1
|
Chung J, Pierce J, Franklin C, Olson RM, Morrison AR, Amos-Landgraf J. Translating animal models of SARS-CoV-2 infection to vascular, neurological and gastrointestinal manifestations of COVID-19. Dis Model Mech 2025; 18:dmm052086. [PMID: 40195851 PMCID: PMC12010913 DOI: 10.1242/dmm.052086] [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: 04/09/2025] Open
Abstract
Since the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) initiated a global pandemic resulting in an estimated 775 million infections with over 7 million deaths, it has become evident that COVID-19 is not solely a pulmonary disease. Emerging evidence has shown that, in a subset of patients, certain symptoms - including chest pain, stroke, anosmia, dysgeusia, diarrhea and abdominal pain - all indicate a role of vascular, neurological and gastrointestinal (GI) pathology in the disease process. Many of these disease processes persist long after the acute disease has been resolved, resulting in 'long COVID' or post-acute sequelae of COVID-19 (PASC). The molecular mechanisms underlying the acute and systemic conditions associated with COVID-19 remain incompletely defined. Appropriate animal models provide a method of understanding underlying disease mechanisms at the system level through the study of disease progression, tissue pathology, immune system response to the pathogen and behavioral responses. However, very few studies have addressed PASC and whether existing models hold promise for studying this challenging problem. Here, we review the current literature on cardiovascular, neurological and GI pathobiology caused by COVID-19 in patients, along with established animal models of the acute disease manifestations and their prospects for use in PASC studies. Our aim is to provide guidance for the selection of appropriate models in order to recapitulate certain aspects of the disease to enhance the translatability of mechanistic studies.
Collapse
Affiliation(s)
- James Chung
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| | - Julia Pierce
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI 02908, USA
- Department of Research, Ocean State Research Institute, Inc., Providence, RI 02908-4734, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, RI 02908, USA
| | - Craig Franklin
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| | - Rachel M. Olson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
- Laboratory for Infectious Disease Research, University of Missouri, Columbia, MO 65211, USA
| | - Alan R. Morrison
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI 02908, USA
- Department of Research, Ocean State Research Institute, Inc., Providence, RI 02908-4734, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, RI 02908, USA
| | - James Amos-Landgraf
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
2
|
Kakavand G, Arabzadeh S, Mohebbi S, Saeedfar K, Abedini A, Mardani M. Impact of remdesivir treatment on factor VIII gene expression and hematological parameters in COVID-19 patients. Microb Pathog 2025; 204:107536. [PMID: 40187577 DOI: 10.1016/j.micpath.2025.107536] [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: 09/28/2024] [Revised: 03/27/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
The novel coronavirus, COVID-19, which was first identified in December 2019 rapidly spread worldwide and was declared a global pandemic. Beyond respiratory symptoms, COVID-19 often results in coagulation and vascular endothelium disorders, causing increased clotting and bleeding, which are closely linked to the acute phase of the infection. Factor VIII is a crucial protein in the blood coagulation cascade, and elevated FVIII levels have been linked to thrombotic events in COVID-19, highlighting the need to understand its behavior during treatment. Remdesivir is an antiviral drug that has shown promise in reducing recovery time and mortality rates in COVID-19 patients. This study aims to examine the changes in blood factors and the expression of the factor VIII gene in patients treated with Remdesivir. Blood samples were collected from 30 COVID-19 patients before and after Remdesivir treatment and from 20 healthy individuals. Patients with underlying diseases were excluded from the study. RNA was extracted from these samples, followed by cDNA synthesis. The expression of the factor VIII gene was analyzed using Real-Time PCR. The results indicated that blood factors such as Urea, ALK, AST, WBC, and CRP were elevated in the patient group compared to the control group. At the same time, FBS, Urea, ALK, AST, WBC, RDW, INR, and K levels increased in the Remdesivir treatment group (P < 0.001). Conversely, MCHC, RBC, and Ca levels decreased in both patient and treatment groups compared to the control group (P < 0.001). The expression of the FVIII gene was upregulated approaching 2 times in COVID-19 patients and 1.5-fold in the treatment group compared to the control group (P < 0.001). However, no significant changes were observed in FVIII expression before and after Remdesivir treatment. However, a positive correlation between RBC, FBS, and Urea in the patient group and a negative correlation between RDW and FVIII expression levels was observed. In the treatment group, FVIII expression level correlated negatively with Urea, P, and RDW. These findings suggest that elevated FVIII levels are associated with disease severity and excessive coagulation in COVID-19 patients. Additionally, Remdesivir does not appear to exacerbate the coagulation process.
Collapse
Affiliation(s)
- Ghazal Kakavand
- Department of Biology, Faculty of Basic Science, Ale Taha Institute of Higher Education, Tehran, Iran
| | - Somayeh Arabzadeh
- Department of Biology, Faculty of Basic Science, Ale Taha Institute of Higher Education, Tehran, Iran
| | - Sohameh Mohebbi
- Department of Biology, Faculty of Basic Science, Ale Taha Institute of Higher Education, Tehran, Iran.
| | - Kayvan Saeedfar
- Tracheal Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Atefeh Abedini
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Mardani
- Shahid Beheshti University of Medical Sciences, Infectious Disease Department, Loghman Hakim Hospital, Tehran, Iran
| |
Collapse
|
3
|
Cho J, Lee NK, Kim JY, Lee CS, Byeon SH, Kim SS, Lee SW, Kim YJ. Trends in the Incidence of Retinal Artery and Vein Occlusion Before and After COVID-19: A National Study in South Korea (2017-2022). Am J Ophthalmol 2025; 274:101-111. [PMID: 40058535 DOI: 10.1016/j.ajo.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/17/2024] [Accepted: 03/03/2025] [Indexed: 03/29/2025]
Abstract
PURPOSE This study investigated the incidence of new retinal artery occlusion (RAO) and retinal vein occlusion (RVO) during the coronavirus disease-19 (COVID-19) pandemic compared to pre-outbreak periods in South Korea. DESIGN Nationwide population-based retrospective cohort study. PARTICIPANTS Individuals diagnosed with RAO or RVO. METHODS We evaluated data from 326 154 patients diagnosed with RAO (n = 32 028), RVO (n = 304 405), or both (n = 10 279) from January 1, 2017 to December 31, 2022. We calculated the incidence rate ratios (IRR) of RAO and RVO using 2019 as the reference year, making comparisons across the total population and within age and sex subgroups. We examined correlations between the number of new RVO patients and COVID-19 infected or vaccinated persons. Additionally, we compared the systemic characteristics of individuals with RAO and RVO, adjusting for multiple comparisons with Bonferroni correction. MAIN OUTCOME MEASURES Annual IRRs of RAO and RVO. RESULTS The IRR for RAO showed no significant increase across all age and sex subgroups. Contrastingly, RVO exhibited a consistent IRR in 2020; however, significant increases were observed in 2021 and 2022 among those aged 20-39 years (adjusted IRR in 2021, 1.22, 95% CI, 1.15-1.29; adjusted IRR in 2022, 1.12, 95% CI, 1.06-1.18) as well as those aged 40-64 years (adjusted IRR in 2021, 1.11; 95% CI, 1.09-1.13; adjusted IRR in 2022, 1.11, 95% CI, 1.08-1.12). No correlations were found between the monthly number of patients with RVO and the number of individuals infected with or vaccinated against COVID-19. Additionally, a higher proportion of patients with RVO having hypertension, diabetes, dyslipidemia, and chronic liver disease were seen in 2021 and 2022, as analyzed by ANOVA with Bonferroni correction. CONCLUSIONS The incidence of RAO did not increase during the COVID-19 outbreak. However, the incidence of RVO increased during the pandemic, especially in 2021 and 2022, with the highest rates observed in the 40-64 years age group, likely linked to increases in metabolic diseases rather than COVID-19 infection and vaccination. Further research is warranted to confirm these findings.
Collapse
Affiliation(s)
- Jin Cho
- From the Department of Ophthalmology (J.C., J.Y.K., C.S.L., S.H.B., S.S.K., Y.J.K.), Institute of Vision Research, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Nang Kyeong Lee
- Department of Precision Medicine (N.K.L., S.W.L.), School of Medicine, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jin Yeong Kim
- From the Department of Ophthalmology (J.C., J.Y.K., C.S.L., S.H.B., S.S.K., Y.J.K.), Institute of Vision Research, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Christopher Seungkyu Lee
- From the Department of Ophthalmology (J.C., J.Y.K., C.S.L., S.H.B., S.S.K., Y.J.K.), Institute of Vision Research, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Suk Ho Byeon
- From the Department of Ophthalmology (J.C., J.Y.K., C.S.L., S.H.B., S.S.K., Y.J.K.), Institute of Vision Research, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sung Soo Kim
- From the Department of Ophthalmology (J.C., J.Y.K., C.S.L., S.H.B., S.S.K., Y.J.K.), Institute of Vision Research, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung Won Lee
- Department of Precision Medicine (N.K.L., S.W.L.), School of Medicine, Sungkyunkwan University, Suwon, Republic of Korea.
| | - Yong Joon Kim
- From the Department of Ophthalmology (J.C., J.Y.K., C.S.L., S.H.B., S.S.K., Y.J.K.), Institute of Vision Research, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
4
|
Pereira de Godoy AC, Bulgarelli Bestetti R. Hospitalization and Mortality in Brazilian Children and Adolescents Due to COVID-19: Retrospective Study. JMIR Pediatr Parent 2025; 8:e67546. [PMID: 40393049 DOI: 10.2196/67546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 01/23/2025] Open
Abstract
Background COVID-19 is currently one of the most important medical challenges as it affects the entire population, with children and adolescents being infected as easily as adults. Objective The objective of this study was to evaluate the prevalence of mortality in children and adolescents aged <19 years, compared to that of adults. Methods This retrospective, observational study analyzed the medical records of all patients diagnosed with COVID-19 by real-time reverse transcription-quantitative polymerase chain reaction who were hospitalized at Hospital de Base and the Infant and Maternal Hospital of São José do Rio Preto, São Paulo, Brazil. Out of a total of 8986 hospitalized patients who were COVID-19 positive, 383 (4.26%) were children and adolescents aged <19 years (group 1), and 8603 (95.74%) were adults (group 2). Results Overall, mortality was significantly higher (P<.001) in group 2 (2185/8603, 25.4%) compared to group 1 (12/383, 3.1%). A total of 11 (92%) of the 12 patients in group 1 that died had associated diseases. The mortality rates by age group were as follows: infants aged <1 year, 1.6% (2/123); children aged 1-4 years, 4% (4/95); children aged 5-9 years, 2% (1/47); adolescents aged 10-14 years, 2% (1/40); and adolescents aged 15-19 years, 5% (4/78). Conclusions Mortality from COVID-19 in children and adolescents was significantly lower than that in adults and was associated with other comorbidities.
Collapse
Affiliation(s)
- Ana Carolina Pereira de Godoy
- Hospital da Criança e Materinade de Sao José do Rio Preto-Brazil, Rua Floriano Peixoto 2950, Sao José do Rio Preto, 15020010, Brazil, 55 11 949830105
| | - Reinaldo Bulgarelli Bestetti
- Faculdade de Medicina de Sao José do Rio Preto-Brazi, University of Ribeirão Preto, Sao José do Rio Preto, Brazil
| |
Collapse
|
5
|
Alves MCS, da Silva RCC, de Leitão-Júnior SSP, de Balbino VQ. Therapeutic Approaches for COVID-19: A Review of Antiviral Treatments, Immunotherapies, and Emerging Interventions. Adv Ther 2025:10.1007/s12325-025-03218-3. [PMID: 40338485 DOI: 10.1007/s12325-025-03218-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Accepted: 04/22/2025] [Indexed: 05/09/2025]
Abstract
The coronavirus disease 2019 (COVID-19) global health crisis, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has presented unprecedented challenges to global healthcare systems, leading to rapid advances in treatment development. This review comprehensively examines the current therapeutic approaches for managing COVID-19, including direct-acting antivirals, immunomodulators, anticoagulants, and adjuvant therapies, as well as emerging and experimental approaches. Direct-acting antivirals target various stages of the viral life cycle, offering specific intervention points, while immunomodulators aim to modulate the host's immune response, reducing disease severity. Anticoagulant therapies address the coagulopathy frequently observed in severe cases, and adjuvant treatments provide supportive care to improve overall outcomes. We also explore the challenges and limitations of implementing these treatments, such as drug resistance, variable patient responses, and access to therapies, especially in resource-limited settings. The review also discusses future perspectives, including the potential of next-generation vaccines, personalized medicine, and global collaboration in shaping future COVID-19 treatment paradigms. Continuous innovation, combined with an integrated and adaptable approach, will be crucial to effectively managing COVID-19 and mitigating the impact of future pandemics.
Collapse
Affiliation(s)
- Maria C S Alves
- Laboratory of Bioinformatics and Evolutionary Biology, Center for Biosciences, Genetics Department, Federal University of Pernambuco, Recife, Pernambuco, 50670-423, Brazil.
| | - Ruana C C da Silva
- Laboratory of Health Sciences Research, Federal University of Grande Dourados, Dourados, Mato Grosso do Sul, 79825-070, Brazil
| | - Sérgio S P de Leitão-Júnior
- Laboratory of Bioinformatics and Evolutionary Biology, Center for Biosciences, Genetics Department, Federal University of Pernambuco, Recife, Pernambuco, 50670-423, Brazil
- Serra Talhada Academic Unit, Federal Rural University of Pernambuco, Serra Talhada, Pernambuco, 56909-535, Brazil
| | - Valdir Q de Balbino
- Laboratory of Bioinformatics and Evolutionary Biology, Center for Biosciences, Genetics Department, Federal University of Pernambuco, Recife, Pernambuco, 50670-423, Brazil.
| |
Collapse
|
6
|
Wada H, Shiraki K, Ichikawa Y, Ito N, Inoue H, Moritani I, Masuda J, Yamamoto A, Tomida M, Yoshida M, Kawamura M, Shimaoka M, Iba T, Shimpo H. Implications of Soluble C-type Lectin-Like Receptor 2 Levels in Patients with Coronavirus Disease 2019-Associated with Thrombosis. Thromb Haemost 2025. [PMID: 40174637 DOI: 10.1055/a-2572-1170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
Coronavirus disease 2019 (COVID-19) is often associated with thrombosis. Elevated levels of soluble C-type lectin-like receptor 2 (sCLEC-2), a biomarker for platelet activation, have been reported in COVID-19. Therefore, we examined the behavior of sCLEC-2 levels and their relationship with thrombosis.The clinical course of inflammatory and thrombotic biomarkers was assessed in 271 patients with COVID-19.Inflammatory biomarkers such as C-reactive protein, procalcitonin, and presepsin levels were significantly increased in patients with COVID-19, and these behaviors differed among the clinical course or stages. The plasma D-dimer levels increased slightly and gradually. Platelet counts were within the normal range, and plasma sCLEC-2 levels were markedly increased in most patients with COVID-19. There were 17 patients with thrombosis in this study. Although there was no significant difference in various biomarkers between COVID-19 patients with and without thrombosis, the super formula of sCLEC-2xD-dimer/platelet count in patients with thrombosis was significantly higher than in those without thrombosis. Furthermore, this super formula was significantly higher in COVID-19 patients with severe or critical illness than in those with mild or moderate illness.Elevation of the super formula of sCLEC-2xD-dimer/platelet count was associated with the thrombosis in patients with COVID-19 suggesting the thrombosis in COVID-19 may be caused by the development of microthrombosis.
