dimers in the severity of COVID-19 infection. Methodology A cross-sectional, observational retrospective pilot study was conducted in Udaipur, Rajasthan, wherein data was collected from 250 subjects, out of which, data of 100 subjects were included as per the inclusion criteria. The data was recorded retrospectively among the health professionals via Google Forms in Udaipur, Rajasthan. Results There were 1 (1%), 3 (3%), 31 (31%) and 65 (65%) participants with minor elevation (0.3-1.0), moderate elevation (1-10), marked elevation (10-50) and severe elevation (>50) of CRP respectively. The difference between the groups was statistically highly significant with a significantly higher number of study participants with a severe elevation of CRP levels (χ2 = 107.84, P < 0.001). The results showed that there was a significant difference between the groups with IL6 in 0-7 range while 96 (96%) study participants had >7 IL6, and the difference was statistically highly significant (2 = 84.640, P 0.001). Conclusion In conclusion, the existing body of research indicates a discernible correlation between COVID-19 infection and the fluctuation of biomarker levels. This supplement has the potential to be utilised in clinical practice as a means of informing treatment decisions and determining the necessity of admission to the intensive care unit (ICU).
Collapse
Affiliation(s)
- Pramod Jahagirdar
- Department of Dentistry, Karnavati School of Research, Gandhinagar, Gujarat, India
| | - Kalpesh Vaishnav
- Department of Prosthodontics, Crown, and Bridge, Karnavati School of Dentistry, Gujarat, India
| | - Niharika Abhay Sarathy
- Department of Oral and Maxillofacial Pathology, R.R. Dental College and Hospital, Udaipur, Rajasthan, India
| | - Harneet Singh
- Department of Oral Medicine and Radiology, Post Graduate Institute of Dental Sciences, Rohtak, Haryana, India
| | - Komal Kumia
- Department of Oral Medicine and Radiology, Post Graduate Institute of Dental Sciences, Rohtak, Haryana, India
| | - Abhishek Banerjee
- Department of Oral and Maxillofacial Pathology, Awadh Dental College and Hospital, Jamshedpur, Jharkhand, India
| |
Collapse
|
29
|
Shweikeh F, Hong G, Rabeeah S, Shabir U, Sofi A. COVID-19 and Its Sequelae Masquerading as Gastrointestinal Ailments: A Report of Gastroesophageal Reflux Disease (GERD) and Review of Recent Cases. Cureus 2024; 16:e57617. [PMID: 38707018 PMCID: PMC11069409 DOI: 10.7759/cureus.57617] [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] [Accepted: 04/01/2024] [Indexed: 05/07/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) predominantly causes respiratory symptoms. However, a rare segment of patients recovering from COVID-19 may develop gastrointestinal (GI) symptoms. We describe a case of a female who presented with symptoms suggestive of refractory gastroesophageal reflux disease (GERD) for 18 months following COVID-19 infection. Her symptoms included epigastric and chest pain, coughing, and vomiting. Upper endoscopy and 24-hour pH monitoring were negative. Following hospital admission due to worsening symptoms, she was diagnosed with chronic pulmonary embolism (PE) presumed to be related to COVID-19. Her reflux symptoms resolved within two days of the initiation of anticoagulation. Our findings suggest that chronic PE should be considered in patients presenting with GERD refractory to treatment following COVID-19 infection. Generally, as COVID-19 and its sequelae may masquerade as GI conditions, they should be on the differential diagnosis, especially in the post-pandemic era when routine testing has significantly declined.
Collapse
Affiliation(s)
- Faris Shweikeh
- Internal Medicine, Cleveland Clinic Akron General, Akron, USA
| | - Gordon Hong
- Internal Medicine, University Hospitals Cleveland Medical Center, Cleveland, USA
| | - Sana Rabeeah
- Internal Medicine, The University of Toledo Medical Center, Toledo, USA
| | - Usma Shabir
- Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, USA
| | - Aijaz Sofi
- Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, USA
| |
Collapse
|
30
|
Ikiz F, Ak A. Investigation of the relationship between coagulation parameters and mortality in COVID-19 infection. BLOOD SCIENCE 2024; 6:e00191. [PMID: 38694496 PMCID: PMC11062700 DOI: 10.1097/bs9.0000000000000191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 04/07/2024] [Indexed: 05/04/2024] Open
Abstract
This study, which included patients over the age of 18 who were diagnosed with coronavirus disease 2019 (COVID-19) in the emergency clinic, aims to determine the relationship between coagulation parameters and mortality. Epidemiologic data such as age, gender, medical history, vital parameters at emergency department admission, clinical findings, coagulation parameters such as d-dimer, prothrombin time (PT), active partial thromboplastin time (aPTT), international normalized ration (INR), fibrinogen, and platelet were evaluated. Patients with positive computerized tomography (CT) findings and positive polymerase chain reaction (PCR) together were included in the study. It was revealed that d-dimer, fibrinogen, INR, and PT values were higher in the elderly group. It was shown that there was a significant relationship between hospitalization days (ward or intensive care unit) and d-dimer levels. It was observed that d-dimer, fibrinogen elevation was significantly associated with prognosis by increasing mortality, and that platelet and aPTT values were also associated with prognosis and were lower in the mortality group. On the other hand, in receiver operating characteristic (ROC) analysis, the sensitivity and specificity data were 80.3%/80.0% for d-dimer, 70.5%/72.2% for fibrinogen, 58.2%/59.4% for aPTT, and 59.7%/59.2% for platelet, respectively. The overall classification success was 88.6% and mortality prediction success was 37.7% in the regression model of some coagulation parameters (d-dimer, fibrinogen, aPTT, and platelet) which were effective on prognosis. In conclusion, it was determined that d-dimer, fibrinogen, aPTT, and platelet parameters were directly associated with mortality and when these coagulation parameters were used together with the clinical, vital, and demographic data of the patients, the success of mortality prediction increased significantly.
Collapse
Affiliation(s)
- Fatih Ikiz
- Department of Emergency Medicine, Beyhekim Training and Research Hospital, Selcuklu, Konya, Turkey
| | - Ahmet Ak
- Department of Emergency Medicine, Faculty of Medicine, Selcuk University, Selcuklu, Konya, Turkey
| |
Collapse
|
31
|
Aggarwal A, Singh TK, Pham M, Godwin M, Chen R, McIntyre TM, Scalise A, Chung MK, Jennings C, Ali M, Park H, Englund K, Khorana AA, Svensson LG, Kapadia S, McCrae KR, Cameron SJ. Dysregulated platelet function in patients with postacute sequelae of COVID-19. Vasc Med 2024; 29:125-134. [PMID: 38334067 PMCID: PMC11164201 DOI: 10.1177/1358863x231224383] [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] [Indexed: 02/10/2024]
Abstract
BACKGROUND Postacute sequelae of COVID-19 (PASC), also referred to as "Long COVID", sometimes follows COVID-19, a disease caused by SARS-CoV-2. Although SARS-CoV-2 is well known to promote a prothrombotic state, less is known about the thrombosis risk in PASC. Our objective was to evaluate platelet function and thrombotic potential in patients following recovery from SARS-CoV-2, but with clear symptoms of patients with PASC. METHODS patients with PASC and matched healthy controls were enrolled in the study on average 15 months after documented SARS-CoV-2 infection. Platelet activation was evaluated by light transmission aggregometry (LTA) and flow cytometry in response to platelet surface receptor agonists. Thrombosis in platelet-deplete plasma was evaluated by Factor Xa activity. A microfluidics system assessed thrombosis in whole blood under shear stress conditions. RESULTS A mild increase in platelet aggregation in patients with PASC through the thromboxane receptor was observed, and platelet activation through the glycoprotein VI (GPVI) receptor was decreased in patients with PASC compared to age- and sex-matched healthy controls. Thrombosis under shear conditions as well as Factor Xa activity were reduced in patients with PASC. Plasma from patients with PASC was an extremely potent activator of washed, healthy platelets - a phenomenon not observed when stimulating healthy platelets after incubation with plasma from healthy individuals. CONCLUSIONS patients with PASC show dysregulated responses in platelets and coagulation in plasma, likely caused by a circulating molecule that promotes thrombosis. A hitherto undescribed protective response appears to exist in patients with PASC to counterbalance ongoing thrombosis that is common to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Anu Aggarwal
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Tamanna K Singh
- Section of Vascular Medicine, Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Michael Pham
- Section of Vascular Medicine, Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Matthew Godwin
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Rui Chen
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Thomas M McIntyre
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Alliefair Scalise
- Section of Vascular Medicine, Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Mina K Chung
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
- Section of Vascular Medicine, Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Courtney Jennings
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Mariya Ali
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Hiijun Park
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Kristin Englund
- Department of Infectious Disease, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Alok A Khorana
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Lars G Svensson
- Department of Cardiac Surgery, Heart Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Samir Kapadia
- Section of Vascular Medicine, Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Keith R McCrae
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
- Section of Vascular Medicine, Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Scott J Cameron
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
- Section of Vascular Medicine, Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
32
|
Chagas LDS, Serfaty CA. The Influence of Microglia on Neuroplasticity and Long-Term Cognitive Sequelae in Long COVID: Impacts on Brain Development and Beyond. Int J Mol Sci 2024; 25:3819. [PMID: 38612629 PMCID: PMC11011312 DOI: 10.3390/ijms25073819] [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: 03/01/2024] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Microglial cells, the immune cells of the central nervous system, are key elements regulating brain development and brain health. These cells are fully responsive to stressors, microenvironmental alterations and are actively involved in the construction of neural circuits in children and the ability to undergo full experience-dependent plasticity in adults. Since neuroinflammation is a known key element in the pathogenesis of COVID-19, one might expect the dysregulation of microglial function to severely impact both functional and structural plasticity, leading to the cognitive sequelae that appear in the pathogenesis of Long COVID. Therefore, understanding this complex scenario is mandatory for establishing the possible molecular mechanisms related to these symptoms. In the present review, we will discuss Long COVID and its association with reduced levels of BDNF, altered crosstalk between circulating immune cells and microglia, increased levels of inflammasomes, cytokines and chemokines, as well as the alterations in signaling pathways that impact neural synaptic remodeling and plasticity, such as fractalkines, the complement system, the expression of SIRPα and CD47 molecules and altered matrix remodeling. Together, these complex mechanisms may help us understand consequences of Long COVID for brain development and its association with altered brain plasticity, impacting learning disabilities, neurodevelopmental disorders, as well as cognitive decline in adults.
Collapse
Affiliation(s)
- Luana da Silva Chagas
- Program of Neuroscience, Department of Neurobiology, Institute of Biology, Federal Fluminense University, Niterói 24210-201, Rio de Janeiro, Brazil;
- National Institute of Science and Technology on Neuroimmunomodulation—INCT-NIM, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21041-250, Rio de Janeiro, Brazil
- Rio de Janeiro Research Network on Neuroinflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21041-250, Rio de Janeiro, Brazil
| | - Claudio Alberto Serfaty
- Program of Neuroscience, Department of Neurobiology, Institute of Biology, Federal Fluminense University, Niterói 24210-201, Rio de Janeiro, Brazil;
- National Institute of Science and Technology on Neuroimmunomodulation—INCT-NIM, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21041-250, Rio de Janeiro, Brazil
- Rio de Janeiro Research Network on Neuroinflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21041-250, Rio de Janeiro, Brazil
| |
Collapse
|
33
|
Youness M, Mansour S, Sakr F, Olabi S, Atwi S, Martinez IY, El Khatib S, Hallit S, Salameh P, Malaeb D, Hosseini H. Odds and associated factors for thrombosis development among Lebanese COVID-19 patients: a case-control retrospective study. J Pharm Policy Pract 2024; 17:2319743. [PMID: 38505825 PMCID: PMC10950289 DOI: 10.1080/20523211.2024.2319743] [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] [Indexed: 03/21/2024] Open
Abstract
Background Thromboembolism is reported to be up to 27% in COVID-19 patients due to SARS-CoV-2 infection. Dysregulated systemic inflammation and various patient traits play a vital role in thrombosis progression. Purpose To assess odds and associated factors for thrombosis development among Lebanese COVID-19 patients. Methods This was a case-control retrospective study conducted in January-May 2021. Patients infected with COVID-19 and developed thrombosis were classified as cases and patients who were thrombosis-free identified as control. A questionnaire assessed socio-demographics, clinical parameters, and WHO COVID-19 disease severity. Results Among 267 patients, 26 (9.7%) developed thrombosis and the majority of thrombosis 34.6% was myocardial infarction, and the least (3.8%) was for catheter-related thrombosis. Results showed that the risk of thrombosis development is higher in patients with previous thromboembolic event (OR = 9.160) and previous intake of anti-hypertensive medications at home (OR = 3.116). However, females (OR = 0.330; CI: 0.118-0.925), intake of anticoagulants during hospital admission (OR = 0.126; CI: 0.053-0.300) and non-severe COVID-19 were at lower thrombosis risk (OR = 0.273). Patients who developed thromboembolic events had longer hospital stay (OR = 0.077). Conclusion Patients with COVID-19 and thromboembolism were at higher risk of mortality as compared to patients with COVID-19 but without thromboembolism. The use of anticoagulants significantly reduced the risk for thromboembolism.
Collapse
Affiliation(s)
- Mahmoud Youness
- Research Department, Beirut Cardiac Institute, Beirut, Lebanon
| | - Sara Mansour
- School of Pharmacy, Lebanese International University, Beirut, Lebanon
| | - Fouad Sakr
- School of Pharmacy, Lebanese International University, Beirut, Lebanon
| | - Samer Olabi
- Rafic Hariri University Hospital, Beirut, Lebanon
| | - Sarah Atwi
- Rafic Hariri University Hospital, Beirut, Lebanon
| | | | - Sami El Khatib
- Department of Biomedical Sciences, Lebanese International University, Bekaa, Lebanon
- Center for Applied Mathematics and Bioinformatics (CAMB), Gulf University for Science and Technology, Mubarak Al-Abdullah, Kuwait
| | - Souheil Hallit
- School of Medicine and Medical Sciences, Holy Spirit University of Kaslik, Jounieh, Lebanon
- Applied Science Research Center, Applied Science Private University, Amman, Jordan
| | - Pascale Salameh
- INSPECT-LB: Institut National de Santé Publique, Epidémiologie Clinique et Toxicologie, Beirut, Lebanon
- Faculty of Pharmacy, Lebanese University, Hadath, Lebanon
- Department of Primary Care and Population Health, University of Nicosia Medical School, Nicosia, Cyprus
- School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Diana Malaeb
- College of Pharmacy, Gulf Medical University, Ajman, United Arab Emirates
| | - Hassan Hosseini
- Neurology Department, Henri Mondor Hospital, AP-HP, Creteil, France
- UPEC-University Paris-Est, Creteil, France
- RAMSAY SANTÉ, HPPE, Champigny sur Marne, France
| |
Collapse
|
34
|
Alsayed AR, Ahmed SI, AL Shweiki AO, Al-Shajlawi M, Hakooz N. The laboratory parameters in predicting the severity and death of COVID-19 patients: Future pandemic readiness strategies. BIOMOLECULES & BIOMEDICINE 2024; 24:238-255. [PMID: 37712883 PMCID: PMC10950347 DOI: 10.17305/bb.2023.9540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/30/2023] [Accepted: 09/14/2023] [Indexed: 09/16/2023]
Abstract
The range of clinical manifestations associated with the infection by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) encompasses a broad spectrum, ranging from flu-like symptoms to the occurrence of multiple organ failure and death. The severity of the coronavirus disease 2019 (COVID-19) is categorized based on clinical presentation and is divided into three distinct levels of severity identified as non-severe, severe, and critical. Although individuals of all age groups are susceptible to SARS-CoV-2 infection, middle-aged and older adults are more frequently impacted, with the latter being more likely to develop severe illness. Various laboratory characteristics observed in hospitalized COVID-19 patients have been correlated with adverse outcomes. These include elevated levels of D-dimer, liver enzymes, lactate dehydrogenase, C-reactive protein, ferritin, prothrombin time, and troponin, as well as decreased lymphocyte and platelets counts. This review investigated the relationship between baseline clinical characteristics, initial laboratory parameters upon hospital admission, and the severity of illness and mortality rates among COVID-19 patients. Although the COVID-19 pandemic has concluded, understanding the laboratory predictors of virus severity and mortality remains crucial, and examining these predictors can have long-term effects. Such insights can help healthcare systems manage resources more effectively and deliver timely and appropriate care by identifying and targeting high-risk individuals. This knowledge can also help us better prepare for future pandemics. By examining these predictors, we can take steps to protect public health and mitigate the impact of future pandemics.