Collapse
Affiliation(s)
- Hideo Wada
- Department of General Medicine, Mie Prefectural General Medical Center, Yokkaichi, Japan
| | - Katsuya Shiraki
- Department of General Medicine, Mie Prefectural General Medical Center, Yokkaichi, Japan
| | - Yuhuko Ichikawa
- Department of Central Laboratory, Mie Prefectural General Medical Center, Yokkaichi, Japan
| | - Nobuo Ito
- Department of Neurology, Mie Prefectural General Medical Center, Yokkaichi, Japan
| | - Hidekazu Inoue
- Department of Gastroenterology, Mie Prefectural General Medical Center, Yokkaichi, Japan
| | - Isao Moritani
- Department of Gastroenterology, Mie Prefectural General Medical Center, Yokkaichi, Japan
| | - Jun Masuda
- Department of Cardiovascular Medicine, Mie Prefectural General Medical Center, Yokkaichi, Japan
| | - Akitaka Yamamoto
- Department of Emergency and Critical Care Center, Mie Prefectural General Medical Center, Yokkaichi, Japan
| | - Masaki Tomida
- Department of Emergency and Critical Care Center, Mie Prefectural General Medical Center, Yokkaichi, Japan
| | - Masamichi Yoshida
- Department of Respiratory Medicine, Mie Prefectural General Medical Center, Yokkaichi, Japan
| | | | - Motomu Shimaoka
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Toshiaki Iba
- Department of Emergency and Disaster Medicine, Juntendo University, Graduate School of Medicine, Tokyo, Japan
| | - Hideto Shimpo
- Mie Prefectural General Medical Center, Yokkaichi, Japan
| |
Collapse
|
7
|
Pederson T, Tainter CK, Self M, Ghobrial M, Sloane C, Mergen S, Kennis B, Aminlari A, McGuire WC, Wardi G. Controversies in the Management of Acute Pulmonary Embolism in the Emergency Department. J Emerg Med 2025; 71:31-43. [PMID: 39979191 DOI: 10.1016/j.jemermed.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/03/2024] [Accepted: 10/02/2024] [Indexed: 02/22/2025]
Abstract
BACKGROUND Acute pulmonary embolism (PE) is frequently diagnosed in the Emergency Department (ED), and the management approach can be nuanced. OBJECTIVE In this narrative review, we synthesize the literature in selected areas of ongoing controversy regarding the diagnostic and management approaches for acute PE in the ED, and provide evidence-based recommendations to empower emergency physicians (EPs) to provide optimal care in these situations. DISCUSSION d-Dimer is used to clinically exclude the diagnosis of PE patients who are stratified as low risk. However by utilizing likelihood ratio and with certain scoring tools, patient historically considered moderate or high risk for PE may safely be able to have the diagnosis excluded with a negative d-dimer. Traditional risk stratification and management strategies can be cautiously applied to patients with concomitant Coronavirus-19 infection while awaiting more definitive studies. There is an increasing trend in the diagnosis of isolated subsegmental PE, and many patients receiving this diagnosis may be treated without anticoagulation provided that they have no evidence of associated deep vein thrombosis (DVT) and can be closely followed as an outpatient. There is a persistent hesitancy to discharge patients with newly diagnosed acute PE, and existing well-supported risk stratification tools and clinical decision frameworks can support the EP's decision to safely discharge low-risk patients. CONCLUSION tThis review of the literature empowers emergency clinicians to manage challenging PE cases in the ED.
Collapse
Affiliation(s)
- Thomas Pederson
- Department of Emergency Medicine, Division of Anesthesiology Critical Care Medicine, University of California, San Diego, California.
| | | | - Michael Self
- Department of Emergency Medicine, Division of Anesthesiology Critical Care Medicine, University of California, San Diego, California
| | - Mina Ghobrial
- Department of Emergency Medicine, Division of Anesthesiology Critical Care Medicine, University of California, San Diego, California
| | - Christian Sloane
- Department of Emergency Medicine, University of California, San Diego, California
| | - Stephanie Mergen
- Department of Emergency Medicine, Division of Pulmonary, Critical Care Medicine, and Sleep Medicine, University of California, San Diego, California
| | - Brent Kennis
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah
| | - Amir Aminlari
- Department of Emergency Medicine, University of California, San Diego, California
| | - William Cameron McGuire
- Division of Pulmonary, Critical Care, Sleep Medicine, and Physiology, University of California, San Diego, California
| | - Gabriel Wardi
- Department of Emergency Medicine, Division of Pulmonary, Critical Care Medicine, and Sleep Medicine, University of California, San Diego, California
| |
Collapse
|
8
|
Tcholadze G, Pantsulaia I, Ratiani L, Kopaleishvili L, Bolotashvili T, Jorbenadze A, Chikovani T. The Prognostic Value of Circulating Cytokines and Complete Blood Count-Based Inflammatory Markers in COVID-19 Patients With Atrial Fibrillation. Cardiol Res 2025; 16:153-160. [PMID: 40051670 PMCID: PMC11882233 DOI: 10.14740/cr2027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 02/05/2025] [Indexed: 03/09/2025] Open
Abstract
Background Atrial fibrillation (AF) is associated with a high burden of cardiovascular disease, which has been worsened during the coronavirus disease 2019 (COVID-19) pandemic. The purpose of this study was to assess the association between clinical markers, especially interleukin-6 (IL-6) and other inflammatory biomarkers, and the severity of COVID-19 in patients with AF. Methods This retrospective cohort study categorized patients based on clinical presentations and laboratory results to investigate the prognostic significance of inflammatory markers in COVID-19 outcomes among those with AF. The study included 100 hospitalized COVID-19 patients aged between 40 to 80 years and was conducted at the Chapidze Hospital in Tbilisi, Georgia. Patients were then grouped by disease severity according to computed tomography (CT) scores, clinical symptoms, respiratory rate and oxygen saturation. Levels of IL-6 were obtained at three time points during hospitalization. A broad range of laboratory tests, including C-reactive protein (CRP), ferritin, and D-dimer, were also conducted. Results Patients with AF demonstrated significantly elevated levels of IL-6 (P = 0.024), CRP (P = 0.001), and ferritin (P < 0.001), suggesting a severe inflammatory response. D-dimer levels were also notably higher in the AF group (P < 0.005), indicating an increased risk of thrombotic complications. Oxygen saturation levels were significantly lower (P = 0.004) and CT scores higher in patients with AF. Furthermore, the length of hospitalization was longer among patients with AF (median duration significantly higher, P = 0.032), indicating a more severe disease course. Conclusions The proinflammatory markers such as IL-6 are independent predictive markers of COVID-19 severity in AF patients. Overall, it highlights urgent treatment approaches, such as available anti-inflammatory drugs, for COVID-19 patients with arrhythmias. Combining these biomarkers into clinical routines helps us better identify patients at risk and how to treat them.
Collapse
Affiliation(s)
- Giorgi Tcholadze
- Department of Immunology, Tbilisi State Medical University, Tbilisi 0177, Georgia
| | - Ia Pantsulaia
- Department of Immunology, Tbilisi State Medical University, Tbilisi 0177, Georgia
- Vl. Bakhutashvili Institute of Medical Biotechnology, Tbilisi State Medical University, Tbilisi 0159, Georgia
| | | | | | | | | | - Tinatin Chikovani
- Department of Immunology, Tbilisi State Medical University, Tbilisi 0177, Georgia
| |
Collapse
|
9
|
Siegal DM, Tseng EK, Schünemann HJ, Angchaisuksiri P, Cuker A, Dane K, DeSancho MT, Diuguid D, Griffin DO, Klok FA, Lee AI, Neumann I, Pai A, Righini M, Sanfilippo KM, Terrell DR, Akl EA, Al Jabiri R, Al Jabiri Y, Barbara AM, Bognanni A, Akl IB, Boulos M, Brignardello-Petersen R, Chan M, Charide R, Colunga-Lozano LE, Dearness K, Darzi AJ, Hussein H, Karam SG, Kolb P, Mansour R, Morgano GP, Morsi RZ, Muti-Schünemann G, Nadim MK, Noori A, Philip BA, Piggott T, Qiu Y, Benitez YR, Schünemann F, Stevens A, Solo K, Wiercioch W, Mustafa RA, Nieuwlaat R. American Society of Hematology living guidelines on use of anticoagulation for thromboprophylaxis for patients with COVID-19: executive summary. Blood Adv 2025; 9:1247-1260. [PMID: 39437797 PMCID: PMC11950770 DOI: 10.1182/bloodadvances.2024014219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND COVID-19-related critical and acute illness is associated with an increased risk of venous thromboembolism (VTE). These evidence-based recommendations of the American Society of Hematology (ASH) are intended to support patients, clinicians, and other health care professionals in decisions about using anticoagulation for thromboprophylaxis for patients with COVID-19-related critical illness; patients with COVID-19-related acute illness; and those being discharged from the hospital, who do not have suspected or confirmed VTE. METHODS ASH formed a multidisciplinary panel, including patient representatives. The Michael G. DeGroote Cochrane Canada and MacGRADE Centres at McMaster University supported guideline development, including performing systematic reviews (up to June 2023). The panel prioritized clinical questions and outcomes according to their importance for clinicians and patients. The panel used the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) approach to assess certainty in the evidence and make recommendations. RESULTS This is an executive summary of 3 updated recommendations that have been published, which concludes the living phase of the guidelines. For patients with COVID-19-related critical illness, the panel issued conditional recommendations suggesting (a) prophylactic-intensity over therapeutic-intensity anticoagulation and (b) prophylactic-intensity over intermediate-intensity anticoagulation. For patients with COVID-19-related acute illness, conditional recommendations were suggested (a) prophylactic-intensity over intermediate-intensity anticoagulation, and (b) therapeutic-intensity over prophylactic-intensity anticoagulation. The panel issued a conditional recommendation suggesting against the use of postdischarge anticoagulant thromboprophylaxis. CONCLUSIONS These conditional recommendations were made based on low or very low certainty in the evidence, underscoring the need for additional, high-quality, randomized controlled trials for patients with COVID-19.
Collapse
Affiliation(s)
- Deborah M. Siegal
- Department of Medicine and Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Eric K. Tseng
- Division of Hematology/Oncology, St Michael’s Hospital, University of Toronto, Toronto, ON, Canada
| | - Holger J. Schünemann
- Department of Health Research Methods, Evidence, and Impact, Department of Medicine, Michael G. DeGroote Cochrane Canada Centre, and McMaster GRADE Centre, McMaster University, Hamilton, ON, Canada
- Institut für Evidenz in der Medizin, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Pantep Angchaisuksiri
- Division of Hematology, Department of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Adam Cuker
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Kathryn Dane
- Department of Pharmacy, The Johns Hopkins Hospital, Baltimore, MD
| | - Maria T. DeSancho
- Division of Hematology-Oncology, Department of Medicine, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY
| | - David Diuguid
- Division of Hematology, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Daniel O. Griffin
- Division of Infectious Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY
- Optum Tristate, Lake Success, NY
| | - Frederikus A. Klok
- Department of Medicine, Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, The Netherlands
| | - Alfred Ian Lee
- Section of Hematology, Yale School of Medicine, New Haven, CT
| | - Ignacio Neumann
- Escuela de Medicina, Facultad de Medicina y Ciencia, Universidad San Sebastian, Santiago, Chile
| | - Ashok Pai
- Division of Hematology and Oncology, Kaiser Permanente, Oakland, CA
| | - Marc Righini
- Division of Angiology and Hemostasis, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Kristen M. Sanfilippo
- Division of Hematology, Washington University School of Medicine St. Louis, St. Louis, MO
| | - Deirdra R. Terrell
- Department of Biostatistics and Epidemiology, Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Elie A. Akl
- Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | | | - Yazan Al Jabiri
- Division of Hospital Medicine, Washington University in St. Louis, St. Louis, MO
| | - Angela M. Barbara
- Canadian Agency for Drugs and Technologies in Health, Ottawa, ON, Canada
| | - Antonio Bognanni
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada Centre, and McMaster GRADE Centre, McMaster University, Hamilton, ON, Canada
| | - Imad Bou Akl
- Faculty of Medicine, American University of Beirut School of Medicine, Beirut, Lebanon
| | - Mary Boulos
- Postgraduate Medical Education, University of Toronto, Toronto, ON, Canada
| | | | - Matthew Chan
- McMaster GRADE Centre, McMaster University Medical Centre, Hamilton, ON, Canada
| | - Rana Charide
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - Luis E. Colunga-Lozano
- Department of Clinical Medicine, Health Science Center, Universidad de Guadalajara, Guadalajara, Mexico
| | - Karin Dearness
- Library Services, St Joseph’s Healthcare Hamilton, Hamilton, ON, Canada
| | - Andrea J. Darzi
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada Centre, and McMaster GRADE Centre, McMaster University, Hamilton, ON, Canada
| | - Heba Hussein
- Department of Oral Medicine, Oral Diagnosis, and Periodontology, Faculty of Dentistry, Cairo University, Cairo, Egypt
| | - Samer G. Karam
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada Centre, and McMaster GRADE Centre, McMaster University, Hamilton, ON, Canada
| | - Philipp Kolb
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada Centre, and McMaster GRADE Centre, McMaster University, Hamilton, ON, Canada
| | - Razan Mansour
- University of Kansas Medical Center, University of Kansas, Kansas City, KS
| | - Gian Paolo Morgano
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada Centre, and McMaster GRADE Centre, McMaster University, Hamilton, ON, Canada
| | - Rami Z. Morsi
- Department of Neurology, University of Chicago, Chicago, IL
| | | | - Menatalla K. Nadim
- Department of Clinical Pathology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Atefeh Noori
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
- Division of Plastic, Reconstructive and Aesthetic Surgery, Hand Program, University Health Network, Toronto Western Hospital, University of Toronto, Toronto, Canada
| | - Binu A. Philip
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada Centre, and McMaster GRADE Centre, McMaster University, Hamilton, ON, Canada
| | - Thomas Piggott
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
- Department of Family Medicine, Queen’s University, Kingston, ON, Canada
| | - Yuan Qiu
- Department of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Yetiani Roldan Benitez
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | | | - Adrienne Stevens
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada Centre, and McMaster GRADE Centre, McMaster University, Hamilton, ON, Canada
| | - Karla Solo
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada Centre, and McMaster GRADE Centre, McMaster University, Hamilton, ON, Canada
| | - Wojtek Wiercioch
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada Centre, and McMaster GRADE Centre, McMaster University, Hamilton, ON, Canada
| | - Reem A. Mustafa
- Division of Nephrology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Robby Nieuwlaat
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada Centre, and McMaster GRADE Centre, McMaster University, Hamilton, ON, Canada
- University of Kansas Medical Center, University of Kansas, Kansas City, KS
| |
Collapse
|
10
|
Weiss S, Lin HM, Acosta E, Komarova NL, Chen P, Wodarz D, Baine I, Duerr R, Wajnberg A, Gervais A, Bastard P, Casanova JL, Arinsburg SA, Swartz TH, Aberg JA, Bouvier NM, Liu ST, Alvarez RA, Chen BK. Post-transfusion activation of coagulation pathways during severe COVID-19 correlates with COVID-19 convalescent plasma antibody profiles. J Clin Invest 2025; 135:e181136. [PMID: 40091845 PMCID: PMC11910229 DOI: 10.1172/jci181136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 01/24/2025] [Indexed: 03/19/2025] Open
Abstract
Early antibody therapy can prevent severe SARS-CoV-2 infection (COVID-19). However, the effectiveness of COVID-19 convalescent plasma (CCP) therapy in treating severe COVID-19 remains inconclusive. To test a hypothesis that some CCP units are associated with a coagulopathy hazard in severe disease that offsets its benefits, we tracked 304 CCP units administered to 414 hospitalized COVID-19 patients to assess their association with the onset of unfavorable post-transfusion D-dimer trends. CCP recipients with increasing or persistently elevated D-dimer trajectories after transfusion experienced higher mortality than those whose D-dimer levels were persistently low or decreasing after transfusion. Within the CCP donor-recipient network, recipients with increasing or persistently high D-dimer trajectories were skewed toward association with a minority of CCP units. In in vitro assays, CCP from "higher-risk" units had higher cross-reactivity with the spike protein of human seasonal betacoronavirus OC43. "Higher-risk" CCP units also mediated greater Fcγ receptor IIa signaling against cells expressing SARS-CoV-2 spike compared with "lower-risk" units. This study finds that post-transfusion activation of coagulation pathways during severe COVID-19 is associated with specific CCP antibody profiles and supports a potential mechanism of immune complex-activated coagulopathy.
Collapse
Affiliation(s)
| | - Hung-Mo Lin
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | | | | - Dominik Wodarz
- Department of Ecology, Behavior and Evolution, UCSD, La Jolla, California, USA
| | - Ian Baine
- Department of Transfusion Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ralf Duerr
- Department of Medicine
- Department of Microbiology, and
- Vaccine Center, NYU Grossman School of Medicine, New York, New York, USA
| | - Ania Wajnberg
- Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Adrian Gervais
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Laboratory of Human Genetics of Infectious Diseases, INSERM, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris, Paris, France
| | - Paul Bastard
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Laboratory of Human Genetics of Infectious Diseases, INSERM, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris, Paris, France
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Laboratory of Human Genetics of Infectious Diseases, INSERM, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris, Paris, France
- Howard Hughes Medical Institute, New York, New York, USA
| | | | | | | | - Nicole M. Bouvier
- Division of Infectious Diseases and
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Sean T.H. Liu
- Division of Infectious Diseases and
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | |
Collapse
|
11
|
Shakhidzhanov S, Filippova A, Bovt E, Gubkin A, Sukhikh G, Tsarenko S, Spiridonov I, Protsenko D, Zateyshchikov D, Vasilieva E, Kalinskaya A, Dukhin O, Novichkova G, Karamzin S, Serebriyskiy I, Lipets E, Kopnenkova D, Morozova D, Melnikova E, Rumyantsev A, Ataullakhanov F. Severely Ill COVID-19 Patients May Exhibit Hypercoagulability Despite Escalated Anticoagulation. J Clin Med 2025; 14:1966. [PMID: 40142778 PMCID: PMC11943368 DOI: 10.3390/jcm14061966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/07/2025] [Accepted: 03/09/2025] [Indexed: 03/28/2025] Open
Abstract
Introduction: Severely ill COVID-19 patients receiving prophylactic-dose anticoagulation exhibit high rates of thrombosis and mortality. The escalation of anticoagulation also does not reduce mortality and has an uncertain impact on thrombosis rates. The reasons why escalated doses fail to outperform prophylactic doses in reducing risks of thrombosis and death in severely ill COVID-19 patients remain unclear. We hypothesized that escalated anticoagulation would not effectively prevent hypercoagulability and, consequently, would not reduce the risk of thrombosis and death in some severely ill patients. Methods: We conducted a prospective multicenter study that enrolled 3860 COVID-19 patients, including 1654 severely ill. They received different doses of low-molecular-weight or unfractionated heparin, and their blood coagulation was monitored with activated partial thromboplastin time, D-dimer, and Thrombodynamics. A primary outcome was hypercoagulability detected by Thrombodynamics. Blood samples were collected at the trough level of anticoagulation. Results: We found that escalated anticoagulation did not prevent hypercoagulability in 28.3% of severely ill patients at the trough level of the pharmacological activity. Severely ill patients with such hypercoagulability had higher levels of inflammation markers and better creatinine clearance compared to severely ill patients without it. Hypercoagulability detected by Thrombodynamics was associated with a 1.68-fold higher hazard rate for death and a 3.19-fold higher hazard rate for thrombosis. Elevated D-dimer levels were also associated with higher hazard rates for thrombosis and death, while shortened APTTs were not. The simultaneous use of Thrombodynamics and D-dimer data enhanced the accuracy for predicting thrombotic events and fatal outcomes in severely ill patients. Conclusions: Thrombodynamics reliably detects hypercoagulability in COVID-19 patients and can be used in conjunction with D-dimer to assess the risk of thrombosis and death in severely ill patients. The pharmacological effect of LMWH at the trough level might be too low to prevent thrombosis in some severely ill patients with severe inflammation and better creatinine clearance, even if escalated doses are used.
Collapse
Affiliation(s)
- Soslan Shakhidzhanov
- Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, 117997 Moscow, Russia; (A.F.); (E.B.); (G.N.); (D.M.); (A.R.)
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Anna Filippova
- Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, 117997 Moscow, Russia; (A.F.); (E.B.); (G.N.); (D.M.); (A.R.)
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Elizaveta Bovt
- Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, 117997 Moscow, Russia; (A.F.); (E.B.); (G.N.); (D.M.); (A.R.)
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Andrew Gubkin
- Central Clinical Hospital No. 2 Named After N.A.Semashko “RZD-Medicine”, 121359 Moscow, Russia;
| | - Gennady Sukhikh
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I.Kulakov, 117997 Moscow, Russia;
| | - Sergey Tsarenko
- City Clinical Hospital No. 52 of Moscow Health Care Department, 123182 Moscow, Russia;
| | - Ilya Spiridonov
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Denis Protsenko
- Moscow Multiprofile Clinical Center “Kommunarka” of Moscow Healthcare Department, 142770 Moscow, Russia; (D.P.); (D.K.)
| | - Dmitriy Zateyshchikov
- City Clinical Hospital No. 51 of Moscow Health Care Department, 121309 Moscow, Russia;
| | - Elena Vasilieva
- City Clinical Hospital No. 23 of Moscow Health Care Department, 109004 Moscow, Russia; (E.V.); (A.K.); (O.D.)
| | - Anna Kalinskaya
- City Clinical Hospital No. 23 of Moscow Health Care Department, 109004 Moscow, Russia; (E.V.); (A.K.); (O.D.)
| | - Oleg Dukhin
- City Clinical Hospital No. 23 of Moscow Health Care Department, 109004 Moscow, Russia; (E.V.); (A.K.); (O.D.)
| | - Galina Novichkova
- Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, 117997 Moscow, Russia; (A.F.); (E.B.); (G.N.); (D.M.); (A.R.)
| | - Sergey Karamzin
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Ilya Serebriyskiy
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Elena Lipets
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Daria Kopnenkova
- Moscow Multiprofile Clinical Center “Kommunarka” of Moscow Healthcare Department, 142770 Moscow, Russia; (D.P.); (D.K.)
| | - Daria Morozova
- Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, 117997 Moscow, Russia; (A.F.); (E.B.); (G.N.); (D.M.); (A.R.)
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Evgeniya Melnikova
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Alexander Rumyantsev
- Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, 117997 Moscow, Russia; (A.F.); (E.B.); (G.N.); (D.M.); (A.R.)
| | - Fazoil Ataullakhanov
- Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, 117997 Moscow, Russia; (A.F.); (E.B.); (G.N.); (D.M.); (A.R.)
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| |
Collapse
|
12
|
He X, Zhang C, Ji J, Liu Y, Feng W, Luo L, Fan H, Guo L. Prognostic factors in hospitalized patients with COVID-19 pneumonia and effectiveness of prophylactic anticoagulant therapy: a single-center retrospective study. BMC Infect Dis 2025; 25:303. [PMID: 40033231 PMCID: PMC11877778 DOI: 10.1186/s12879-025-10666-3] [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: 11/05/2024] [Accepted: 02/18/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND COVID-19 pneumonia patients encounter the potential risk of venous thromboembolism (VTE) and mortality during hospitalization. This study aimed to analyzed risk factors of all-cause mortality in hospitalized patients with COVID-19 pneumonia, and investigated the effectiveness of prophylactic anticoagulation and hospital stays on the mortality in hospitalized patients with nonVTE. METHODS We retrospectively analyzed all COVID-19 pneumonia patients who were admitted to our medical center from December 2022 to January 2023. Clinical data and outcome events were collected from patients' electronic medical records. Cox regression was used to identify poor prognostic factors of COVID-19 pneumonia patients with VTE and nonVTE. Landmark analysis was conducted to identify time points of hospital stays between anticoagulation treatment and in-hospital survival outcomes in COVID-19 pneumonia patients with nonVTE. Binary logistic regression analysis was performed to investigate factors related to prolonged hospital stays. RESULTS Among 2,520 COVID-19 pneumonia patients, 1047 received prophylactic anticoagulation and 76 complicated with VTE during hospitalization. Survival curve analysis showed no statistically significant difference in mortality between COVID-19 pneumonia patients with VTE and nonVTE in prophylactic anticoagulant group (P = 0.63). Multivariate cox regression analysis revealed that male(HR = 1.398, 95%CI= [1.021,1.915]), BMI (HR = 0.935, 95%CI= [0.900,0.972]), lymphocytes (HR = 0.576, 95%CI= [0.409,0.809]), platelets (HR = 0.997, 95%CI= [0.995,0.999]), albumin (HR = 0.950, 95%CI= [0.926,0.975]), lactate dehydrogenase (HR = 1.001, 95%CI= [1.001,1.002]) were risk factors for mortality in COVID-19 pneumonia patients with nonVTE, while sCRP (HR = 1.010, 95%CI= [1.004,1.015]), anticoagulant therapy (HR = 0.247, 95%CI= [0.096,0.632]) were risk factors for mortality in COVID-19 pneumonia patients with VTE. Landmark analysis showed that for the hospital stays of 11 days, the difference in the impact of prophylactic anticoagulation on mortality was statistically significant in COVID-19 pneumonia patients with nonVTE (≤ 11days, P = 0.014; > 11days, P = 0.01). CVD (OR = 1.717, 95%CI= [1.248,2.363]), CRD (OR = 1.605, 95%CI= [1.133,2.274]), sCRP (OR = 1.003, 95%CI= [1.000,1.006]), Alb (OR = 0.959, 95%CI = [0.932,0.987]) and use of glucocorticoid (OR = 1.428, 95%CI= [1.057,1.930]) were independent factors associated with hospital stays > 11 days in anticoagulant group. CONCLUSIONS This study indicated that Male, lower BMI, peripheral blood lymphocytes, platelets, albumin and elevated lactate dehydrogenase were associated with poor hospitalisation outcomes in COVID-19 pneumonia patients with nonVTE. As for COVID-19 pneumonia patients with VTE, poor hospitalisation outcomes were associated with elevated sCRP levels and no given anticoagulant therapy. No significant difference in mortality between hospitalized COVID-19 pneumonia patients with VTE and nonVTE when receiving prophylactic anticoagulation. Prolonged hospital stays (> 11 days) may limit the effectiveness of prophylactic anticoagulation on lower in-hospital mortality for COVID-19 pneumonia patients with nonVTE.
Collapse
Affiliation(s)
- Xing He
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, China
| | - Chun Zhang
- Department of Pulmonary and Critical Care Medicine, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jiaqi Ji
- Department of Pulmonary and Critical Care Medicine, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yang Liu
- Department of Pulmonary and Critical Care Medicine, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Wanjie Feng
- Department of Internal Medicine, Wenjiang District Third People's Hospital of Chengdu, Chengdu, 611130, China
| | - Linjie Luo
- Department of Critical Care Medicine, Wenjiang District People's Hospital of Chengdu, Chengdu, Sichuan, China
| | - Hong Fan
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, China
| | - Lu Guo
- Department of Pulmonary and Critical Care Medicine, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.
| |
Collapse
|
13
|
Zhang H, Yang P, Gu X, Sun Y, Zhang R, Zhang D, Zhang J, Wang Y, Ma C, Liu M, Ma J, Li A, Wang Y, Ma X, Cui X, Wang Y, Liu Z, Wang W, Zheng Z, Li Y, Wu J, Wang Q, Cao B. Health outcomes one year after Omicron infection among 12,789 adults: a community-based cross-sectional study. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2025; 56:101507. [PMID: 40226780 PMCID: PMC11992576 DOI: 10.1016/j.lanwpc.2025.101507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/19/2024] [Accepted: 02/14/2025] [Indexed: 04/15/2025]
Abstract
Background Characterizing the paradigm and impact of long COVID is crucial for addressing this worldwide health challenge. This study aimed to investigate the prevalence of long COVID one year after primary Omicron infection and characterize differences in long-term health consequence between participants with persistent long COVID and those who fully recovered. Methods This a community-based cross-sectional study conducted from December 2023 to March 2024 at the China-Japan Friendship Hospital and 16 administrative districts in Beijing. 12,789 participants infected with Omicron between December 2022 and January 2023 were recruited through stratified multistage random sampling and included in the final analysis. Of them, 376 participants with persistent long COVID and 229 without long COVID were matched for further physical examinations. The primary outcome was the prevalence of long COVID one year after infection. Secondary outcomes included muscle strength, exercise capacity, health-related quality of life (HRQoL), mental health, work status, laboratory tests, and examinations. Findings Among 12,789 participants (media [IQR] age, 48.4 [37.3 to 61.4] years; 7817 females [61.1%]), 995 of them (7.8%) experienced long COVID within one year, with 651 (5.1%) having persistent symptoms. Fatigue (598/995 [60.1%]) and post-exertional malaise (367/995 [36.9%]) were the most common symptoms. Brain fog had the lowest resolution proportion as 4.2% within one year. The odds of long COVID increased with reinfections (odds ratios for one reinfection 2.592 [95% CI: 2.188 to 3.061]; two or more: 6.171 [3.227 to 11.557]; all p < 0.001). Participants with persistent long COVID had markedly lower muscle strength (upper-limb: 26.9 ± 12.4 vs. 29.1 ± 14.5 Kg; lower-limb: 40.0 [27.0 to 62.0] vs. 43.0 [28.0 to 59.0] s), worse exercise capacity and poorer HRQoL, and meaningful difference in laboratory tests results compared to those without long COVID. They also exhibited significantly higher proportions of abnormal lung function (FEV1 %pred<80%: 13.0% vs. 2.0%; DLco %pred<80%: 32.7% vs. 19.9%) and lung imaging abnormalities (23.5% vs. 13.6%). Interpretation The considerable health burden of long COVID and the progression of neurological symptoms following Omicron infection warrant close monitoring. Utilizing professional questionnaires and developing reliable diagnostic tools are necessary for improving diagnosis and treatment of long COVID. Funding This work was supported by Beijing Research Center for Respiratory Infectious Diseases (BJRID2024-012), Chinese Academy of Medical Sciences Innovation Fund for Medical Sciences (2022-I2M-CoV19-005/CIFMS 2021-I2M-1-048), the National Natural Science Foundation of China (82241056/82200114/82200009), the New Cornerstone Science Foundation.
Collapse
Affiliation(s)
- Hui Zhang
- National Center for Respiratory Medicine, Beijing, PR China
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, PR China
- National Clinical Research Center for Respiratory Diseases, Beijing, PR China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, PR China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, PR China
| | - Peng Yang
- Beijing Key Laboratory of Surveillance, Early Warning and Pathogen Research on Emerging Infectious Diseases, Beijing Center for Disease Prevention and Control, Beijing, PR China
- Beijing Research Center for Respiratory Infectious Diseases, Beijing, PR China
- School of Public Health, Capital Medical University, Beijing, PR China
| | - Xiaoying Gu
- National Center for Respiratory Medicine, Beijing, PR China
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, PR China
- National Clinical Research Center for Respiratory Diseases, Beijing, PR China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, PR China
- Department of Clinical Research and Data Management, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, PR China
| | - Ying Sun
- Beijing Key Laboratory of Surveillance, Early Warning and Pathogen Research on Emerging Infectious Diseases, Beijing Center for Disease Prevention and Control, Beijing, PR China
- Beijing Research Center for Respiratory Infectious Diseases, Beijing, PR China
- School of Public Health, Capital Medical University, Beijing, PR China
| | - Rongling Zhang
- National Center for Respiratory Medicine, Beijing, PR China
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, PR China
- National Clinical Research Center for Respiratory Diseases, Beijing, PR China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, PR China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, PR China
- Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| | - Daitao Zhang
- Beijing Key Laboratory of Surveillance, Early Warning and Pathogen Research on Emerging Infectious Diseases, Beijing Center for Disease Prevention and Control, Beijing, PR China
- Beijing Research Center for Respiratory Infectious Diseases, Beijing, PR China
- School of Public Health, Capital Medical University, Beijing, PR China
| | - Jiaojiao Zhang
- Beijing Key Laboratory of Surveillance, Early Warning and Pathogen Research on Emerging Infectious Diseases, Beijing Center for Disease Prevention and Control, Beijing, PR China
- Beijing Research Center for Respiratory Infectious Diseases, Beijing, PR China
- School of Public Health, Capital Medical University, Beijing, PR China
| | - Yeming Wang
- National Center for Respiratory Medicine, Beijing, PR China
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, PR China
- National Clinical Research Center for Respiratory Diseases, Beijing, PR China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, PR China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, PR China
| | - Chunna Ma
- Beijing Key Laboratory of Surveillance, Early Warning and Pathogen Research on Emerging Infectious Diseases, Beijing Center for Disease Prevention and Control, Beijing, PR China
- Beijing Research Center for Respiratory Infectious Diseases, Beijing, PR China
- School of Public Health, Capital Medical University, Beijing, PR China
| | - Min Liu
- Department of Radiology, China-Japan Friendship Hospital, Beijing, PR China
| | - Jiaxin Ma
- Beijing Key Laboratory of Surveillance, Early Warning and Pathogen Research on Emerging Infectious Diseases, Beijing Center for Disease Prevention and Control, Beijing, PR China
- Beijing Research Center for Respiratory Infectious Diseases, Beijing, PR China
- School of Public Health, Capital Medical University, Beijing, PR China
| | - Aili Li
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, PR China
| | - Yingying Wang
- Beijing Key Laboratory of Surveillance, Early Warning and Pathogen Research on Emerging Infectious Diseases, Beijing Center for Disease Prevention and Control, Beijing, PR China
- Beijing Research Center for Respiratory Infectious Diseases, Beijing, PR China
- School of Public Health, Capital Medical University, Beijing, PR China
| | - Xiao Ma
- Health Examination Center, China-Japan Friendship Hospital, Beijing, PR China
| | - Xiaojing Cui
- National Center for Respiratory Medicine, Beijing, PR China
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, PR China
- National Clinical Research Center for Respiratory Diseases, Beijing, PR China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, PR China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, PR China
| | - Yimin Wang
- National Center for Respiratory Medicine, Beijing, PR China
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, PR China
- National Clinical Research Center for Respiratory Diseases, Beijing, PR China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, PR China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, PR China
- Department of Pulmonary and Critical Care Medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei Province, PR China
| | - Zhibo Liu
- National Center for Respiratory Medicine, Beijing, PR China
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, PR China
- National Clinical Research Center for Respiratory Diseases, Beijing, PR China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, PR China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, PR China
| | - Wei Wang
- Department of Outpatients, China-Japan Friendship Hospital, Beijing, PR China
| | - Zhi Zheng
- Nursing Department, China-Japan Friendship Hospital, Beijing, PR China
| | - Yong Li
- National Center for Respiratory Medicine, Beijing, PR China
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, PR China
- National Clinical Research Center for Respiratory Diseases, Beijing, PR China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, PR China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, PR China
| | - Jin Wu
- Health Examination Center, China-Japan Friendship Hospital, Beijing, PR China
| | - Quanyi Wang
- Beijing Key Laboratory of Surveillance, Early Warning and Pathogen Research on Emerging Infectious Diseases, Beijing Center for Disease Prevention and Control, Beijing, PR China
- Beijing Research Center for Respiratory Infectious Diseases, Beijing, PR China
- School of Public Health, Capital Medical University, Beijing, PR China
| | - Bin Cao
- National Center for Respiratory Medicine, Beijing, PR China
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, PR China
- National Clinical Research Center for Respiratory Diseases, Beijing, PR China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, PR China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, PR China
- Beijing Key Laboratory of Surveillance, Early Warning and Pathogen Research on Emerging Infectious Diseases, Beijing Center for Disease Prevention and Control, Beijing, PR China
- Beijing Research Center for Respiratory Infectious Diseases, Beijing, PR China
- Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
- Tsinghua University-Peking University Joint Center for Life Sciences, Beijing, PR China
| |
Collapse
|
14
|
Niu ZS, Liu RX, Hu Y, Meng XR, Liu LH, Yang LT, Bai X, Chen MF, Pan DF. Complex causal relationships between genetic predictions of 731 immune cell phenotypes and novel coronavirus: A two-sample Mendelian randomization analysis. J Chin Med Assoc 2025; 88:231-237. [PMID: 39725715 DOI: 10.1097/jcma.0000000000001201] [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: 12/28/2024] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has had a significant impact on global health. While the virus primarily affects the respiratory system, the intricate interplay between immune cells and the virus remains poorly understood. This study investigates the causal relationship between 731 immune cell phenotypes and COVID-19 using Mendelian randomization (MR) analysis. METHODS A bidirectional two-sample MR analysis was conducted using genetic variants strongly associated with immune cell phenotypes as instrumental variables. Data for 731 immune cell phenotypes were sourced from the Genome-Wide Association Study (GWAS) catalog, while data for COVID-19 susceptibility were obtained from the OPEN GWAS database. Five MR methods (inverse variance weighted [IVW], MR-Egger, weighted median, simple mode, and weighted mode) were used to estimate causal effects, with IVW as the primary analysis method. RESULTS The study identified 57 immune cell phenotypes causally associated with COVID-19 risk across two independent GWAS datasets. Five immune cell phenotypes were consistently associated with COVID-19 risk across both datasets: CD3- lymphocyte %lymphocyte (protective), CD27 on CD20- (protective), CD20 on IgD+ CD38- unsw mem (increased risk), CD27 on IgD- CD38- (increased risk), and CD19 on B cell (increased risk). Sensitivity analyses confirmed the robustness of the findings. CONCLUSION This study provides compelling evidence for a causal relationship between specific immune cell phenotypes and COVID-19 risk. These findings highlight the potential for targeting these immune cell phenotypes as novel therapeutic targets for COVID-19 treatment and prevention.