Collapse
Affiliation(s)
- Ahmad R Alsayed
- Department of Clinical Pharmacy and Therapeutics, Faculty of Pharmacy, Applied Science Private University, Amman, Jordan
| | - Syed Imran Ahmed
- College of Health and Science, School of Pharmacy, University of Lincoln, Lincoln, United Kingdom
| | - Anas Osama AL Shweiki
- Department of Clinical Pharmacy and Therapeutics, Faculty of Pharmacy, Applied Science Private University, Amman, Jordan
| | - Mustafa Al-Shajlawi
- Department of Clinical Pharmacy and Therapeutics, Faculty of Pharmacy, Applied Science Private University, Amman, Jordan
| | - Nancy Hakooz
- School of Pharmacy, The University of Jordan, Amman, Jordan
| |
Collapse
|
35
|
Spiezia L, Campello E, Simioni P, Lumbreras-Marquez MI. Whole blood viscoelastic testing profile and mortality in patients hospitalized with acute COVID-19 pneumonia: A systematic review and meta-analysis. Thromb Res 2024; 234:21-31. [PMID: 38142487 DOI: 10.1016/j.thromres.2023.12.009] [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: 10/24/2023] [Revised: 12/16/2023] [Accepted: 12/18/2023] [Indexed: 12/26/2023]
Abstract
BACKGROUND Several studies have evaluated the possible association between whole blood viscoelastic testing (VET) parameters in patients hospitalized for acute Coronavirus disease 2019 (COVID-19) pneumonia and mortality. A few studies found no significant differences between survivors and non-survivors, though other studies identified potential predictors of COVID-19-related mortality. We conducted a systematic review and meta-analysis of the literature to evaluate the possible association between standard thromboelastometry/graphy parameters and mortality in patients hospitalized for acute COVID-19 pneumonia. METHODS Relevant studies were searched through MEDLINE, EMBASE, and Google Scholar from their inception until 15th June 2023. We aimed to identify any study including: i) adults admitted to intensive care units (ICU) or medicine wards (MW) for acute COVID-19 pneumonia; ii) viscoelastic testing; iii) mortality. RESULTS We included 13 studies: nine prospective and four retrospective, 231 (30.4 %) non-survivors and 528 (69.6 %) survivors. Mortality rates ranged from 12.8 % to 67.5 %. The studies using the TEG apparatus found a significant difference in K time in the Kaolin test among survivors vs. non-survivors (mean difference [MD] 0.20, 95 % confidence interval [CI] 0.12, 0.28, I2 0%). The studies using the rotational thromboelastometry apparatus found a significant difference in CT-INTEM (MD -17.14, 95 % CI -29.23, -5.06, I2 0%) and LI60-EXTEM (MD -1.00, 95 % CI -1.00, -1.00, I2 0%) assays among survivors vs. non-survivors. CONCLUSION We identified no specific hypercoagulable or hypocoagulable profile associated with mortality in patients with COVID-19-related pneumonia. Large prospective studies are needed to explore the possible prognostic role of VET in this subset of patients.
Collapse
Affiliation(s)
- Luca Spiezia
- General Internal Medicine and Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine, Padova University School of Medicine, Padova, Italy.
| | - Elena Campello
- General Internal Medicine and Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine, Padova University School of Medicine, Padova, Italy
| | - Paolo Simioni
- General Internal Medicine and Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine, Padova University School of Medicine, Padova, Italy
| | - Mario I Lumbreras-Marquez
- Universidad Panamericana School of Medicine, Mexico City, Mexico; Maternal-Fetal Medicine Division, Instituto Nacional de Perinatologia, Mexico City, Mexico
| |
Collapse
|
36
|
S B MJ, Chacko B, Selvarajan S, Peter JV, Geevar T, Dave RG, Georgy JT, Zachariah A, George T, Sathyendra S, Hansdak SG, Krishnaswami RK, Thangakunam B, Gupta R, Karuppusami R, Nair SC, Srivastava A. Biomarkers of coagulation, endothelial, platelet function, and fibrinolysis in patients with COVID-19: a prospective study. Sci Rep 2024; 14:2011. [PMID: 38263377 PMCID: PMC10805716 DOI: 10.1038/s41598-024-51908-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/11/2024] [Indexed: 01/25/2024] Open
Abstract
Prospective and sequential evaluation of homeostatic changes leading to thrombosis across COVID 19 disease severity spectrum are limited. In this prospective observational study, haemostasis was evaluated in patients with mild, moderate-severe, and critical COVID-19 infection. Markers of endothelial activation [Soluble thrombomodulin (sTM), von Willebrand Factor (VWF)], platelet activation [Soluble P-selectin, beta-thromboglobulin (BTG)] and global haemostasis [Rotational thromboelastometry (ROTEM)] were evaluated on days 1 and 5 after admission. The study cohort comprised of 100 adult patients (mild = 20, moderate-severe = 22, critical = 58). Sixty-five patients received anticoagulation for 10 (7-14) days. Thrombotic events were seen in 9 patients. In-hospital mortality was 21%. Endothelial activation markers were elevated at baseline in all subgroups, with levels in moderate-severe (sTM = 4.92 ng/ml, VWF = 295 U/dl) [reference-ranges: sTM = 2.26-4.55 ng/ml; Soluble P-selectin = 13.5-31.5 ng/ml; BTG = 0.034-1.99 ng/ml] and critical patients (sTM = 6.07 ng/ml, VWF = 294 U/dl) being significantly higher than in the mild group (sTM = 4.18 ng/ml, VWF = 206 U/dl). In contrast, platelet activation markers were elevated only in critically ill patients at baseline (Soluble P-selectin = 37.3 ng/ml, BTG = 2.51 ng/ml). The critical group had significantly lower fibrinolysis on days 1 and 5 when compared with the moderate-severe arm. COVID-19 infection was associated with graded endothelial activation and lower fibrinolysis that correlated with illness severity.
Collapse
Affiliation(s)
- Manoj Job S B
- Department of Critical Care, Christian Medical College, Vellore, 632004, India.