Collapse
Affiliation(s)
- Ze-Su Niu
- Department of Emergency Medicine, The Third Clinical Medical College of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Ru-Xin Liu
- Department of Emergency Medicine, The Third Clinical Medical College of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yi Hu
- Department of Emergency Medicine, The Third Clinical Medical College of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Xiao-Rui Meng
- Department of Emergency Medicine, The Third Clinical Medical College of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Li-Hong Liu
- Department of Emergency Medicine, The Third Clinical Medical College of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Li-Ting Yang
- Department of Emergency Medicine, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China
| | - Xue Bai
- Department of Emergency Medicine, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China
| | - Meng-Fei Chen
- Department of Emergency Medicine, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China
| | - Dong-Feng Pan
- Department of Emergency Medicine, The Third Clinical Medical College of Ningxia Medical University, Yinchuan, Ningxia, China
- Department of Emergency Medicine, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China
| |
Collapse
|
15
|
Popazu C, Romila A, Petrea M, Grosu RM, Lescai AM, Vlad AL, Oprea VD, Baltă AAȘ. Overview of Inflammatory and Coagulation Markers in Elderly Patients with COVID-19: Retrospective Analysis of Laboratory Results. Life (Basel) 2025; 15:370. [PMID: 40141715 PMCID: PMC11943672 DOI: 10.3390/life15030370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Elderly patients with COVID-19 often exhibit a complex interplay between hypercoagulability and coagulopathy, key factors in determining the risk of severe complications and mortality. This study aimed to analyze coagulation and inflammatory markers to identify critical predictors of adverse outcomes in this vulnerable population. Material and Methods: The retrospective study was conducted on a sample of 1429 elderly patients (≥60 years) diagnosed with COVID-19, hospitalized in "Sf. Ap. Andrei" St. Apostle Andrew's County Emergency Hospital in various wards between March 2020 and August 2022. Data were collected from medical records and included inflammatory markers (C-reactive protein, procalcitonin, ESR) and coagulation markers (prothrombin time, INR, fibrinogen, D-dimer). The SPSS 2.0 statistical software was used to conduct the study. Results:Coagulation markers: Prothrombin activity averaged 74.22%, below normal levels, indicating a heightened bleeding risk, while fibrinogen levels were significantly elevated (mean: 531.69 mg/dL), reflecting hypercoagulability. Prolonged prothrombin time (mean: 17.28 s) and elevated INR (International normalized ratio) (mean: 1.51) were associated with increased mortality, emphasizing their role in risk stratification. Elevated D-dimer levels (mean: 2.75 mg/L) further highlighted thromboembolic risks. Inflammatory markers: C-reactive protein (CRP) and erythrocyte sedimentation rate (ESR) showed marked elevations (mean CRP: 92.09 mg/L, mean ESR: 58.47 mm/h), correlating with heightened systemic inflammation and poor outcomes. Bacterial infections: Elevated procalcitonin (mean: 1.98 ng/mL) suggested secondary bacterial infections, particularly in mechanically ventilated patients, significantly worsening prognosis. Conclusions: The duality of hypercoagulability and coagulopathy in elderly COVID-19 patients underscores the importance of consistently monitoring coagulation markers such as prothrombin time, INR, D-dimer, and fibrinogen. Simultaneously, elevated inflammatory markers and secondary bacterial infections require prompt therapeutic interventions. This study highlights the critical need for personalized management strategies to mitigate complications and reduce mortality in this high-risk population.
Collapse
Affiliation(s)
- Corina Popazu
- Clinical-Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, Str. Domnească 35, 800201 Galaţi, Romania; (C.P.); (A.L.V.); (A.A.Ș.B.)
| | - Aurelia Romila
- Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, Str. Domnească 35, 800201 Galaţi, Romania; (A.R.); (V.D.O.)
| | - Marius Petrea
- Pre-Clinical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, Str. Domnească 35, 800201 Galaţi, Romania; (M.P.); (R.M.G.)
| | - Robert Marius Grosu
- Pre-Clinical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, Str. Domnească 35, 800201 Galaţi, Romania; (M.P.); (R.M.G.)
| | - Alina-Maria Lescai
- Clinical-Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, Str. Domnească 35, 800201 Galaţi, Romania; (C.P.); (A.L.V.); (A.A.Ș.B.)
| | - Adriana Liliana Vlad
- Clinical-Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, Str. Domnească 35, 800201 Galaţi, Romania; (C.P.); (A.L.V.); (A.A.Ș.B.)
| | - Violeta Diana Oprea
- Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, Str. Domnească 35, 800201 Galaţi, Romania; (A.R.); (V.D.O.)
| | - Alexia Anastasia Ștefania Baltă
- Clinical-Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, Str. Domnească 35, 800201 Galaţi, Romania; (C.P.); (A.L.V.); (A.A.Ș.B.)
| |
Collapse
|
16
|
Dinoi G, Togo MV, Guida P, Deruvo C, Samarelli F, Imbrici P, Nicolotti O, De Luca A, Mastroianni F, Liantonio A, Altomare CD. Retrospective Clinical Investigation into the Association Between Abnormal Blood Clotting, Oral Anticoagulant Therapy, and Medium-Term Mortality in a Cohort of COVID-19 Patients. Biomedicines 2025; 13:535. [PMID: 40149514 PMCID: PMC11940371 DOI: 10.3390/biomedicines13030535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/14/2025] [Accepted: 02/16/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: People affected by COVID-19 are exposed to abnormal clotting and endothelial dysfunction, which may trigger thromboembolic events. This study aimed at retrospectively investigating whether oral anticoagulant therapy (OAT), encompassing either direct oral anticoagulants (DOACs), mainly apixaban, or the vitamin K antagonist (VKA) warfarin, could have impacted medium-term mortality in a cohort of SARS-CoV-2 patients. Methods: Among 1238 COVID-19 patients, hospitalized from 17 March 2020 to 15 June 2021, 247 survivors and 247 deceased within 90 days from hospitalization were matched 1:1 based on age, sex, and intensive care unit (ICU) admission within three days. Conditional logistic regression was used to estimate associations by means of odds ratio (OR) with a 95% confidence interval (CI). Results: A univariate regression analysis suggested that OAT, no differently from subcutaneous low-molecular-weight heparins (LMWHs) during hospitalization, has no significant impact (p value > 0.05) on medium-term mortality. A multivariate analysis, limited to baseline variables (i.e., comorbidities and pharmacotherapies at hospital admission) showing significant association (p < 0.05) to mortality in a univariate analysis, revealed that, compared to patients living at 90 days from hospitalization, deceased patients had cancer histories (OR 1.75, CI 1.06-2.90, p = 0.029) or suffered from asthma (OR 2.25, CI 1.13-4.47, p = 0.021). In contrast, heart failure (HF), atrial fibrillation (AF), arteriopathy, chronic obstructive pulmonary disease (COPD), and kidney failure (KF), which, in a univariate analysis, were found to be associated with the endpoint (p < 0.05), lost significance in a multivariate analysis. Therapy at admission with aldosterone antagonists also appeared to be associated with medium-term mortality (OR 2.49, CI 1.52-4.08, p < 0.001); whereas, vitamin D supplementation during hospitalization appeared to be beneficial. Although not conclusive, a search into the Eudravigilance database, combined with consulting a digital predictive platform (PLATO, polypharmacology platform prediction), suggested potential off-target activities, which might contribute to increasing the severity of SARS-CoV-2 infection. Conclusions: This retrospective clinical study furnished evidences of the impact of OAT, comorbidities and other pharmacological treatments on COVID-19 clinical course.
Collapse
Affiliation(s)
- Giorgia Dinoi
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| | - Maria Vittoria Togo
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| | - Pietro Guida
- Department of Internal Medicine, F. Miulli General Hospital, 70021 Bari, Italy; (P.G.); (F.M.)
| | - Caterina Deruvo
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| | - Francesco Samarelli
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| | - Paola Imbrici
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| | - Orazio Nicolotti
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| | - Annamaria De Luca
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| | - Franco Mastroianni
- Department of Internal Medicine, F. Miulli General Hospital, 70021 Bari, Italy; (P.G.); (F.M.)
| | - Antonella Liantonio
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| | - Cosimo Damiano Altomare
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| |
Collapse
|
17
|
Ayyoub S, Dhillon NK, Tura-Ceide O. Genetics of Long COVID: Exploring the Molecular Drivers of Persistent Pulmonary Vascular Disease Symptoms. Infect Dis Rep 2025; 17:15. [PMID: 39997467 PMCID: PMC11855385 DOI: 10.3390/idr17010015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/24/2025] [Accepted: 02/08/2025] [Indexed: 02/26/2025] Open
Abstract
Background/ Objectives: Long COVID or post-acute sequelae of SARS-CoV-2 infection (PASC) are symptoms that manifest despite passing the acute infection phase. These manifestations encompass a wide range of symptoms, the most common being fatigue, shortness of breath, and cognitive dysfunction. Genetic predisposition is clearly involved in the susceptibility of individuals to developing these persistent symptoms and the variation in the severity and forms. This review summarizes the role of genetic factors and gene polymorphisms in the development of major pulmonary vascular disorders associated with long COVID. Methods: A comprehensive review of current literature was conducted to examine the genetic contributions to pulmonary complications following SARS-CoV-2 infection. Studies investigating genetic polymorphisms linked to pulmonary hypertension, pulmonary thromboembolism, and pulmonary vascular endothelialitis were reviewed and summarized. Results: Findings show that specific genetic variants contribute to increased susceptibility to pulmonary vascular complications in long COVID patients. Variants associated with endothelial dysfunction, coagulation pathways, and inflammatory responses have been implicated in the development of pulmonary hypertension and thromboembolic events. Genetic predispositions influencing vascular integrity and immune responses appear to influence disease severity and progression. Conclusions: Understanding these mechanisms and genetic predispositions could pave the way for targeted therapeutic interventions to alleviate the burden on patients experiencing long COVID.
Collapse
Affiliation(s)
- Sana Ayyoub
- Department of Medical Sciences, Faculty of Medicine, University of Girona, 17004 Girona, Spain;
| | - Navneet Kaur Dhillon
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Kansas Medical Center, Mail Stop 3007, 3901 Rainbow Blvd, Kansas City, KS 66160, USA;
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Olga Tura-Ceide
- Translational Research Group on Cardiovascular Respiratory Diseases (CAREs), Girona Biomedical Research Institute (IDIBGI-CERCA), Martí i Julià, Hospital Park Building M2, 17190 Salt, Spain
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029 Madrid, Spain
| |
Collapse
|
18
|
van der Mescht MA, Steel HC, Anderson R, Rossouw TM. Vascular endothelial growth factor A: friend or foe in the pathogenesis of HIV and SARS-CoV-2 infections? Front Cell Infect Microbiol 2025; 14:1458195. [PMID: 40008234 PMCID: PMC11850333 DOI: 10.3389/fcimb.2024.1458195] [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: 07/02/2024] [Accepted: 10/28/2024] [Indexed: 02/27/2025] Open
Abstract
This review article discusses the role of vascular endothelial growth factor A (VEGF-A) in the pathogenesis of SARS-CoV-2 and HIV infection, both conditions being renowned for their impact on the vascular endothelium. The processes involved in vascular homeostasis and angiogenesis are reviewed briefly before exploring the interplay between hypoxia, VEGF-A, neuropilin-1 (NRP-1), and inflammatory pathways. We then focus on SARS-CoV-2 infection and show how the binding of the viral pathogen to the angiotensin-converting enzyme 2 receptor, as well as to NRP-1, leads to elevated levels of VEGF-A and consequences such as coagulation, vascular dysfunction, and inflammation. HIV infection augments angiogenesis via several mechanisms, most prominently, by the trans-activator of transcription (tat) protein mimicking VEGF-A by binding to its receptor, VEGFR-2, as well as upregulation of NRP-1, which enhances the interaction between VEGF-A and VEGFR-2. We propose that the elevated levels of VEGF-A observed during HIV/SARS-CoV-2 co-infection originate predominantly from activated immune cells due to the upregulation of HIF-1α by damaged endothelial cells. In this context, a few clinical trials have described a diminished requirement for oxygen therapy during anti-VEGF treatment of SARS-CoV-2 infection. The currently available anti-VEGF therapy strategies target the binding of VEGF-A to both VEGFR-1 and VEGFR-2. The blocking of both receptors could, however, lead to a negative outcome, inhibiting not only pathological, but also physiological angiogenesis. Based on the examination of published studies, this review suggests that treatment targeting selective inhibition of VEGFR-1 may be beneficial in the context of SARS-CoV-2 infection.
Collapse
Affiliation(s)
| | | | | | - Theresa M. Rossouw
- Department of Immunology, Faculty of Health Sciences, University of
Pretoria, Pretoria, South Africa
| |
Collapse
|
19
|
Arnold JR, Yeo JL, Budgeon CA, Shergill S, England R, Shiwani H, Artico J, Moon JC, Gorecka M, Roditi G, Morrow A, Mangion K, Shanmuganathan M, Miller CA, Chiribiri A, Alzahir M, Ramirez S, Lin A, Swoboda PP, McDiarmid AK, Sykes R, Singh T, Bucciarelli-Ducci C, Dawson D, Fontana M, Manisty C, Treibel TA, Levelt E, Young R, McConnachie A, Neubauer S, Piechnik SK, Davies RH, Ferreira VM, Dweck MR, Berry C, McCann GP, Greenwood JP. Myocardial ischaemia following COVID-19: a cardiovascular magnetic resonance study. Int J Cardiovasc Imaging 2025; 41:247-256. [PMID: 39738791 PMCID: PMC11811239 DOI: 10.1007/s10554-024-03304-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 11/27/2024] [Indexed: 01/02/2025]
Abstract
The pathophysiology of myocardial injury following COVID-19 remains uncertain. COVID-HEART was a prospective, multicentre study utilising cardiovascular magnetic resonance (CMR) to characterise COVID-related myocardial injury. In this pre-specified analysis, the objectives were to examine (1) the frequency of myocardial ischaemia following COVID-19, and (2) the association between ischaemia and myocardial injury. We studied 59 patients hospitalised with COVID-19 and elevated serum troponin (COVID + /troponin + , age 61 ± 11 years) and 37 control subjects without COVID-19 or elevated troponin and similar by age and cardiovascular comorbidities (COVID -/comorbidity + , 64 ± 10 years). Subjects underwent multi-parametric CMR (comprising assessment of ventricular volumes, stress perfusion, T1/T2 mapping and scar). The primary endpoint was the frequency of inducible myocardial ischaemia. Inducible ischaemia was evident in 11 (19%) COVID + /troponin + patients and in 8 (22%) control subjects (p = 0.72). In COVID + /troponin + patients with ischaemia, epicardial coronary disease pattern ischaemia was present in eight patients and microvascular disease pattern, in three patients. There was no significant difference in the frequency of inducible ischaemia in COVID + /troponin + patients with previous myocardial infarction and/or revascularisation compared to those without (2/12 [17%] vs. 9/47 [19%] respectively, p = 0.84), or in those with and without scar (7/27 [26%] vs. 4/32 [13%] respectively, p = 0.19). Myocardial ischaemia was present in ~ 20% of patients recently hospitalised with COVID-19 and with elevated cardiac troponin, but this was not different to matched comorbid controls. This finding coupled with the lack of an association between ischaemia and myocardial scar suggests that coronary artery abnormalities are unlikely to be the predominant mechanism underlying COVID-19 induced myocardial injury.
Collapse
Affiliation(s)
- J Ranjit Arnold
- University of Leicester and The NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK.
| | - Jian L Yeo
- University of Leicester and The NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Charley A Budgeon
- University of Leicester and The NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
- Cardiovascular Epidemiology Research Centre, School of Population and Global Health, University of Western Australia, Perth, Australia
| | - Simran Shergill
- University of Leicester and The NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Rachel England
- University of Leicester and The NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Hunain Shiwani
- Institute of Cardiovascular Science, University College London, London, UK
| | - Jessica Artico
- Institute of Cardiovascular Science, University College London, London, UK
| | - James C Moon
- Institute of Cardiovascular Science, University College London, London, UK
| | - Miroslawa Gorecka
- Institute of Cardiovascular and Metabolic Medicine, University of Leeds, and Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Giles Roditi
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Andrew Morrow
- Institute of Cardiovascular and Medical Sciences and British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Kenneth Mangion
- Institute of Cardiovascular and Medical Sciences and British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Mayooran Shanmuganathan
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford Centre for Clinical Magnetic Resonance Research, Oxford, UK
- British Heart Foundation Centre of Research Excellence, Oxford, UK
- Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Christopher A Miller
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Amedeo Chiribiri
- School of Biomedical Engineering and Imaging Sciences, King's College London, BHF Centre of Excellence and The NIHR Biomedical Research Centre at Guy's and St. Thomas' NHS Foundation Trust, The Rayne Institute, St. Thomas' Hospital, London, UK
| | - Mohammed Alzahir
- Institute of Cardiovascular Science, University College London, London, UK
| | - Sara Ramirez
- Institute of Cardiovascular Science, University College London, London, UK
| | - Andrew Lin
- Institute of Cardiovascular Science, University College London, London, UK
| | - Peter P Swoboda
- Institute of Cardiovascular and Metabolic Medicine, University of Leeds, and Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Adam K McDiarmid
- Adult Congenital and Paediatric Heart Unit, Freeman Hospital, Newcastle Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Robert Sykes
- Institute of Cardiovascular and Medical Sciences and British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Trisha Singh
- University of Edinburgh and British Heart Foundation Centre for Cardiovascular Science, Edinburgh, UK
| | - Chiara Bucciarelli-Ducci
- School of Biomedical Engineering and Imaging Sciences, King's College London, BHF Centre of Excellence and The NIHR Biomedical Research Centre at Guy's and St. Thomas' NHS Foundation Trust, The Rayne Institute, St. Thomas' Hospital, London, UK
- Royal Brompton and Harefield Hospitals, London, UK
- Guys' and St Thomas NHS Trust, London, UK
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Trust, Bristol, UK
| | - Dana Dawson
- Department of Cardiology, Aberdeen Cardiovascular and Diabetes Centre, Aberdeen Royal Infirmary and University of Aberdeen, Aberdeen, UK
| | - Marianna Fontana
- Division of Medicine, Royal Free Hospital, University College London, London, UK
| | - Charlotte Manisty
- Institute of Cardiovascular Science, University College London, London, UK
| | - Thomas A Treibel
- Institute of Cardiovascular Science, University College London, London, UK
| | - Eylem Levelt
- Institute of Cardiovascular and Metabolic Medicine, University of Leeds, and Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Robin Young
- Robertson Centre for Biostatistics, Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Alex McConnachie
- Robertson Centre for Biostatistics, Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford Centre for Clinical Magnetic Resonance Research, Oxford, UK
| | - Stefan K Piechnik
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford Centre for Clinical Magnetic Resonance Research, Oxford, UK
| | - Rhodri H Davies
- Institute of Cardiovascular Science, University College London, London, UK
| | - Vanessa M Ferreira
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford Centre for Clinical Magnetic Resonance Research, Oxford, UK
| | - Marc R Dweck
- University of Edinburgh and British Heart Foundation Centre for Cardiovascular Science, Edinburgh, UK
| | - Colin Berry
- Institute of Cardiovascular and Medical Sciences and British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Gerry P McCann
- University of Leicester and The NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - John P Greenwood
- Institute of Cardiovascular and Metabolic Medicine, University of Leeds, and Leeds Teaching Hospitals NHS Trust, Leeds, UK
- Baker Heart and Diabetes Institute, Melbourne, Australia
| |
Collapse
|
20
|
Lai J, Yu S, Li X, Wei Q, Qin J. METTL14/IGF2BP2-MEDIATED M6A MODIFICATION OF STEAP1 AGGRAVATES ACUTE LUNG INJURY INDUCED BY SEPSIS. Shock 2025; 63:217-225. [PMID: 39193903 DOI: 10.1097/shk.0000000000002456] [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: 08/29/2024]
Abstract
ABSTRACT Background: Acute lung injury (ALI) is a severe complication of sepsis, characterized by inflammation, edema, and injury to alveolar cells, leading to high mortality rates. Septic ALI is a complex disease involving multiple factors and signaling pathways. STEAP family member 1 (STEAP1) has been reported to be upregulated in a sepsis-induced ALI model. However, the role of STEAP1 in the regulation of septic ALI is not yet fully understood. Methods: The study stimulated human pulmonary microvascular endothelial cells (HPMECs) using lipopolysaccharides (LPS) to establish an in vitro ALI model. The study used quantitative real-time polymerase chain reaction to measure mRNA expression, and western blotting assay or immunohistochemistry assay to analyze protein expression. Cell Counting Kit-8 assay was performed to assess cell viability. Flow cytometry was conducted to analyze cell apoptosis. Tube formation assay was used to analyze the tube formation rate of human umbilical vein endothelial cells. Enzyme-linked immunosorbent assays were used to measure the levels of interleukin 1beta and tumor necrosis factor alpha. The levels of Fe 2+ and reactive oxygen species were determined using colorimetric and fluorometric assays, respectively. The glutathione level was also determined using a colorimetric assay. m6A RNA immunoprecipitation assay, dual-luciferase reporter assay, and RNA immunoprecipitation assay were performed to identify the association of STEAP1 with methyltransferase 14, N6-adenosine-methyltransferase noncatalytic subunit (METTL14) and insulin like growth factor 2 mRNA binding protein 2 (IGF2BP2). The transcript half-life of STEAP1 was analyzed by actinomycin D assay. Finally, a rat model of polymicrobial sepsis was established to analyze the effects of STEAP1 knockdown on lung injury in vivo . Results: We found that the mRNA expression levels of STEAP1 and METTL14 were upregulated in the blood of ALI patients induced by sepsis compared to healthy volunteers. LPS treatment increased the protein levels of STEAP1 and METTL14 in HPMECs. STEAP1 depletion attenuated LPS-induced promoting effects on HPMECs' apoptosis, inflammatory response, and ferroptosis, as well as LPS-induced inhibitory effect on tube formation. We also found that METTL14 and IGF2BP2 stabilized STEAP1 mRNA expression through the m6A methylation modification process. Moreover, METTL14 silencing attenuated LPS-induced effects by decreasing STEAP1 expression in HPMECs, and STEAP1 silencing ameliorated cecal ligation and puncture-induced lung injury of mice. Conclusion: METTL14/IGF2BP2-mediated m6A modification of STEAP1 aggravated ALI induced by sepsis. These findings suggest potential therapeutic targets for the treatment of this disease.
Collapse
Affiliation(s)
- Junhua Lai
- Intensive Care Unit, Liuzhou Worker's Hospital, Liuzhou City, Guangxi, China
| | | | | | | | | |
Collapse
|
21
|
Xiang Z, Hu J, Bu S, Ding J, Chen X, Li Z. Machine learning based prediction models for the prognosis of COVID-19 patients with DKA. Sci Rep 2025; 15:2633. [PMID: 39837852 PMCID: PMC11751332 DOI: 10.1038/s41598-025-85357-9] [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: 06/17/2024] [Accepted: 01/02/2025] [Indexed: 01/23/2025] Open
Abstract
Patients with Diabetic ketoacidosis (DKA) have increased critical illness and mortality during coronavirus diseases 2019 (COVID-19). The aim of our study was to develop a predictive model for the occurrence of critical illness and mortality in COVID-19 patients with DKA utilizing machine learning. Blood samples and clinical data from 242 COVID-19 patients with DKA collected from December 2022 to January 2023 at Second Xiangya Hospital. Patients were categorized into non-death (n = 202) and death (n = 38) groups, and non-severe (n = 146) and severe (n = 96) groups. We developed five machine learning-based prediction models-Extreme Gradient Boosting (XGB), Logistic Regression (LR), Random Forest (RF), Support Vector Machine (SVM), and Multilayer Perceptron (MLP)-to evaluate the prognosis of COVID-19 patients with DKA. We employed 5-fold cross-validation for model evaluation and used the Shapley Additive Explanations (SHAP) algorithm for result interpretation to ensure reliability. The LR model demonstrated the highest accuracy (AUC = 0.933) in predicting mortality. Additionally, the LR model excelled (AUC = 0.898) in predicting progression to severe disease. This study developed a machine learning-based predictive model for the progression to severe disease or death in COVID-19 patients with DKA, which can serve as a valuable tool to guide clinical treatment decisions.
Collapse
Affiliation(s)
- Zhongyuan Xiang
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Jingyi Hu
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Shengfang Bu
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jin Ding
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Department of Metabolism and Endocrinology, Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xi Chen
- Information Network Center of Xiangya Second Hospital, Central South University, Changsha, China
| | - Ziyang Li
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
22
|
Murad D, Paracha RZ, Nisar M. Unravelling the impact of SARS-CoV-2 on hemostatic and complement systems: a systems immunology perspective. Front Immunol 2025; 15:1457324. [PMID: 39885991 PMCID: PMC11781117 DOI: 10.3389/fimmu.2024.1457324] [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: 06/30/2024] [Accepted: 12/17/2024] [Indexed: 02/01/2025] Open
Abstract
The hemostatic system prevents and stops bleeding, maintaining circulatory integrity after injury. It directly interacts with the complement system, which is key to innate immunity. In coronavirus disease 2019 (COVID-19), dysregulation of the hemostatic and complement systems has been associated with several complications. To understand the essential balance between activation and regulation of these systems, a quantitative systems immunology model can be established. The dynamics of the components are examined under three distinct conditions: the disease state representing symptomatic COVID-19 state, an intervened disease state marked by reduced levels of regulators, and drug interventions including heparin, tranexamic acid, avdoralimab, garadacimab, and tocilizumab. Simulation results highlight key components affected, including thrombin, tissue plasminogen activator, plasmin, fibrin degradation products, interleukin 6 (IL-6), the IL-6 and IL-6R complex, and the terminal complement complex (C5b-9). We explored that the decreased levels of complement factor H and C1-inhibitor significantly elevate these components, whereas tissue factor pathway inhibitor and alpha-2-macroglobulin have more modest effects. Furthermore, our analysis reveals that drug interventions have a restorative impact on these factors. Notably, targeting thrombin and plasmin in the early stages of thrombosis and fibrinolysis can improve the overall system. Additionally, the regulation of C5b-9 could aid in lysing the virus and/or infected cells. In conclusion, this study explains the regulatory mechanisms of the hemostatic and complement systems and illustrates how the biopathway machinery sustains the balance between activation and inhibition. The knowledge that we have acquired could contribute to designing therapies that target the hemostatic and complement systems.
Collapse
Affiliation(s)
| | - Rehan Zafar Paracha
- School of Interdisciplinary Engineering and Sciences (SINES), Department of Sciences,
National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | | |
Collapse
|
23
|
Lee RH, Wang S, Akerman M, Joseph D. Role of peak D-dimer in predicting mortality and venous thromboembolism in COVID-19 patients. Sci Prog 2025; 108:368504241247982. [PMID: 40012497 PMCID: PMC11866355 DOI: 10.1177/00368504241247982] [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] [Indexed: 02/28/2025]
Abstract
Covid 19 patients often present with elevated D-dimer levels. The purpose of this study is to evaluate the role of D-Dimer levels in Covid 19 patients to predict mortality and venous thromboembolism (VTE) events. This is a retrospective chart review study from 1 April 2020 to 30 June 2020, during the peak Covid pandemic. A total of 350 patients were enrolled in this study; 69 (19.7%) patients died; 12 (3.4%) had a deep venous thrombosis; and 8 (2.3%) had a pulmonary embolism outcome. Peak D-dimer levels were collected with median levels of 765 ng/ml (266, 3135). Patients with VTE outcomes had significantly higher levels of peak D-dimers than patients in the non-VTE group (4876 vs 680, p < 0.0001). Patients who died had higher peak D-dimer levels than those who survived (4690 vs 501, p < 0.0001). The optimal cutoff point in peak D-dimer in predicting VTE events was 1437, yielding a sensitivity of 84.2% and a specificity of 65.0%. The optimal cutoff point in peak D-dimer in predicting mortality was 2004, yielding a sensitivity of 71.0% and a specificity of 77.9%. This study suggests that D-dimer levels can be elevated in Covid 19 hospitalized patients and can serve as indicators for mortality and VTE events.
Collapse
Affiliation(s)
- Rachel H Lee
- Department of Pharmacy, NYU Langone Hospital–Long Island, Mineola, NY, USA
| | - Shan Wang
- Department of Pharmacy, NYU Langone Hospital–Long Island, Mineola, NY, USA
- NYU Long Island School of Medicine, Mineola, NY, USA
| | - Meredith Akerman
- NYU Long Island School of Medicine, Mineola, NY, USA
- Department of Foundations of Medicine, NYU Langone Hospital– Long Island, Mineola, NY, USA
| | - D’Andrea Joseph
- NYU Long Island School of Medicine, Mineola, NY, USA
- Department of Surgery, NYU Langone Hospital– Long Island, Mineola, NY, USA
| |
Collapse
|
24
|
Eslamifar Z, Behzadifard M, Zare E. Investigation of homocysteine, D-dimer and platelet count levels as potential predictors of thrombosis risk in COVID-19 patients. Mol Cell Biochem 2025; 480:439-444. [PMID: 38502382 DOI: 10.1007/s11010-024-04967-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 02/13/2024] [Indexed: 03/21/2024]
Abstract
Thrombosis plays an important role in induction of Coronavirus disease 19 (COVID-19) complications including heart attack and stroke. Reliable biomarkers are needed to predict thrombosis risk for better management and improve patient outcomes. This study aimed to investigate the relationship between homocysteine, a thrombosis-related biomarker, and other thrombosis-related parameters, such as D-dimer and platelet count with disease outcome in COVID-19 patients. This case-control study including 50 intensive care unit hospitalized patients with Covid-19 with a positive RT-PCR test for SARS-CoV-2 infection and 50 healthy individuals as a control group was conducted. Both groups were matched for age and body mass index (BMI) and had no history of underlying diseases such as cardiovascular, liver, kidney or smoking. Blood samples were collected from both groups to measure serum homocysteine, platelet count and D-dimer levels. Data were analyzed using GraphPad Prism version 8.3 software. The study found no statistically significant difference in homocysteine levels between COVID-19 patients and the control group. However, D-dimer levels were significantly higher in the patient group. Platelet count analysis revealed a significant difference between patients who died and those who were discharged from the hospital (P < 0.05). Despite previous studies suggesting a link between homocysteine and thrombosis, this study found no significant difference in homocysteine levels between COVID-19 patients and the control group. The significantly elevated D-dimer levels in the death group patient suggest that D-dimer and thrombocytopenia may be more reliable predictors of thrombosis and worse outcome in COVID-19 patients without underlying diseases.