| | - Binila Chacko
- Department of Critical Care, Christian Medical College, Vellore, 632004, India
| | - Sushil Selvarajan
- Department of Clinical Hematology, Christian Medical College, Vellore, India
| | - John Victor Peter
- Department of Critical Care, Christian Medical College, Vellore, 632004, India
| | - Tulasi Geevar
- Department of Transfusion Medicine, Christian Medical College, Vellore, India
| | - Rutvi Gautam Dave
- Department of Transfusion Medicine, Christian Medical College, Vellore, India
| | - Josh Thomas Georgy
- Department of General Medicine, Christian Medical College, Vellore, India
| | - Anand Zachariah
- Department of General Medicine, Christian Medical College, Vellore, India
| | - Tina George
- Department of General Medicine, Christian Medical College, Vellore, India
| | - Sowmya Sathyendra
- Department of General Medicine, Christian Medical College, Vellore, India
| | | | | | | | - Richa Gupta
- Department of Respiratory Medicine, Christian Medical College, Vellore, India
| | - Reka Karuppusami
- Department of Biostatistics, Christian Medical College, Vellore, India
| | | | - Alok Srivastava
- Department of Clinical Hematology, Christian Medical College, Vellore, India
| |
Collapse
|
37
|
Iwamoto T, Hatayama Y, Yamashita N, Ichikawa H, Kawamura K. Association Between Coagulation Fibrinolysis Markers and Severity in Patients with COVID-19. Clin Appl Thromb Hemost 2024; 30:10760296241298233. [PMID: 39529256 PMCID: PMC11555749 DOI: 10.1177/10760296241298233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
INTRODUCTION Thrombosis is a major complication of COVID-19. D-dimer (DD) is an important coagulation fibrinolysis marker in COVID-19 and has been extensively studied. However, very little is known about the role of other fibrinolysis markers, plasmin-plasmin inhibitor complex (PIC), and fibrin monomer complex (FMC) in COVID-19. This study investigated and compared the associations of DD, PIC, and FMC with COVID-19 severity. METHODS Archived plasma samples from patients with COVID-19 (n = 50) were assessed for DD, FMC, and PIC levels. We compared the levels between patients with mild (n = 36) and moderate (n = 14) COVID-19 and evaluated their correlation with other COVID-19 severity markers, including lactate dehydrogenase (LD), serum albumin (Alb), C-reactive protein (CRP), and neutrophil-lymphocyte ratio (NLR). RESULTS Patients with moderate COVID-19 had significantly higher DD and PIC levels than those with mild disease, while FMC levels were comparable. DD and PIC levels significantly correlated with LD, Alb, and NLR. Additionally, PIC levels significantly correlated with CRP levels. However, FMC levels correlated only with Alb but not with LD, NLR, or CRP. CONCLUSIONS COVID-19 severity was significantly associated with PIC and DD levels but not with FMC levels. PIC was the variable with the most clearly significant difference between patients with moderate disease and those with mild disease, highlighting its potential as an indicator of coagulation and fibrinolysis imbalance in patients with COVID-19. Further studies with larger sample sizes and more diverse severity groups are required to confirm these findings.
Collapse
Affiliation(s)
- Takuya Iwamoto
- Division of Clinical Laboratory Medicine, Tottori University Hospital, Yonago, Tottori, Japan
| | - Yuki Hatayama
- Division of Clinical Laboratory Medicine, Tottori University Hospital, Yonago, Tottori, Japan
- Department of Multidisciplinary Internal Medicine, School of Medicine, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Noriko Yamashita
- Division of Clinical Laboratory Medicine, Tottori University Hospital, Yonago, Tottori, Japan
| | - Hitomi Ichikawa
- Division of Clinical Laboratory Medicine, Tottori University Hospital, Yonago, Tottori, Japan
| | - Koji Kawamura
- Division of Clinical Laboratory Medicine, Tottori University Hospital, Yonago, Tottori, Japan
- Department of Hematology, Tottori University Hospital, Yonago, Tottori, Japan
| |
Collapse
|
38
|
Mendes-Filho SPDM, de Souza Pinheiro R, Martins FS, Giroldi PJ, e Melo RH, de Oliveira EL, dos Santos AB, Medeiros DCO, Lopes JA, Chaves YO, Zuliani JP, Nogueira PA. Kinetics of IL-6, C-reactive Protein and Fibrinogen Levels in COVID-19 Outpatients Who Evolved to Hypoxemia. CLINICAL PATHOLOGY (THOUSAND OAKS, VENTURA COUNTY, CALIF.) 2024; 17:2632010X231222795. [PMID: 38188270 PMCID: PMC10768631 DOI: 10.1177/2632010x231222795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 12/08/2023] [Indexed: 01/09/2024]
Abstract
Introduction Despite the efficacy of the COVID-19, the search for improvements in the management of severe/critical cases continues to be important. The aim is to demonstrate the kinetics of 4 serological markers in patients with COVID-19 who evolved in hypoxemia. Methods From June to December 2020, the Health Secretariat of Rondônia State, Brazil, established a home medical care service team (HMCS) that provided clinical follow-up for health professionals and military personnel with COVID-19. The clinical and laboratory monitoring was individualized at home by a nursing and medical team. In addition to laboratory parameters, C-reactive protein (CRP), interleukin-6 (IL-6), fibrinogen, and D-dimer levels were periodically taken to monitor the evolution of treatment. Results Of 218 patients telemonitored, 48 patients needed special care by the HMCS team due to shortness of breath. Chest tomography showed multiple ground-glass shadows and lung parenchymal condensations that was compatible with secondary bacterial infection associated with leukocytosis, for which antibiotics were prescribed. The symptoms were accompanied by increases of CRP and IL-6 levels followed by fibrinogen after a few days, for which an anticoagulant therapy was included. Thirty-three patients evolved to improvements in clinical signs and laboratory results. Between the sixth and eighth day of illness, 15 patients presented signs of hypoxemia with low O2 saturation accompanied with an increase in the respiratory rate, with some of them requiring oxygen therapy. As they did not present signs of clinical severity, but their laboratory markers showed an abrupt IL-6 peak that was higher than the increase in CRP and a new alteration in fibrinogen levels, they received a supplemental dose of anticoagulant and a high dose of corticosteroids, which resulted in clinical improvement. Conclusion Our study demonstrates that monitoring of IL-6 and CRP may identify precocious hypoxemia in COVID-19 patients and prevented the progressive deterioration of the lung injury.
Collapse
Affiliation(s)
| | - Rebeca de Souza Pinheiro
- Programa de Pós-graduação de Imunologia Básica e Aplicada, Universidade Federal do Amazonas, Manaus/AM, Brazil
| | - Fernanda Simão Martins
- Serviço de Assistência Médica Domiciliar (SAMD), Secretaria Estadual da Saúde (SESAU), Porto Velho/RO, Brazil
| | - Paulo Jose Giroldi
- Laboratório Estadual de Patologia e Análises Clínicas (LEPAC), Porto Velho/RO, Brazil
| | - Raul Honorato e Melo
- Serviço de Assistência Médica Domiciliar (SAMD), Secretaria Estadual da Saúde (SESAU), Porto Velho/RO, Brazil
- Hospital Cemetron, Porto Velho/RO, Brazil
| | | | - Anibal Borin dos Santos
- Serviço de Assistência Médica Domiciliar (SAMD), Secretaria Estadual da Saúde (SESAU), Porto Velho/RO, Brazil
| | | | | | - Yury Oliveira Chaves
- Instituto Leônidas e Maria Deane (ILMD), Fundação Oswaldo Cruz – Amazonas (FIOCRUZ – AMAZONAS), Manaus/AM, Brazil
| | | | - Paulo Afonso Nogueira
- Programa de Pós-graduação de Imunologia Básica e Aplicada, Universidade Federal do Amazonas, Manaus/AM, Brazil
- Instituto Leônidas e Maria Deane (ILMD), Fundação Oswaldo Cruz – Amazonas (FIOCRUZ – AMAZONAS), Manaus/AM, Brazil
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus/AM, Brazil
| |
Collapse
|
39
|
Guo J, Zhang Y, Gao Y, Li S, Xu G, Tian Z, Xu Q, Li X, Li Y, Zhang Y. Systematical analyses of large-scale transcriptome reveal viral infection-related genes and disease comorbidities. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2023; 51:453-465. [PMID: 37651591 DOI: 10.1080/21691401.2023.2252477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 08/13/2023] [Accepted: 08/17/2023] [Indexed: 09/02/2023]
Abstract
Perturbation of transcriptome in viral infection patients is a recurrent theme impacting symptoms and mortality, yet a detailed understanding of pertinent transcriptome and identification of robust biomarkers is not complete. In this study, we manually collected 23 datasets related to 6,197 blood transcriptomes across 16 types of respiratory virus infections. We applied a comprehensive systems biology approach starting with whole-blood transcriptomes combined with multilevel bioinformatics analyses to characterize the expression, functional pathways, and protein-protein interaction (PPI) networks to identify robust biomarkers and disease comorbidities. Robust gene markers of infection with different viruses were identified, which can accurately classify the normal and infected patients in train and validation cohorts. The biological processes (BP) of different viruses showed great similarity and enriched in infection and immune response pathways. Network-based analyses revealed that a variety of viral infections were associated with nervous system diseases, neoplasms and metabolic diseases, and significantly correlated with brain tissues. In summary, our manually collected transcriptomes and comprehensive analyses reveal key molecular markers and disease comorbidities in the process of viral infection, which could provide a valuable theoretical basis for the prevention of subsequent public health events for respiratory virus infections.
Collapse
Affiliation(s)
- Jing Guo
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
| | - Ya Zhang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
| | - Yueying Gao
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
| | - Si Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
| | - Gang Xu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
| | - Zhanyu Tian
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
| | - Qi Xu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
| | - Xia Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yongsheng Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
| | - Yunpeng Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
40
|
Siddique YA, Chaudhry R, Ahmad M, Sebai A, Sharma L, Hassouba M, Virk GS. The Trend of Arrhythmias in Patients With COVID-19: A Complication or Late Manifestation? Cureus 2023; 15:e50746. [PMID: 38239526 PMCID: PMC10794791 DOI: 10.7759/cureus.50746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2023] [Indexed: 01/22/2024] Open
Abstract
Patients diagnosed with coronavirus disease (CVD) who experience cardiovascular complications or have pre-existing cardiovascular disease are at an increased risk of death. The primary heart-related consequences associated with COVID-19 encompass venous thromboembolism, shock, heart failure, arrhythmias, myocarditis, acute myocardial infarction, and acute cardiac damage. The coronavirus has the potential to induce cardiovascular complications or exacerbate pre-existing CVD through various mechanisms. These mechanisms include dysregulation of the renin-angiotensin-aldosterone system; direct viral toxicity; damage to endothelial cells; formation of blood clots and subsequent inflammation, a phenomenon known as thromboinflammation; an excessive immune response known as cytokine storm; and an imbalance between the demand and supply of oxygen in the body. In this study, we comprehensively analyze the cardiovascular symptoms, histology, and underlying mechanisms associated with COVID-19. Our aim is to contribute to the identification of future research objectives and aid in the advancement of therapeutic management approaches.
Collapse
Affiliation(s)
- Yusuf A Siddique
- Basic Sciences, St. George's University School of Medicine, True Blue, GRD
| | | | | | - Ahmad Sebai
- School of Medicine, California University of Science and Medicine, Colton, USA
| | - Lubhani Sharma
- Family Medicine, Dayanand Medical College and Hospital, Ludhiana, IND
| | - Mohamed Hassouba
- Pediatrics, SUNY Downstate Health Sciences University, Brooklyn, USA
| | - Ghazala S Virk
- Internal Medicine, Avalon University School of Medicine, Ohio, USA
| |
Collapse
|
41
|
Singh M, Lo SH, Dubey R, Kumar S, Chaubey KK, Kumar S. Plant-Derived Natural Compounds as an Emerging Antiviral in Combating COVID-19. Indian J Microbiol 2023; 63:429-446. [PMID: 38031604 PMCID: PMC10682353 DOI: 10.1007/s12088-023-01121-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a human virus that burst at Wuhan in China and spread quickly over the world, leading to millions of deaths globally. The journey of this deadly virus to different mutant strains is still ongoing. The plethora of drugs and vaccines have been tested to cope up this pandemic. The herbal plants and different spices have received great attention during pandemic, because of their anti-inflammatory, and immunomodulatory properties in treating viruses and their symptoms. Also, it has been shown that nano-formulation of phytochemicals has potential therapeutic effect against COVID-19. Furthermore, the plant derived compound nano-formulation specifically increases its antiviral property by enhancing its bioavailability, solubility, and target-specific delivery system. This review highlights the potentiality of herbal plants and their phytochemical against SARS-CoV-2 utilizing different mechanisms such as blocking the ACE-2 receptors, inhibiting the main proteases, binding spike proteins and reducing the cytokine storms.
Collapse
Affiliation(s)
- Mansi Singh
- Department of Pharmacy, Institute of Pharmaceutical Research, GLA University, Mathura, UP 281406 India
| | - Shih-Hsiu Lo
- Department of Urology, Taipei Medical University Hospital, Taipei, Taiwan
| | - Rajni Dubey
- Division of Cardiology, Department of Internal Medicine, Taipei Medical University Hospital, No. 252, Wuxing Street, Taipei, 11031 Taiwan
| | - Sudhashekhar Kumar
- Department of Physiology, School of Medical Sciences and Research, Sharda University, Greater Noida, UP 201310 India
| | - Kundan Kumar Chaubey
- Division of Research and Innovation, School of Applied and Life Sciences, Uttaranchal University, Arcadia Grant, P.O. Chandanwari, Premnagar, Dehradun, Uttarakhand 248007 India
- School of Basic and Applied Sciences, Sanskriti University, Mathura, UP 281401 India
| | - Sanjay Kumar
- Biological and Bio-Computational Lab, Department of Life Science, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida, UP 201310 India
| |
Collapse
|
42
|
Landi A, Morici N, Vranckx P, Frigoli E, Bonacchini L, Omazzi B, Tresoldi M, Camponovo C, Moccetti T, Valgimigli M. Edoxaban and/or colchicine in outpatients with COVID-19: rationale and design of the CONVINCE trial. J Cardiovasc Med (Hagerstown) 2023; 24:920-930. [PMID: 37942793 DOI: 10.2459/jcm.0000000000001556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
BACKGROUND An excessive inflammatory response and a hypercoagulable state are not infrequent in patients with coronavirus disease-2019 (COVID-19) and are associated with adverse clinical outcomes. However, the optimal treatment strategy for COVID-19 patients managed in the out-of-hospital setting is still uncertain. DESIGN The CONVINCE (NCT04516941) is an investigator-initiated, open-label, blinded-endpoint, 2 × 2 factorial design randomized trial aimed at assessing two independently tested hypotheses (anticoagulation and anti-inflammatory ones) in COVID-19 patients. Adult symptomatic patients (≥18 years of age) within 7 days from reverse transcription-PCR (RT-PCR) diagnosis of SARS-CoV-2 infection managed at home or in nursery settings were considered for eligibility. Eligible patients fulfilling all inclusion and no exclusion criteria were randomized to edoxaban versus no treatment (anticoagulation hypothesis) and colchicine versus no treatment (anti-inflammatory hypothesis) in a 1 : 1:1 : 1 ratio. The study had two co-primary endpoints (one for each randomization), including the composite of major vascular thrombotic events at 25 ± 3 days for the anticoagulation hypothesis and the composite of SARS-CoV-2 detection rates at 14 ± 3 days by RT-PCR or freedom from death or hospitalizations (anti-inflammatory hypothesis). Study endpoints will be adjudicated by a blinded Clinical Events Committee. With a final sample size of 420 patients, this study projects an 80% power for each of the two primary endpoints appraised separately. CONCLUSION The CONVINCE trial aims at determining whether targeting anticoagulation and/or anti-inflammatory pathways may confer benefit in COVID-19 patients managed in the out-of-hospital setting. TRIAL REGISTRATION ClinicalTrials.gov number, NCT04516941.