Collapse
Affiliation(s)
- Zahra Eslamifar
- Department of Medical Laboratory Sciences, School of Paramedical Sciences, Dezful University of Medical Sciences, Dezful, Iran
| | - Mahin Behzadifard
- Department of Medical Laboratory Sciences, School of Paramedical Sciences, Dezful University of Medical Sciences, Dezful, Iran.
| | - Ehsan Zare
- Student Research Committee, Dezful University of Medical Sciences, Dezful, Iran
| |
Collapse
|
25
|
Becker RC, Tantry US, Khan M, Gurbel PA. The COVID-19 thrombus: distinguishing pathological, mechanistic, and phenotypic features and management. J Thromb Thrombolysis 2025; 58:15-49. [PMID: 39179952 PMCID: PMC11762605 DOI: 10.1007/s11239-024-03028-4] [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] [Accepted: 08/01/2024] [Indexed: 08/26/2024]
Abstract
A heightened risk for thrombosis is a hallmark of COVID-19. Expansive clinical experience and medical literature have characterized small (micro) and large (macro) vessel involvement of the venous and arterial circulatory systems. Most events occur in patients with serious or critical illness in the hyperacute (first 1-2 weeks) or acute phases (2-4 weeks) of SARS-CoV-2 infection. However, thrombosis involving the venous, arterial, and microcirculatory systems has been reported in the subacute (4-8 weeks), convalescent (> 8-12 weeks) and chronic phases (> 12 weeks) among patients with mild-to-moderate illness. The purpose of the current focused review is to highlight the distinguishing clinical features, pathological components, and potential mechanisms of venous, arterial, and microvascular thrombosis in patients with COVID-19. The overarching objective is to better understand the proclivity for thrombosis, laying a solid foundation for screening and surveillance modalities, preventive strategies, and optimal patient management.
Collapse
Affiliation(s)
- Richard C Becker
- Cardiovascular Center, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267, USA.
| | - Udaya S Tantry
- Sinai Center for Thrombosis Research and Drug Development, Baltimore, USA
| | - Muhammad Khan
- Division of General Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, USA
| | - Paul A Gurbel
- Sinai Center for Thrombosis Research and Drug Development, Baltimore, USA
| |
Collapse
|
26
|
Héja M, Fekete I, Márton S, Horváth L, Fekete K. Impact of COVID-19 pandemic on acute stroke care in a tertiary stroke centre. Sci Rep 2024; 14:31408. [PMID: 39733029 PMCID: PMC11682369 DOI: 10.1038/s41598-024-83016-z] [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: 06/14/2024] [Accepted: 12/10/2024] [Indexed: 12/30/2024] Open
Abstract
The aim of this study was to evaluate how COVID-19 affected acute stroke care and outcome in patients with acute ischemic or hemorrhagic stroke. We performed a retrospective analysis on patients who were admitted with acute ischemic (AIS) or hemorrhagic (ICH) stroke from September 2020 to May 2021 with and without COVID-19. We recorded demographic and clinical data, imaging parameters, functional outcome and mortality at one year. Beside descriptive statistics we performed χ2-probe, Mann-Witney U-test, Student t-probe and multivariate testing. We found a 29%-reduction in the number of AIS cases during the pandemic. The number of the large vessel occlusions /LVOs/ (N = 83, 41.7%), from them 37 (17.7%) had mechanical thrombectomy (MT), was higher than before the COVID-19 period (p = 0.02 and p = 0.001, respectively). From all patients needing acute revascularization therapy (N = 137) 118 patients received it, among them 20 (16.9%) had COVID-19. Those positive for COVID-19 were more likely to have a higher median NIHSS score at baseline and at 24 h (p = 0.02 and p = 0.03, respectively). They also had a lower rate of favourable outcome at discharge (15% vs. 41.8%; p = 0.024) and at three months (25% vs. 52%, p = 0.02), longer median hospitalization (p < 0.0001), and a higher mortality rate (52% vs. 25%; p = 0.03). The incidence of symptomatic intracerebral hemorrhage (sICH) did not differ between the groups. Regarding the ICH patients, NIHSS score at 24 h (p = 0.036), mortality at 3 months (p = 0.004) and at one year (p = 0.00) were higher in the COVID-19 group. We concluded that the pandemic resulted fewer admission due to AIS with an increased number of LVOs and MTs. AIS patients with concomitant SARS-CoV-2 infection have more severe strokes and unfavorable long term outcome. The risk of sICH was not increased in COVID-19 positive patients therefore reperfusion therapies appear to be safe and beneficial for some individuals. Patients with ICH and comorbid COVID-19 have a very poor prognosis.
Collapse
Affiliation(s)
- Máté Héja
- Department of Neurology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| | - István Fekete
- Department of Neurology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Sándor Márton
- Faculty of Arts, Institute of Political Science and Sociology, University of Debrecen, Debrecen, Hungary
| | - László Horváth
- Department of Pharmaceutical Surveillance and Economics, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Klára Fekete
- Department of Neurology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
27
|
Svobodová B, Löfdahl A, Nybom A, Wigén J, Hirdman G, Olm F, Brunnström H, Lindstedt S, Westergren-Thorsson G, Elowsson L. Overlapping Systemic Proteins in COVID-19 and Lung Fibrosis Associated with Tissue Remodeling and Inflammation. Biomedicines 2024; 12:2893. [PMID: 39767799 PMCID: PMC11727205 DOI: 10.3390/biomedicines12122893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/09/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
Background/Objectives: A novel patient group with chronic pulmonary fibrosis is emerging post COVID-19. To identify patients at risk of developing post-COVID-19 lung fibrosis, we here aimed to identify systemic proteins that overlap with fibrotic markers identified in patients with idiopathic pulmonary fibrosis (IPF) and may predict COVID-19-induced lung fibrosis. Methods: Ninety-two proteins were measured in plasma samples from hospitalized patients with moderate and severe COVID-19 in Sweden, before the introduction of the vaccination program, as well as from healthy individuals. These measurements were conducted using proximity extension assay (PEA) technology with a panel including inflammatory and remodeling proteins. Histopathological alterations were evaluated in explanted lung tissue. Results: Connecting to IPF pathology, several proteins including decorin (DCN), tumor necrosis factor receptor superfamily member 12A (TNFRSF12A) and chemokine (C-X-C motif) ligand 13 (CXCL13) were elevated in COVID-19 patients compared to healthy subjects. Moreover, we found incrementing expression of monocyte chemotactic protein-3 (MCP-3) and hepatocyte growth factor (HGF) when comparing moderate to severe COVID-19. Conclusions: Both extracellular matrix- and inflammation-associated proteins were identified as overlapping with pulmonary fibrosis, where we found DCN, TNFRSF12A, CXCL13, CXCL9, MCP-3 and HGF to be of particular interest to follow up on for the prediction of disease severity.
Collapse
Affiliation(s)
- Barbora Svobodová
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden; (B.S.); (J.W.); (G.W.-T.)
| | - Anna Löfdahl
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden; (B.S.); (J.W.); (G.W.-T.)
| | - Annika Nybom
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden; (B.S.); (J.W.); (G.W.-T.)
| | - Jenny Wigén
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden; (B.S.); (J.W.); (G.W.-T.)
| | - Gabriel Hirdman
- Department of Cardiothoracic Surgery and Transplantation, Skåne University Hospital, 222 42 Lund, Sweden; (G.H.); (F.O.); (S.L.)
- Wallenberg Center for Molecular Medicine, Lund University, 221 84 Lund, Sweden
- Department of Clinical Sciences, Lund University, 221 84 Lund, Sweden;
| | - Franziska Olm
- Department of Cardiothoracic Surgery and Transplantation, Skåne University Hospital, 222 42 Lund, Sweden; (G.H.); (F.O.); (S.L.)
- Wallenberg Center for Molecular Medicine, Lund University, 221 84 Lund, Sweden
- Department of Clinical Sciences, Lund University, 221 84 Lund, Sweden;
| | - Hans Brunnström
- Department of Clinical Sciences, Lund University, 221 84 Lund, Sweden;
| | - Sandra Lindstedt
- Department of Cardiothoracic Surgery and Transplantation, Skåne University Hospital, 222 42 Lund, Sweden; (G.H.); (F.O.); (S.L.)
- Wallenberg Center for Molecular Medicine, Lund University, 221 84 Lund, Sweden
- Department of Clinical Sciences, Lund University, 221 84 Lund, Sweden;
| | - Gunilla Westergren-Thorsson
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden; (B.S.); (J.W.); (G.W.-T.)
| | - Linda Elowsson
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden; (B.S.); (J.W.); (G.W.-T.)
| |
Collapse
|
28
|
Wang KC, Wang S, Serna J, Su F, Halvorson RT, Lansdown DA, Ma CB, Zhang AL. COVID-19 Diagnosis Within 6 Weeks of Arthroscopic Knee Surgery Leads to Higher Risk for Postoperative Venous Thromboembolism. Arthroscopy 2024:S0749-8063(24)01076-4. [PMID: 39709106 DOI: 10.1016/j.arthro.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 12/23/2024]
Abstract
PURPOSE To determine if preoperative infection with COVID-19 increased risk for postoperative venous thromboembolism (VTE) in patients undergoing arthroscopic knee surgery. METHODS PearlDiver Mariner 165 database was queried for patients undergoing knee arthroscopy between 2010 and October 2022. Patients were categorized by history of COVID-19 diagnosis and timing in relation to surgery. Multivariate logistic regression was performed to isolate the effect of COVID-19 diagnosis on postoperative VTE rates. Covariates included age, obesity, smoking, oral contraceptive pill use, hypertension, coronary artery disease, congestive heart failure, malignancy, renal disease, and diabetes. RESULTS A total of 954,294 patients met inclusion criteria, and 7,637 patients experienced VTE, including deep vein thrombosis (n = 5,830, 0.61%) and pulmonary embolism (n = 2,790, 0.29%). Patients with a COVID-19 diagnosis before surgery (7,858) had an overall higher incidence of VTE (1.72%) compared to patients without a preoperative COVID-19 diagnosis (0.81%) (P < .001). There was no difference in VTE incidence among patients with a preoperative COVID-19 diagnosis undergoing knee arthroscopy in the pre- and post-COVID-19 vaccination eras (odds ratio, 0.82; 95% CI, 0.51-1.31; P = .48). Multivariate regression accounting for covariates showed that patients with a preoperative COVID-19 diagnosis within 6 weeks before arthroscopy had significantly increased odds of experiencing postoperative VTE (odds ratio, 1.68; 95% CI, 1.09-2.45; P = .012). There was no significant difference in VTE risk among patients with a preoperative COVID-19 diagnosis between 6 and 12 weeks before arthroscopy and more than 12 weeks before arthroscopy compared to those with no prior COVID-19 diagnosis. CONCLUSIONS COVID-19 diagnosis within 6 weeks preceding arthroscopic knee surgery leads to a significantly higher risk for postoperative VTE. LEVEL OF EVIDENCE Level III, retrospective case control study.
Collapse
Affiliation(s)
- Kevin C Wang
- Department of Orthopedic Surgery, University of California San Francisco, San Francisco, California, USA
| | - Sophia Wang
- Department of Orthopedic Surgery, University of California San Francisco, San Francisco, California, USA
| | - Juan Serna
- Department of Orthopedic Surgery, University of California San Francisco, San Francisco, California, USA
| | - Favian Su
- Department of Orthopedic Surgery, University of California San Francisco, San Francisco, California, USA
| | - Ryan T Halvorson
- Department of Orthopedic Surgery, University of California San Francisco, San Francisco, California, USA
| | - Drew A Lansdown
- Department of Orthopedic Surgery, University of California San Francisco, San Francisco, California, USA
| | - C Benjamin Ma
- Department of Orthopedic Surgery, University of California San Francisco, San Francisco, California, USA
| | - Alan L Zhang
- Department of Orthopedic Surgery, University of California San Francisco, San Francisco, California, USA.
| |
Collapse
|
29
|
El-Menyar A, Ramzee AF, Asim M, Shahid F, Ata YM, El Baba H, Fino A, Nair AP, Peralta R, Almaslamani MA, Al Suwaidi J, Al-Thani H, Rizoli S. COVID-19 Increases the Risk of New Myocardial Infarction in Patients with Old Myocardial Infarction: A Retrospective Observational Study. CLINICAL MEDICINE INSIGHTS-CARDIOLOGY 2024; 18:11795468241301133. [PMID: 39697349 PMCID: PMC11653445 DOI: 10.1177/11795468241301133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 10/07/2024] [Indexed: 12/20/2024]
Abstract
Background We aimed to investigate the incidence of new acute myocardial infarction (AMI), in patients with Coronavirus disease (COVID-19) who had old MI. We hypothesized that COVID-19 increases the rate of repeated AMI in this population regardless of age and gender. Methods A retrospective analysis was conducted for adult patients admitted with COVID-19 and developed thromboembolic event (TEE) in 2020. Patients were categorized based on the history of old MI, new MI, age, and gender. Results Among 16,903 patients with COVID-19 who were admitted, 210 (1.2%) developed TEE (89% were males, 55% were <55 years old, and 80.5% had an old MI). COVID-19 was severe in 32% of cases. AMI occurred in 160 patients (42.5% STEMI and 57.5% NSTEMI). In patients with prior MI, 92.5% developed another AMI. NSTEMI was higher in patients with severe COVID-19 than STEMI (33% vs 21%). Patients with severe COVID-19 had higher mortality (39.4% vs 5.6%), fewer rates of prior MI (74% vs 83%), hypertension (40% vs 60%), and STEMI (31.8% vs 46.5%) than mild COVID-19 patients. On multivariable analysis, COVID-19 severity was an independent predictor of mortality (OR10; 95%CI 1.62-67.19) after adjustment for age, gender, diabetes mellitus, C-reactive protein, serum Ferritin, Procalcitonin, and Fibrinogen values, and prior or new MI. Conclusions Patients with old MI could develop a new AMI in 80% of COVID-19. However, the mortality was higher in patients without a history of MI due to the severity of COVID-19. Attention should be given to patients who possess thrombotic risk factors in pandemics.
Collapse
Affiliation(s)
- Ayman El-Menyar
- Clinical Research, Trauma and Vascular Surgery, Hamad Medical Corporation, Doha, Qatar
- Clinical Medicine, Weill Cornell Medical College, Doha, Qatar
| | | | - Mohammad Asim
- Clinical Research, Trauma and Vascular Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Fakhar Shahid
- Department of Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Yaser M Ata
- Department of Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Hamzah El Baba
- Department of Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Areen Fino
- Department of Family Medicine, Hamad Medical Corporation, Doha, Qatar
| | - Arun P Nair
- Communicable Disease Center (CDC), Hamad Medical Corporation, Doha, Qatar
| | - Ruben Peralta
- Trauma Surgery Section, Hamad Medical Corporation, Doha, Qatar
- Department of Surgery, Universidad Nacional Pedro Henriquez Urena, Santo Domingo, Dominican Republic
| | - Muna A Almaslamani
- Communicable Disease Center (CDC), Hamad Medical Corporation, Doha, Qatar
| | - Jassim Al Suwaidi
- Department of Cardiology, Heart Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Hassan Al-Thani
- Trauma Surgery Section, Hamad Medical Corporation, Doha, Qatar
| | - Sandro Rizoli
- Trauma Surgery Section, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
30
|
Cao J, Huang Z, Zeng J, Liu J, Zuo W, Su Z, Chen Y, Yu W, Ye H. Maternal and neonatal outcomes and clinical laboratory testing of pregnant women with COVID-19 during the BA.5.2/BF.7 surge. Virulence 2024; 15:2360130. [PMID: 38803076 PMCID: PMC11152110 DOI: 10.1080/21505594.2024.2360130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/22/2024] [Indexed: 05/29/2024] Open
Abstract
The impact of COVID-19 on pregnant women and newborns continues to be a critical societal concern. However, the majority of research focuses on the disease resulting from the early pandemic variants, without sufficient study on the more recent BA.5.2/BF.7. We retrospectively recruited pregnant women giving birth during the surge of the BA.5.2/BF.7 and analysed the risk impact of COVID-19 on maternal and neonatal outcomes. Furthermore, subjects matched through propensity scores were used for the analysis of clinical laboratory tests. A total of 818 pregnant women were enrolled, among 276 (33.7%) were diagnosed with SARS-CoV-2 during childbirth. COVID-19 significantly increased the risk of a hospital length of stay equal to or greater than seven days and neonatal admission to the neonatal intensive care unit, with an aHR of 2.03 (95% CI, 1.22-3.38) and 1.51 (95% CI, 1.12-2.03), respectively. In the analysis of 462 matched subjects, it was found that subjects infected with SARS-CoV-2 tended slight leucopenia and coagulation abnormalities. We found that during the surge of the BA.5.2/BF.7, COVID-19 increased the risk of maternal and neonatal outcomes among Chinese pregnant women. This finding offers significant insights to guide clinical practices involving pregnant women infected with the recently emerged Omicron subvariants.