Collapse
Affiliation(s)
- Antonio Landi
- Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale (EOC)
- Department of Biomedical Sciences, University of Italian Switzerland, Lugano, Switzerland
| | - Nuccia Morici
- IRCCS S. Maria Nascente - Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| | - Pascal Vranckx
- the Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Enrico Frigoli
- Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale (EOC)
| | - Luca Bonacchini
- Emergency Department, ASST Great Metropolitan Hospital Niguarda, Milan
| | - Barbara Omazzi
- Emergency Unit, ASST Rhodense, Garbagnate Milanese, Italy
| | - Moreno Tresoldi
- Unit of General Medicine and Advanced Care, IRCCS San Raffaele Hospital, Milan
| | - Claudio Camponovo
- Department of Anesthesiology, Clinica Ars Medica, Genolier Swiss Medical Network, Gravesano
| | | | - Marco Valgimigli
- Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale (EOC)
- Department of Biomedical Sciences, University of Italian Switzerland, Lugano, Switzerland
- University of Bern, Bern, Switzerland
| |
Collapse
|
43
|
Yousaf MH, Ali M, Ahmad N, Yousaf G. Development and Validation of RP-HPLC Method for the Determination of Enoxaparin Sodium in Dry Injection Formulation. ACS OMEGA 2023; 8:44988-44994. [PMID: 38046333 PMCID: PMC10688014 DOI: 10.1021/acsomega.3c06445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/26/2023] [Accepted: 10/30/2023] [Indexed: 12/05/2023]
Abstract
Enoxaparin sodium is an anticoagulant medication that is used as a blood thinning agent. It is mostly used for the treatment and prevention of deep vein thrombosis (DVT) and pulmonary embolism (PE). It is also used in certain surgeries and during pregnancy. For the treatment of acute coronary syndrome (ACS) and heart attacks, it may be used. Enoxaparin sodium was validated by the RP-HPLC method. A simple RP-HPLC method was developed in a single HPLC run in a dry powder injection formulation. All injections of HPLC sample were 20 μL volume. The chromatographic separation was completed in the isocratic mode. The used column was USP-L8 (250 mm × 4.6 mm) of BDS type of 10 μm meters in the same mobile phase throughout the analysis by using methanol and ultrapure water with a ratio of 7:93, respectively. The flow rate was 1.0 mL/min. The mobile phase was filtered through 0.45 μm filter paper, and isocratic elution was performed. The refractive index (RI) detector was used to analyze this sample. The specific peak of enoxaparin sodium was observed at 5:56 min. The calculated detection limit (LOD) was 0.351 ppm, and the calculated quantitation limit was 1.063 ppm. In repeatability of precision, the average calculated assay (%) was 100.85%, and the calculated RSD (%) was 0.01. In the accuracy test, the RSD (%) was 0.50, and the mean recovery (%) was 100.35. The system's suitability was within the limit. This newly developed method is proposed according to ICH guidelines, and rules and can be applied effectively for the exact estimation of enoxaparin sodium in injection formulation. This newly developed methodology is affordable in cost as long as less time is taken and the consumption of samples is in smaller quantities for every investigation. In medicinal chemistry, the USP (United States Pharmacopeia) and BP (British Pharmacopeia) are directly involved in production as well as in quality testing.
Collapse
Affiliation(s)
| | - Majid Ali
- Department
of Chemistry, Lahore Garrison University, Lahore 54792, Pakistan
| | - Naveed Ahmad
- Department
of Chemistry, University of Agriculture, Faisalabad 38000, Pakistan
| | - Ghufran Yousaf
- Department
of Agronomy, Faculty of Crop & Food Science, PMAS Arid Agriculture University, Rawalpindi 46300, Pakistan
| |
Collapse
|
44
|
Aggarwal A, Singh TK, Pham M, Godwin M, Chen R, McIntyre TM, Scalise A, Chung MK, Jennings C, Ali M, Park H, Englund K, Khorana AA, Svensson LG, Kapadia S, McCrae KR, Cameron SJ. Dysregulated Platelet Function in Patients with Post-Acute Sequelae of COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.18.545507. [PMID: 38045316 PMCID: PMC10690211 DOI: 10.1101/2023.06.18.545507] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Background Post-acute sequelae of COVID-19 (PASC), also referred as Long-COVID, sometimes follows COVID-19, a disease caused by SARS-CoV-2. While SARS-CoV-2 is well-known to promote a prothrombotic state, less is known about the thrombosis risk in PASC. Aim Our objective was to evaluate the platelet function and thrombotic potential in patients following recovery from SARS-CoV-2 with clear symptoms of PASC. Methods PASC patients and matched healthy controls were enrolled in the study on average 15 months after documented SARS-CoV-2 infection. Platelet activation was evaluated by Light Transmission Aggregometry (LTA) and flow cytometry in response to platelet surface receptor agonists. Thrombosis in platelet-deplete plasma was evaluated by Factor Xa activity. A microfluidics system assessed thrombosis in whole blood under shear stress conditions. Results A mild increase in platelet aggregation in PASC patients through the thromboxane receptor was observed and platelet activation through the glycoprotein VI (GPVI) receptor was decreased in PASC patients compared to age- and sex-matched healthy controls. Thrombosis under shear conditions as well as Factor Xa activity were reduced in PASC patients. Plasma from PASC patients was an extremely potent activator of washed, healthy platelets - a phenomenon not observed when stimulating healthy platelets after incubation with plasma from healthy individuals. Conclusions PASC patients show dysregulated responses in platelets and coagulation in plasma, likely caused by a circulating molecule that promotes thrombosis. A hitherto undescribed protective response appears to exists in PASC patients to counterbalance ongoing thrombosis that is common to SARS-CoV-2 infection.
Collapse
|
45
|
Kusudo E, Murata Y, Kawamoto S, Egi M. Variant-derived SARS-CoV-2 spike protein does not directly cause platelet activation or hypercoagulability. Clin Exp Med 2023; 23:3701-3708. [PMID: 37208552 PMCID: PMC10198021 DOI: 10.1007/s10238-023-01091-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/11/2023] [Indexed: 05/21/2023]
Abstract
Thrombosis has been associated with severity and mortality in COVID-19. SARS-CoV-2 infects the host via its spike protein. However, direct effects of spike proteins from SARS-CoV-2 variants on platelet activity and coagulability have not been examined. An ethically approved ex vivo study was performed under a preplanned power analysis. Venous blood was collected from 6 healthy subjects who gave prior written consent. The samples were divided into 5 groups: without spike proteins (group N) and with spike proteins derived from alpha, beta, gamma, and delta SARS-CoV-2 variants (groups A, B, C, and D, respectively). Platelet aggregability, P-selectin expression, platelet-associated complement-1 (PAC-1) binding, platelet count, and mean platelet volume (MPV) were measured in all 5 groups, and thromboelastography (TEG) parameters were measured in groups N and D. The % change in each parameter in groups A to D was calculated relative to the value in group N. Data were analyzed by Friedman test, except for TEG parameters, which were evaluated by Wilcoxon matched pairs test. P < 0.05 was considered significant. This study included 6 participants based on a power analysis. There were no significant differences in platelet aggregability under stimulation with adenosine diphosphate 5 µg/ml, collagen 0.2 or 0.5 µg/ml, and Ser-Phe-Leu-Leu-Arg-Asn-amide trifluoroacetate salt (SFLLRN) 0.5 or 1 µM in groups A-D compared to group N. There were also no significant differences in P-selectin expression and PAC-1 binding under basal conditions or SFLLRN stimulation, and no significant differences in platelet count, MPV and TEG parameters. Platelet hyperactivity and blood hypercoagulability have been reported in COVID-19 patients, but spike proteins at 5 µg/ml from SARS-CoV-2 variants (alpha, beta, gamma, delta) did not directly cause these effects in an ex vivo study. This study was approved by the Ethics Committee of Kyoto University Hospital (R0978-1) on March 06, 2020.
Collapse
Affiliation(s)
- Eriko Kusudo
- Department of Anesthesia, Kyoto University Hospital, 54 Shogoin-kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Yutaka Murata
- Department of Anesthesia, Kyoto University Hospital, 54 Shogoin-kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
- Department of Anesthesia, Kitano Hospital, 2-4-20 Ohgimachi, Kita-ku, Osaka, 530-8480, Japan
| | - Shuji Kawamoto
- Department of Anesthesia, Kyoto University Hospital, 54 Shogoin-kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan.