Collapse
Affiliation(s)
- Jiali Cao
- Department of Laboratory Medicine, Fujian Key Clinical Specialty of Laboratory Medicine, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Zehong Huang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
| | - Jing Zeng
- School of Pharmacy, Xiamen University, Xiamen, China
| | - Jumei Liu
- Department of Laboratory Medicine, Fujian Key Clinical Specialty of Laboratory Medicine, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Weilun Zuo
- Department of Laboratory Medicine, Fujian Key Clinical Specialty of Laboratory Medicine, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Zhiying Su
- Department of Obstetrics, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Yujuan Chen
- Department of Obstetrics, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Weiwei Yu
- Department of Obstetrics, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Huiming Ye
- Department of Laboratory Medicine, Fujian Key Clinical Specialty of Laboratory Medicine, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
31
|
Indra S, Chalak K, Das P, Mukhopadhyay A. Placenta a potential gateway of prenatal SARS-CoV-2 infection: A review. Eur J Obstet Gynecol Reprod Biol 2024; 303:123-131. [PMID: 39461078 DOI: 10.1016/j.ejogrb.2024.10.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/29/2024]
Abstract
SARS-CoV-2, the causative agent of COVID-19, can infect various tissues in the body apart from the lungs. Although placental infection remains controversial, COVID-19-associated placental abnormalities have been reported worldwide. Therefore, COVID-19 poses a significant risk for fetal distress as well. Scientists are currently debating whether such distress results from direct viral induced assault or placental damage caused by the mother's immune response. The placenta develops different histopathological lesions in response to maternal SARS-CoV-2 infection. While some studies support both theories, the transmission rate through the placenta remains low. Therefore, a more in-depth study is necessary to determine the primary cause of maternal SARS-CoV-2-induced fetal distress. This comprehensive review is aimed to shed light on the possible reasons towards fetal distress among mothers with COVID-19. This review describes the various mechanisms of viral entry along with the mechanisms by which the virus could affect the placenta. Reported cases of placental abnormalities and fetal distress symptoms have been collated to provide an overview of the current state of knowledge on vertical transmission of COVID-19.
Collapse
Affiliation(s)
- Subhashis Indra
- Department of Life Sciences, Presidency University, Kolkata 700073, India
| | - Kuheli Chalak
- Department of Life Sciences, Presidency University, Kolkata 700073, India
| | - Purbasha Das
- Department of Life Sciences, Presidency University, Kolkata 700073, India
| | | |
Collapse
|
32
|
Gabet A, Blacher J, Tuppin P, Lailler G, Grave C, Sanchez O, Mahe I, Emmerich J, Olié V. Epidemiology of venous thromboembolism in France. Arch Cardiovasc Dis 2024; 117:715-724. [PMID: 39632128 DOI: 10.1016/j.acvd.2024.10.325] [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: 06/28/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Few epidemiological data are available for venous thromboembolism (VTE) at French national and subnational levels. AIMS To quantify VTE events in France in 2022 and describe the features of hospital management and outcomes. METHODS Adults hospitalized for a VTE as the primary reason for hospitalization or treatment in a medical unit in 2022 were identified from medical administrative data. Data were stratified as pulmonary embolism (PE) and deep vein thrombosis (DVT), and by French department and various sociodemographic indicators. VTE prevalence at 1 January 2023 was defined as the number of people alive at that date with a history of hospitalization for VTE or a chronic long-term disease status due to VTE (2012-2022). RESULTS VTE cases reached 896,846 adults on 1 January 2023. VTE was the primary diagnosis for a hospital stay or medical unit in 62,055 patients hospitalized in 2022. The age-standardized rate of hospitalized patients was 23.0% higher for men versus women. There were considerable variations between departments of residence, while Martinique had the highest age-standardized rate. The prevalence of triggering factors was high, with almost 30% having cancer and 20% a recent long hospitalization. One-year mortality was approximately 20% for both PE and DVT, despite rehospitalization rates <5%. CONCLUSION The high prevalence of cancer among patients hospitalized due to VTE partly explains the high 1-year mortality. As VTE is partially avoidable, the prevention of VTE needs to be improved in France and whether thromboprophylaxis guidance is being followed should regularly be assessed.
Collapse
Affiliation(s)
- Amélie Gabet
- Santé Publique France, 94410 Saint-Maurice, France.
| | - Jacques Blacher
- Paris public hospitals (AP-HP), Hôtel-Dieu Hospital, Paris Cité University, 75004 Paris, France
| | | | | | | | - Olivier Sanchez
- Paris Cité University, Respiratory Medicine and Intensive Care Department, Georges-Pompidou European Hospital, AP-HP, Inserm UMRS 1140, 75015 Paris, France
| | - Isabelle Mahe
- Paris Cité University, Internal Medicine Department, Louis-Mourier Hospital, AP-HP, Inserm UMRS 1140, 92700 Colombes, France
| | - Joseph Emmerich
- Paris Cité University, Inserm CRESS 1153 and Paris Saint-Joseph Hospital, 75013 Paris, France
| | - Valérie Olié
- Santé Publique France, 94410 Saint-Maurice, France
| |
Collapse
|
33
|
Asgari A, Franczak A, Herchen A, Jickling GC, Jurasz P. Elevated levels of pro-thrombotic eNOS-negative platelets in COVID-19 patients. Thromb Res 2024; 244:109178. [PMID: 39369655 DOI: 10.1016/j.thromres.2024.109178] [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: 05/09/2024] [Revised: 09/13/2024] [Accepted: 10/02/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Platelet-rich microvascular thrombi are common in severe COVID-19. Endogenous nitric oxide (NO)-signaling limits thrombus formation and previously we identified platelet subpopulations with a differential ability to produce NO based on the presence or absence of endothelial nitric oxide synthase (eNOS). eNOS expression is counter-regulated by cytokines, and COVID-19-associated immune/inflammatory responses may affect the transcriptome profile of megakaryocytes and their platelet progeny. OBJECTIVES We investigated whether the percentage of eNOS-negative to eNOS-positive platelets increases in COVID-19 patients and whether this change may be due to the actions of pro-inflammatory cytokines on megakaryocytes. METHODS Platelets were isolated from hospitalized COVID-19 patients and COVID-19-negative controls. Platelet eNOS was measured by flow cytometry and plasma inflammatory cytokines by ELISA. Megakaryocytes from eNOS-GFP transgenic mice and the Meg-01 cell line were characterized to identify an appropriate model to study eNOS-based platelet subpopulation formation in response to inflammatory cytokines. RESULTS COVID-19 patients demonstrated a significant increase in eNOS-negative and a concomitant decrease in eNOS-positive platelets compared to controls, and this change was associated with disease severity as assessed by ICU admission. A higher eNOS-negative to -positive platelet percentage was associated with enhanced platelet activation as measured by surface CD62P. Accordingly, COVID-19 patients demonstrated higher TNF-α, IL-6, and IL-1β plasma concentrations than controls. Inflammatory cytokines associated with COVID-19 promoted eNOS-negative Meg-01 formation and enhanced subsequent eNOS-negative platelet-like particle formation. CONCLUSIONS COVID-19 patients have a higher percentage of eNOS-negative to -positive platelets, likely as a result of inflammatory response reducing megakaryocyte eNOS expression, which predisposes to thrombosis.
Collapse
Affiliation(s)
- Amir Asgari
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Aleksandra Franczak
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Alex Herchen
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada; School of Medicine and Dentistry, Griffith University, Queensland, Australia
| | - Glen C Jickling
- Department of Medicine, Division of Neurology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Paul Jurasz
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada; Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada; Cardiovascular Research Institute, University of Alberta, Edmonton, AB, Canada; Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
34
|
Wang K, Nie Y, Maguire C, Syphurs C, Sheen H, Karoly M, Lapp L, Gygi JP, Jayavelu ND, Patel RK, Hoch A, Corry D, Kheradmand F, McComsey GA, Fernandez-Sesma A, Simon V, Metcalf JP, Higuita NIA, Messer WB, Davis MM, Nadeau KC, Kraft M, Bime C, Schaenman J, Erle D, Calfee CS, Atkinson MA, Brackenridge SC, Hafler DA, Shaw A, Rahman A, Hough CL, Geng LN, Ozonoff A, Haddad EK, Reed EF, van Bakel H, Kim-Schultz S, Krammer F, Wilson M, Eckalbar W, Bosinger S, Langelier CR, Sekaly RP, Montgomery RR, Maecker HT, Krumholz H, Melamed E, Steen H, Pulendran B, Augustine AD, Cairns CB, Rouphael N, Becker PM, Fourati S, Shannon CP, Smolen KK, Peters B, Kleinstein SH, Levy O, Altman MC, Iwasaki A, Diray-Arce J, Ehrlich LIR, Guan L. Unraveling SARS-CoV-2 Host-Response Heterogeneity through Longitudinal Molecular Subtyping. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624784. [PMID: 39651165 PMCID: PMC11623532 DOI: 10.1101/2024.11.22.624784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Hospitalized COVID-19 patients exhibit diverse immune responses during acute infection, which are associated with a wide range of clinical outcomes. However, understanding these immune heterogeneities and their links to various clinical complications, especially long COVID, remains a challenge. In this study, we performed unsupervised subtyping of longitudinal multi-omics immunophenotyping in over 1,000 hospitalized patients, identifying two critical subtypes linked to mortality or mechanical ventilation with prolonged hospital stay and three severe subtypes associated with timely acute recovery. We confirmed that unresolved systemic inflammation and T-cell dysfunctions were hallmarks of increased severity and further distinguished patients with similar acute respiratory severity by their distinct immune profiles, which correlated with differences in demographic and clinical complications. Notably, one critical subtype (SubF) was uniquely characterized by early excessive inflammation, insufficient anticoagulation, and fatty acid dysregulation, alongside higher incidences of hematologic, cardiac, and renal complications, and an elevated risk of long COVID. Among the severe subtypes, significant differences in viral clearance and early antiviral responses were observed, with one subtype (SubC) showing strong early T-cell cytotoxicity but a poor humoral response, slower viral clearance, and greater risks of chronic organ dysfunction and long COVID. These findings provide crucial insights into the complex and context-dependent nature of COVID-19 immune responses, highlighting the importance of personalized therapeutic strategies to improve both acute and long-term outcomes.
Collapse
|
35
|
Ozer V, Gonenc Cekic O, Bulbul O, Aydın D, Bulut E, Aksoy F, Pehlivanlar Kucuk M, Caner Karahan S, Emel Sozen E, Ozkaya E, Kosucu P, Karaca Y, Turedi S. Diagnostic and Prognostic Value of SCUBE-1 in COVID-19 Patients. West J Emerg Med 2024; 25:975-984. [PMID: 39625772 PMCID: PMC11610722 DOI: 10.5811/westjem.18586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 12/06/2024] Open
Abstract
Introduction The workload of physicians increased due to the number of patients presenting with suspicion of coronavirus 2019 (COVID-19) and the prolonged wait times in the emergency department during the COVID-19 pandemic. Signal peptide-CUB-EGF domain-containing protein 1 (SCUBE-1) is a protein present in platelets and endothelial cells; it is activated by inflammation from COVID-19 and may be associated with COVID-19's known thrombotic risk. We aimed to determine whether SCUBE-1 levels are diagnostically correlated in suspected COVID-19 patients, and whether SCUBE-1 correlated with severity of disease and, therefore, might be useful to guide hospitalization/discharge decisions. Methods The suspected COVID-19 patients cared for at tertiary healthcare institutions for one year between May 2021-May 2022 were examined in this study. The subjects were both suspected COVID-19 patients not ultimately found to have COVID-19 and those who were diagnosed with COVID-19. By modifying the disease severity scoring systems present in COVID-19 guidelines in 2021, the COVID-19-positive patient group was classified as mild, moderate, severe, and critical, and compared using the SCUBE-1 levels. Moreover, SCUBE-1 levels were compared between the COVID-19 positive group and the COVID-19 negative group. Results A total of 507 patients were considered for the present study. After excluding 175 patients for incomplete data and alternate comorbid organ failure. we report on 332 patients (65.5%). Of these 332 patients, 80 (24.0%) were COVID-19 negative, and 252 (76.0%) were COVID-19 positive. Of 252 (100%) patients diagnosed with COVID-19, 74 (29.4%) were classified as mild, 95 (37.7%) moderate, 45 (17.8%) severe, and 38 (15.1%) critical. The SCUBE-1 levels were statistically different between COVID-19 positive (8.48 ± 7.42 nanograms per milliliter [ng/mL]) and COVID-19 negative (1.86 ± 0.92 ng/mL) patients (P < 0.001). In the COVID-19 positive group, SCUBE-1 levels increased with disease severity (mild = 3.20 ± 1.65 ng/mL, moderate = 4.78 ± 2.26 ng/mL, severe = 13.68 ± 3.95 ng/mL, and critical = 21.87 ± 5.39 ng/mL) (P < 0.001). The initial SCUBE-1 levels of discharged patients were significantly lower than those requiring hospitalization (discharged = 2.89 ng/mL [0.55-8.60 ng/mL]; ward admitted = 7.13 ng/mL [1.38-21.29 ng/mL], and ICU admitted = 21.19 ng/mL [10.58-37.86 ng/mL]) (P < 0.001). Conclusion The SCUBE-1 levels were found to be differentiated between patients with and without COVID-19 and to be correlated with the severity of illness.
Collapse
Affiliation(s)
- Vildan Ozer
- Karadeniz Technical University, School of Medicine, Department of Emergency Medicine, Trabzon, Türkiye
| | - Ozgen Gonenc Cekic
- SBU Kanuni Training and Research Hospital, Department of Emergency Medicine, Trabzon, Türkiye
| | - Ozlem Bulbul
- Karadeniz Technical University, School of Medicine, Department of Emergency Medicine, Trabzon, Türkiye
| | - Davut Aydın
- SBU Kanuni Training and Research Hospital, Department of Chest Diseases, Division of Intensive Care Medicine, Trabzon, Türkiye
| | - Eser Bulut
- SBU Kanuni Training and Research Hospital, Department of Radiology, Trabzon, Türkiye
| | - Firdevs Aksoy
- Karadeniz Technical University, School of Medicine, Department of Infectious Diseases and Clinical Microbiology, Trabzon, Türkiye
| | - Mehtap Pehlivanlar Kucuk
- Karadeniz Technical University, School of Medicine, Department of Chest Diseases, Division of Intensive Care Medicine, Trabzon, Türkiye
| | - Suleyman Caner Karahan
- Karadeniz Technical University, School of Medicine, Department of Biochemistry, Trabzon, Türkiye
| | - Ebru Emel Sozen
- SBU Kanuni Training and Research Hospital, Department of Infectious Diseases and Clinical Microbiology, Trabzon, Türkiye
| | - Esra Ozkaya
- Karadeniz Technical University, School of Medicine, Department of Medical Microbiology, Trabzon, Türkiye
| | - Polat Kosucu
- Karadeniz Technical University, School of Medicine, Department of Radiology, Trabzon, Türkiye
| | - Yunus Karaca
- Karadeniz Technical University, School of Medicine, Department of Emergency Medicine, Trabzon, Türkiye
| | - Suleyman Turedi
- SBU Kanuni Training and Research Hospital, Department of Emergency Medicine, Trabzon, Türkiye
| |
Collapse
|
36
|
Georgescu I, Artene SA, Giubelan LI, Tache DE, Dumitrescu F, Duta C, Mirea AA, Manea Carneluti EV, Dricu A, Popescu OS. Evaluation of the Demographics, Clinical Laboratory Parameters, and Outcomes of Hospitalized Oncological Versus Non-oncological COVID-19 Patients. Cureus 2024; 16:e73313. [PMID: 39655133 PMCID: PMC11626416 DOI: 10.7759/cureus.73313] [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: 11/08/2024] [Indexed: 12/12/2024] Open
Abstract
Introduction The COVID-19 pandemic emerged globally in 2019, exposing healthcare vulnerabilities. This study delves into the impact of COVID-19 on cancer patients, a high-risk group with increased susceptibility and mortality rates. Recent research underscores cancer patients' vulnerability to severe disease, often due to compromised immunity. Materials and methods This retrospective study analyzed data from 474 adult COVID-19 patients, admitted between March 2020 and July 2023. Patients were categorized into two groups: those with a medically recorded oncological disease (237) and those without any malignant history (237). Demographic and hematologic analysis aim to unveil COVID-19 impact on individuals with cancer history. Results Statistically significant differences in blood parameters highlighted distinctions, with cancer patients exhibiting higher creatinine, leukocyte, and D Dimers levels as well as lower hemoglobin, neutrophile, lymphocyte, and Serum Glutamate-Pyruvate Transaminase (SGPT) levels. Non-significant differences in certain parameters prompted a nuanced exploration of metabolic and coagulation variations. Conclusion This study unveils global COVID-19 effects on cancer patients, emphasizing clinical and laboratory differences. Findings underscore the imperative need for targeted interventions and enhanced support for cancer patients during the pandemic. Study limitations stress careful interpretation, urging further exploration of COVID-19 and cancer interplay.