| | - Moritoki Egi
- Department of Anesthesia, Kyoto University Hospital, 54 Shogoin-kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| |
Collapse
|
46
|
Dalbeni A, Susca N, Daidone M, Rossi I, Giontella A, Cimellaro A, Talerico G, Pietrangelo A, Sesti G, Zaccone V, Villani R. Low dose aspirin and clinical outcomes in patients with SARS-CoV-2 pneumonia: a propensity score-matched cohort analysis from the National SIMI‑COVID‑19 Registry. Intern Emerg Med 2023; 18:2311-2319. [PMID: 37751084 DOI: 10.1007/s11739-023-03432-3] [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: 01/17/2023] [Accepted: 09/11/2023] [Indexed: 09/27/2023]
Abstract
BACKGROUND SARS- CoV-2 virus has had dramatic consequences worldwide being able to cause acute respiratory distress syndrome (ARDS), massive thrombosis and pulmonary embolism and, finally, patients' death. In COVID-19 infection, platelets have a procoagulant phenotype that can cause thrombosis in the pulmonary and systemic vascular network. Aspirin is a well-known anti-platelet drug widely used for the prevention of cardiovascular events and systematic reviews suggest a possible benefit of low-dose aspirin (LDA) use in the prevention and treatment of ARDS in patients with COVID-19 infection. However, several studies are available in the literature which do not support any benefits and no association with the patients' outcome. Therefore, currently available data are inconclusive. MATERIALS AND PATIENTS Data from the nationwide cohort multicenter study of the Italian Society of Internal Medicine (SIMI) were analyzed. We conducted a propensity score-matched cohort analysis to investigate the impact of chronic assumption of LDA on mortality of adult COVID-19 patients admitted in Internal Medicine Units (IMU). Data from 3044 COVID-19 patients who referred to 41 Italian hospitals between February 3rd to May 8th 2020 were analyzed. A propensity score-matched analysis was conducted using the following variables: age, sex, hypertension, hyperlipidemia diabetes, atrial fibrillation, cerebrovascular disease, COPD, CKD and stratified upon LDA usage, excluding anticoagulant treatment. After matching, 380 patients were included in the final analysis (190 in LDA group and 190 in no-LDA group). RESULTS 66.2% were male, median age was 77 [70-83]. 34.8% of the population died during the hospitalization. Cardiovascular diseases were not significantly different between the groups. After comparison of LDA and no-LDA subgroups, we didn't record a significant difference in mortality rate (35.7% vs 33.7%) duration of hospital stay and ICU admission. In a logistic regression model, age (OR 1.05; 95% CI 1.01-1.09), FiO2 (OR 1.024; 95% CI 1.03-1.04) and days between symptoms onset and hospitalization (OR 0.93; 95% CI 0.87-0.99) were the only variables independently associated with death.
Collapse
Affiliation(s)
- A Dalbeni
- Section General Medicine C and Liver Unit, Department of Medicine, Azienda Ospedaliera Universitaria Integrata Verona, University of Verona, Verona, Italy
| | - N Susca
- Department of Biomedical Sciences and Human Oncology, "Aldo Moro" University of Bari Medical School, 70124, Bari, Italy
| | - M Daidone
- Internal Medicine and Stroke Care Ward. Department of Health Promotion, Maternal and Infant Care, Internal Medicine and Medical Specialties, "G. D'Alessandro", University of Palermo, Piazza delle Cliniche N.2, Palermo, Italy
| | - I Rossi
- Department of Medicine and Aging Sciences, Clinica Medica" Institute, "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - A Giontella
- Section General Medicine C and Liver Unit, Department of Medicine, Azienda Ospedaliera Universitaria Integrata Verona, University of Verona, Verona, Italy
| | - A Cimellaro
- Internal Medicine Unit, Pugliese-Ciaccio, Hospital, 88100, Catanzaro, Italy
| | - G Talerico
- Internal Medicine Unit, Policlinico Casilino, Rome, Italy
| | - A Pietrangelo
- Internal Medicine Unit, Department of Surgical and Medical Sciences for Children and Adults, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - G Sesti
- Department of Clinical and Molecular Medicine, University of Rome-Sapienza, 00189, Rome, Italy
| | - V Zaccone
- Department of Emergency Medicine, Internal and Sub-Intensive Medicine, Azienda Ospedaliero-Universitaria "Ospedali Riuniti", 60166, Ancona, Italy.
| | - R Villani
- Liver Unit, Centro Universitario per la Ricerca e la Cura delle Epatopatie (C.U.R.E.), Università di Foggia, 71100, Foggia, Italy
| |
Collapse
|
47
|
DE Vito A, Saderi L, Fiore V, Geremia N, Princic E, Fanelli C, Muredda AA, Panu Napodano C, Moi G, Maida I, Fois AG, Sotgiu G, Madeddu G, Babudieri S. Early treatment with low-molecular-weight heparin reduces mortality rate in SARS-CoV-2 patients. Panminerva Med 2023; 65:286-291. [PMID: 35622392 DOI: 10.23736/s0031-0808.22.04572-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Since the beginning of the SARS-CoV-2 pandemic, millions of people have been infected and died. Different therapeutic approaches have been recommended, but only a few have shown clinical advantages. Low-molecular-weight heparin (LMWH) has been recommended to prevent COVID-19-related thrombo-embolic events. We aimed to evaluate the impact of early treatment with LMWH on hospital admission and death in patients with SARS-CoV-2 infection. METHODS We conducted an observational monocentric retrospective study to evaluate the preventive role of LMWH on the mortality rate of COVID-19 patients. SARS-CoV-2 infected patients were recruited from the beginning of the Italian epidemic to March 31, 2021. We excluded patients with missing data and those chronically exposed to LMWH. Treatment prescription was based on international and national guidelines and modified depending on clinical presentation and drug-drug interactions. RESULTS Seven hundred thirty-four SARS-CoV-2 infected patients were recruited, with 357 (48.6%) males and a median (IQR) age of 77.9 (65-85.7) years. 77.5% of people developed SARS-CoV-2-related symptoms and 62.8% were admitted to the hospital, and 20.2% died. Four hundred ninety-two (67%) started LMWH. In particular, 296 (40.3%) were treated within five days since symptoms onset. At logistic regression, early LMWH therapy was associated with lower mortality. Furthermore, remdesivir treatment showed a lower risk of death. On the contrary, age, BMI>30 kg/m2, neurological diseases, fever or dyspnea were associated with an increased risk of death. CONCLUSIONS Early treatment with LMWH was associated with lower mortality in our cohort. Further studies are needed to better assess the role of wider LMWH administration in terms of timing and regimen dose.
Collapse
Affiliation(s)
- Andrea DE Vito
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy -
| | - Laura Saderi
- Unit of Clinical Epidemiology and Medical Statistics, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Vito Fiore
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Nicholas Geremia
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Elija Princic
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Chiara Fanelli
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Alberto A Muredda
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Catello Panu Napodano
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Giulia Moi
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Ivana Maida
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Alessandro G Fois
- Unit of Respiratory Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Giovanni Sotgiu
- Unit of Clinical Epidemiology and Medical Statistics, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Giordano Madeddu
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Sergio Babudieri
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
48
|
Forlenza EM, Serino J, Weintraub MT, Burnett RA, Karas V, Della Valle CJ. Elective Joint Arthroplasty Should be Delayed by One Month After COVID-19 Infection to Prevent Postoperative Complications. J Arthroplasty 2023; 38:1676-1681. [PMID: 36813216 PMCID: PMC9941067 DOI: 10.1016/j.arth.2023.02.032] [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: 07/04/2022] [Revised: 02/05/2023] [Accepted: 02/11/2023] [Indexed: 02/23/2023] Open
Abstract
BACKGROUND It remains unclear whether a history of recent COVID-19 infection affects the outcomes and risks of complications of total joint arthroplasty (TJA). The purpose of this study was to compare the outcomes of TJA in patients who have and have not had a recent COVID-19 infection. METHODS A large national database was queried for patients undergoing total hip and total knee arthroplasty. Patients who had a diagnosis of COVID-19 within 90-days preoperatively were matched to patients who did not have a history of COVID-19 based on age, sex, Charlson Comorbidity Index, and procedure. A total of 31,453 patients undergoing TJA were identified, of which 616 (2.0%) had a preoperative diagnosis of COVID-19. Of these, 281 COVID-19 positive patients were matched with 281 patients who did not have COVID-19. The 90-day complications were compared between patients who did and did not have a diagnosis of COVID-19 at 1, 2, and 3 months preoperatively. Multivariate analyses were used to further control for potential confounders. RESULTS Multivariate analysis of the matched cohorts showed that COVID-19 infection within 1 month prior to TJA was associated with an increased rate of postoperative deep vein thrombosis (odds ratio [OR]: 6.50, 95% confidence interval: 1.48-28.45, P = .010) and venous thromboembolic events (odds ratio: 8.32, confidence interval: 2.12-34.84, P = .002). COVID-19 infection within 2 and 3 months prior to TJA did not significantly affect outcomes. CONCLUSION COVID-19 infection within 1 month prior to TJA significantly increases the risk of postoperative thromboembolic events; however, complication rates returned to baseline after that time point. Surgeons should consider delaying elective total hip arthroplasty and total knee arthroplasty until 1 month after a COVID-19 infection.
Collapse
Affiliation(s)
| | | | | | | | - Vasili Karas
- Rush University Medical Center, Chicago, Illinois
| | | |
Collapse
|
49
|
Jovanikić O, Stevanović G, Đorđevic B, Jovanović M, Lepić M. Mathematical model of aging in COVID-19. J Med Biochem 2023; 42:383-391. [PMID: 37814624 PMCID: PMC10560502 DOI: 10.5937/jomb0-39602] [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: 06/17/2022] [Accepted: 11/21/2022] [Indexed: 10/11/2023] Open
Abstract
Background The aim was examination of the intimamedia thickness of carotid arteries in COVID-19 infection. Methods In 50 patients, the thickness of the intimomedial complex (IMT) in the common carotid arteries was measured. The values were compared with the control group in 2006-9. The condition of the lungs was assessed by ultrasound score (It score) (0-42) as mild (0-14) or mediumsevere (15-28) Covid. IMT thickening risk factors and the value of fibrinogen, IL-6 and CRP were recorded. Two IMT prediction models were formed. The socio-epidemiological model predicts the development of IMT based on epidemiological factors. Apart from these factors, the second model also includes the values of the mentioned biomarkers. Results It score 20±6, IMT values right: median 0.99 mm, p25=0.89, p75=1.14; left: 1±0.22 mm. Control: IMTright: median 0.7 mm, p25=0.68 mm; p75=0-9 mm; left: median=0.75 mm, p25=0.6 mm, p75=1.0 mm. The group/control difference is highly significant. Epide mio - logical model: logit (IMT)= 4.463+(2.021+value for GEN)+(0.055x AGE value)+(-3.419x RF value)+(-4.447x SM value)+(5.115x HTA value)+(3.56x DM value)+ (22.389x LIP value)+(24.206x CVD value)+(1.449x other value)+(-0.138x It score value)+(0.19xBMI value). Epidemiological-inflammatory model: logit (IMT)=5.204+ (2.545x GEN value)+(0.076x AGE value)+(-6.132x RF value)+(-7.583x SM value)+(8.744x HTA value)+(6.838x DM value)+(25.446x LIP value)+(28.825x CVD value)+ (2.487x other value)+(-0.218xIt score value)+(0.649x BMI value) +(-0.194x fibrinogen value)+(0.894x IL-6 value)+(0.659x CRP value). Values for both models Exp(B)=4.882; P of sample=0.83; logit=-0.19; OR= 23.84; model accuracy for the first model 87% and for the second 88%; Omnibus test of the first model c2=34.324; p=0.000; reliability coefficient -2LogLH=56.854; Omnibus test of the second model c2=39.774; p=0.000; and -2LogLH=51.403. Conclusions The ageing of blood vessels in COVID-19 can be predicted.
Collapse
Affiliation(s)
| | - G. Stevanović
- University of Belgrade, Faculty of Medicine, Belgrade
| | | | | | - Milan Lepić
- University of Defense, Faculty of Medicine, Belgrade
| |
Collapse
|
50
|
Bregel LV, Efremova OS, Kostyunin KY, Rudenko NY, Kozlov YA, Albot VV, Knyzeva NА, Tolmacheva OV, Ovanesyan SV, Barakin AO, Pak KO, Belousova LV, Korinets TS, Kostik MM. Thrombosis in Multisystem Inflammatory Syndrome Associated with COVID-19 in Children: Retrospective Cohort Study Analysis and Review of the Literature. Biomedicines 2023; 11:2206. [PMID: 37626703 PMCID: PMC10452691 DOI: 10.3390/biomedicines11082206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/03/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
Background: The causative agent of the new coronavirus infection SARS-CoV-2 has unique properties causing hyperinflammatory syndrome and cytokine storm, as well as widespread endotheliitis and thrombotic microangiopathy, initially detected in the lungs of adult patients who died from a severe form of the disease. Venous and arterial thrombosis in adults were identified as common causes of severe complications and deaths in new coronavirus infections. There are very few reports of thrombotic events in children with COVID-19 in the literature. Methods: We conducted a retrospective analysis of the histories of 60 patients in the Irkutsk Regional Children's Clinical Hospital from November 2020 to November 2022 with a MIS-C diagnosis established according to WHO criteria, of which 8 (13.3%) were diagnosed with venous and/or arterial thrombosis, confirmed by laboratory and ultrasound and/or X-ray methods. Results: The average age of children with thrombosis (Me) was 7.5 years (min 4 months, max 17 years), with a M:F ratio of 3.0. Venous thrombosis was detected in six of the eight patients, including in the deep veins of the lower extremities in four. Pulmonary embolism occurred in two (one of them was fatal), and cerebral venous sinus thrombosis and thrombosis of the branches of the upper and lower vena cava were found in one patient. Extensive bilateral stroke due to thrombosis of the large cerebral arteries occurred in two patients, including one in combination with distal gangrene. Secondary thrombotic renal microangiopathy took place in three of the eight patients. Among these three, atypical HUS was diagnosed in one case. Multiple thrombosis involving the venous and arterial bed was detected in four of the eight patients. High levels of D-dimer, thrombocytopenia, increased NT-proBNP, cerebral coma, and aseptic meningitis were the events most often associated with thrombosis. All patients received immunomodulatory therapy (immunoglobulin, dexamethasone/methylprednisolone), pathogenetic therapy for multiorgan failure, anticoagulant therapy with heparin/LMWH, and acetylsalicylic acid. Biologics were used in two patients. Conclusions: The main predictors of thrombosis in children with MIS-C were increased D-dimer, thrombocytopenia, hospitalization in the ICU, and noncardiogenic pulmonary edema. Thrombosis of the deep veins of the lower extremities, large cerebral arteries, and secondary thrombotic microangiopathy was common. There was a single death (12.5% of the eight patients), associated with PE.
Collapse
Affiliation(s)
- Liudmila V. Bregel
- Department of Pediatrics, Irkutsk State Medical Academy of Postgraduate Education, Branch of Russian Medical Academy of Continuous Professional Education, 664049 Irkutsk, Russia
- Irkutsk Regional Children’s Hospital, 664022 Irkutsk, Russia
| | - Olesya S. Efremova
- Department of Pediatrics, Irkutsk State Medical Academy of Postgraduate Education, Branch of Russian Medical Academy of Continuous Professional Education, 664049 Irkutsk, Russia
- Irkutsk Regional Children’s Hospital, 664022 Irkutsk, Russia
| | - Kirill Y. Kostyunin
- Irkutsk Regional Diagnostic Centre, Department of Clinical Pathomorpholigy, 664047 Irkutsk, Russia;
- Pathology Department, Irkutsk State Medical University, 664003 Irkutsk, Russia
| | | | - Yury A. Kozlov
- Irkutsk Regional Children’s Hospital, 664022 Irkutsk, Russia
- Pathology Department, Irkutsk State Medical University, 664003 Irkutsk, Russia
| | - Vadim V. Albot
- Department of Pediatrics, Irkutsk State Medical Academy of Postgraduate Education, Branch of Russian Medical Academy of Continuous Professional Education, 664049 Irkutsk, Russia
- Irkutsk Regional Children’s Hospital, 664022 Irkutsk, Russia
| | | | | | | | - Alexander O. Barakin
- Department of Pediatrics, Irkutsk State Medical Academy of Postgraduate Education, Branch of Russian Medical Academy of Continuous Professional Education, 664049 Irkutsk, Russia
- Irkutsk Regional Children’s Hospital, 664022 Irkutsk, Russia
| | - Ki O. Pak
- Irkutsk Regional Children’s Hospital, 664022 Irkutsk, Russia
| | | | | | - Mikhail M. Kostik
- Hospital Pediatry, Saint Petersburg State Pediatric Medical University, 194100 Saint Petersburg, Russia
| |
Collapse
|