Collapse
Affiliation(s)
- Ilona Georgescu
- Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, Craiova, ROU
- Infectious Diseases, "Victor Babeş" Clinical Hospital of Infectious Diseases and Pneumo-phtisiology, Craiova, ROU
| | - Stefan Alexandru Artene
- Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, Craiova, ROU
| | - Lucian-Ion Giubelan
- Infectious Diseases, "Victor Babeş" Clinical Hospital of Infectious Diseases and Pneumo-phtisiology, Craiova, ROU
| | - Daniela Elise Tache
- Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, Craiova, ROU
| | - Florentina Dumitrescu
- Infectious Diseases, "Victor Babeş" Clinical Hospital of Infectious Diseases and Pneumo-phtisiology, Craiova, ROU
| | - Carmen Duta
- Biochemistry, Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest, ROU
| | - Adina Andreea Mirea
- Dental Medicine, University of Medicine and Pharmacy of Craiova, Craiova, ROU
| | | | - Anica Dricu
- Biochemistry, Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest, ROU
| | - Oana Stefana Popescu
- Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, Craiova, ROU
| |
Collapse
|
37
|
Avdonin PP, Blinova MS, Serkova AA, Komleva LA, Avdonin PV. Immunity and Coagulation in COVID-19. Int J Mol Sci 2024; 25:11267. [PMID: 39457048 PMCID: PMC11508857 DOI: 10.3390/ijms252011267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/23/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Discovered in late 2019, the SARS-CoV-2 coronavirus has caused the largest pandemic of the 21st century, claiming more than seven million lives. In most cases, the COVID-19 disease caused by the SARS-CoV-2 virus is relatively mild and affects only the upper respiratory tract; it most often manifests itself with fever, chills, cough, and sore throat, but also has less-common mild symptoms. In most cases, patients do not require hospitalization, and fully recover. However, in some cases, infection with the SARS-CoV-2 virus leads to the development of a severe form of COVID-19, which is characterized by the development of life-threatening complications affecting not only the lungs, but also other organs and systems. In particular, various forms of thrombotic complications are common among patients with a severe form of COVID-19. The mechanisms for the development of thrombotic complications in COVID-19 remain unclear. Accumulated data indicate that the pathogenesis of severe COVID-19 is based on disruptions in the functioning of various innate immune systems. The key role in the primary response to a viral infection is assigned to two systems. These are the pattern recognition receptors, primarily members of the toll-like receptor (TLR) family, and the complement system. Both systems are the first to engage in the fight against the virus and launch a whole range of mechanisms aimed at its rapid elimination. Normally, their joint activity leads to the destruction of the pathogen and recovery. However, disruptions in the functioning of these innate immune systems in COVID-19 can cause the development of an excessive inflammatory response that is dangerous for the body. In turn, excessive inflammation entails activation of and damage to the vascular endothelium, as well as the development of the hypercoagulable state observed in patients seriously ill with COVID-19. Activation of the endothelium and hypercoagulation lead to the development of thrombosis and, as a result, damage to organs and tissues. Immune-mediated thrombotic complications are termed "immunothrombosis". In this review, we discuss in detail the features of immunothrombosis associated with SARS-CoV-2 infection and its potential underlying mechanisms.
Collapse
Affiliation(s)
| | | | | | | | - Pavel V. Avdonin
- Koltzov Institute of Developmental Biology RAS, ul. Vavilova, 26, 119334 Moscow, Russia; (P.P.A.)
| |
Collapse
|
38
|
Liu Y, Lou X. The Bidirectional Association Between Metabolic Syndrome and Long-COVID-19. Diabetes Metab Syndr Obes 2024; 17:3697-3710. [PMID: 39398386 PMCID: PMC11471063 DOI: 10.2147/dmso.s484733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/22/2024] [Indexed: 10/15/2024] Open
Abstract
Background The rapid global spread of a new coronavirus disease known as COVID-19 has led to a significant increase in mortality rates, resulting in an unprecedented worldwide pandemic. Methods The impact of COVID-19, particularly its long-term effects, has also had a profound effect on the health and well-being of individuals.Metabolic syndrome increases the risk of heart and brain diseases, presenting a significant danger to human well-being. Purpose The prognosis of long COVID and the progression of metabolic syndrome interact with each other, but there is currently a lack of systematic reports.In this paper, the pathogenesis, related treatment and prognosis of long COVID and metabolic syndrome are systematically reviewed.
Collapse
Affiliation(s)
- Yanfen Liu
- Department of Endocrinology at Zhejiang University School of Medicine, Jinhua Hospital, Jinhua, People’s Republic of China
| | - Xueyong Lou
- Department of Endocrinology at Zhejiang University School of Medicine, Jinhua Hospital, Jinhua, People’s Republic of China
| |
Collapse
|
39
|
Tsai J, Malik S, Tjen-A-Looi SC. Pulmonary Hypertension: Pharmacological and Non-Pharmacological Therapies. Life (Basel) 2024; 14:1265. [PMID: 39459565 PMCID: PMC11509317 DOI: 10.3390/life14101265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 09/23/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Pulmonary hypertension (PH) is a severe and chronic disease characterized by increased pulmonary vascular resistance and remodeling, often precipitating right-sided heart dysfunction and death. Although the condition is progressive and incurable, current therapies for the disease focus on multiple different drugs and general supportive therapies to manage symptoms and prolong survival, ranging from medications more specific to pulmonary arterial hypertension (PAH) to exercise training. Moreover, there are multiple studies exploring novel experimental drugs and therapies including unique neurostimulation, to help better manage the disease. Here, we provide a narrative review focusing on current PH treatments that target multiple underlying biochemical mechanisms, including imbalances in vasoconstrictor-vasodilator and autonomic nervous system function, inflammation, and bone morphogenic protein (BMP) signaling. We also focus on the potential of novel therapies for managing PH, focusing on multiple types of neurostimulation including acupuncture. Lastly, we also touch upon the disease's different subgroups, clinical presentations and prognosis, diagnostics, demographics, and cost.
Collapse
Affiliation(s)
- Jason Tsai
- Susan Samueli Integrative Health Institute, College of Health Sciences, University of California-Irvine, Irvine, CA 92617, USA;
| | | | - Stephanie C. Tjen-A-Looi
- Susan Samueli Integrative Health Institute, College of Health Sciences, University of California-Irvine, Irvine, CA 92617, USA;
| |
Collapse
|
40
|
Valente Silva B, Jorge C, Plácido R, Nobre Menezes M, Mendonça C, Luísa Urbano M, Rigueira J, G Almeida A, Pinto FJ. Comparison of the accuracy of four diagnostic prediction rules for pulmonary embolism in patients admitted to the emergency department. Rev Port Cardiol 2024; 43:551-559. [PMID: 38663529 DOI: 10.1016/j.repc.2024.02.006] [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: 05/10/2023] [Revised: 12/21/2023] [Accepted: 02/07/2024] [Indexed: 07/12/2024] Open
Abstract
INTRODUCTION AND OBJECTIVES Ruling out pulmonary embolism (PE) through a combination of clinical assessment and D-dimer level can potentially avoid excessive use of computed tomography pulmonary angiography (CTPA). We aimed to compare the diagnostic accuracy of the standard approach based on the Wells and Geneva scores combined with a standard D-dimer cut-off (500 ng/ml), with three alternative strategies (age-adjusted and the YEARS and PEGeD algorithms) in patients admitted to the emergency department (ED) with suspected PE. METHODS Consecutive outpatients admitted to the ED who underwent CTPA due to suspected PE were retrospectively assessed. Sensitivity, specificity, positive and negative predictive values, likelihood ratios and diagnostic odds ratios were calculated and compared between the different diagnostic prediction rules. RESULTS We included 1402 patients (mean age 69±18 years, 54% female), and PE was confirmed in 25%. Compared to the standard approach (p<0.001), an age-adjusted strategy increased specificity with a non-significant decrease in sensitivity only in patients older than 70 years. Compared to the standard and age-adjusted approaches, the YEARS and PEGeD algorithms had the highest specificity across all ages, but were associated with a significant decrease in sensitivity (p<0.001), particularly in patients aged under 60 years (sensitivity of 81% in patients aged between 51 and 60 years). CONCLUSION Compared to the standard approach, all algorithms were associated with increased specificity. The age-adjusted strategy was the only one not associated with a significant decrease in sensitivity compared to the standard approach, enabling CTPA requests to be reduced safely.
Collapse
Affiliation(s)
- Beatriz Valente Silva
- Cardiology Department, Centro Hospitalar Universitário Lisboa Norte, CAML, CCUL, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
| | - Cláudia Jorge
- Cardiology Department, Centro Hospitalar Universitário Lisboa Norte, CAML, CCUL, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Rui Plácido
- Cardiology Department, Centro Hospitalar Universitário Lisboa Norte, CAML, CCUL, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Miguel Nobre Menezes
- Cardiology Department, Centro Hospitalar Universitário Lisboa Norte, CAML, CCUL, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Carlos Mendonça
- Radiology Department, Centro Hospitalar Universitário Lisboa Norte, Lisboa, Portugal
| | - Maria Luísa Urbano
- Radiology Department, Centro Hospitalar Universitário Lisboa Norte, Lisboa, Portugal
| | - Joana Rigueira
- Cardiology Department, Centro Hospitalar Universitário Lisboa Norte, CAML, CCUL, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Ana G Almeida
- Cardiology Department, Centro Hospitalar Universitário Lisboa Norte, CAML, CCUL, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Fausto J Pinto
- Cardiology Department, Centro Hospitalar Universitário Lisboa Norte, CAML, CCUL, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
41
|
Qu P, Lou A, Rong D, Wang C, Zhong Q, Cui W, Gong J, Xu Q, Chen Z, Bathaiian LS, Li X, Chen C. Effect of amubarvimab-romlusevimab for treatment of severe COVID-19 in intensive care units: A retrospective cohort study. Heliyon 2024; 10:e37663. [PMID: 39315175 PMCID: PMC11417170 DOI: 10.1016/j.heliyon.2024.e37663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 08/08/2024] [Accepted: 09/07/2024] [Indexed: 09/25/2024] Open
Abstract
Amubarvimab-romlusevimab is a commonly recommended antiviral treatment in China for adult patients with mild or moderate SARS-CoV-2 infections, especially for patients with a high risk factor for progression to severe COVID-19. However, its exact efficacy in patients with severe Covid-19 is not yet known.This is a single-center retrospective cohort study, in which we collected the general data, laboratory tests, radiological characteristics, viral conversion status, and prognosis of the disease from patients with COVID-19 hospitalized, from December 2022 to March 2023 in the Department of Critical Care Medicine. The amubarvimab-romlusevimab therapy can reduce the 28-day mortality (29.79 % vs 51.35 %, p = 0.02), and ICU mortality (29.79 % vs 55.41 %, p = 0.006) of severe COVID-19.A 1:1 PSM (Propensity Score Matching) was performed to reduce bias, in order to ensure the two groups were balanced and comparable. In the matched population (n = 47), there were no statistically significant differences between the mAbs (monoclonal antibody)group and the Non-antiviral group in 28-day, and thromboembolic events in COVID-19 patients. The 40-day survival analysis shows that mAbs therapy can improve patient prognosis (HR = 0.45, 95%CI = 0.26-0.76, p = 0.008). However, no significant intergroup difference in the 40-day cumulative viral conversion rate. In a univariate Cox regression analysis, The Amubarvimab - romlusevimab therapy(HR:0.464; CI:[0.252-0.853]; p:0.013) is a protective factor and CRP, PCT, PLT, Lactate, PT, PT-INR, and pt% level at admission were risk factors for clinical prognosis. After including the above covariates, Multifactorial COX regression shows that the Amubarvimab - romlusevimab therapy(HR:0.392; CI:[0.211-0.729]; p:0.003), CRP, Lactate and PT-INR at admission are independent factors for mortality of severe COVID-19. Based on the current data, we conclude that amubarvimab-romlusevimab therapy is beneficial for patients with severe COVID-19.
Collapse
Affiliation(s)
- Peng Qu
- Department of Emergency Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Anni Lou
- Department of Emergency Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Dan Rong
- Department of Emergency Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Canmin Wang
- Intensive Care Unit, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Qinglei Zhong
- Department of Emergency Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Wanfu Cui
- Department of Emergency Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Jiacheng Gong
- Department of Emergency Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Qihan Xu
- Department of Emergency Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Zhuoer Chen
- Department of Emergency Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | | | - Xu Li
- Department of Emergency Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Cheng Chen
- Department of Critical Care Medicine, Baiyun Branch, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
42
|
Li B, Bai J, Xiong Y, Guo D, Fu B, Deng G, Wu H. Understanding the mechanisms and treatments of long COVID to address future public health risks. Life Sci 2024; 353:122938. [PMID: 39084516 DOI: 10.1016/j.lfs.2024.122938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/20/2024] [Accepted: 07/27/2024] [Indexed: 08/02/2024]
Abstract
The 2019 coronavirus disease (COVID-19), triggered by the severe acute respiratory syndrome coronavirus (SARS-CoV-2), has seen numerous individuals undergo and recover from it, drawing extensive attention to their health conditions. Extensive studies indicate that even after surpassing the acute phase of infection, patients continue to experience persistent symptoms such as fatigue, pain, depression, weakening, and anosmia. COVID-19 appears not to have concluded but rather to persist long-term in certain individuals, termed as "long COVID." This represents a heterogeneous ailment involving multiple organ systems, with a perceived complex and still elusive pathogenesis. Among patients with long COVID, observations reveal immune dysregulation, coagulation impairments, and microbial dysbiosis, considered potential mechanisms explaining sustained adverse outcomes post COVID-19. Based on the multifactorial nature, varied symptoms, and heterogeneity of long COVID, we have summarized several categories of current therapeutic approaches. Furthermore, the symptoms of long COVID resemble those of other viral illnesses, suggesting that existing knowledge may offer novel insights into long-term COVID implications. Here, we provide an overview of existing literature associated with long COVID and summarize potential mechanisms, treatment modalities, and other analogous conditions. Lastly, we underscore the inadequacies in long COVID treatment approaches and emphasize the significance of conducting further research and clinical trials.
Collapse
Affiliation(s)
- Bohao Li
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Junlu Bai
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Yan Xiong
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Dong Guo
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Beibei Fu
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Guohong Deng
- Department of Infectious Diseases, First Affiliated Hospital, Army Medical University, Chongqing, China.
| | - Haibo Wu
- School of Life Sciences, Chongqing University, Chongqing 401331, China.
| |
Collapse
|
43
|
Tahir A, Draxler A, Stelzer T, Blaschke A, Laky B, Széll M, Binar J, Bartak V, Bragagna L, Maqboul L, Herzog T, Thell R, Wagner KH. A comprehensive IDA and SWATH-DIA Lipidomics and Metabolomics dataset: SARS-CoV-2 case control study. Sci Data 2024; 11:998. [PMID: 39266559 PMCID: PMC11393081 DOI: 10.1038/s41597-024-03822-y] [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: 10/05/2023] [Accepted: 08/27/2024] [Indexed: 09/14/2024] Open
Abstract
A significant hurdle in untargeted lipid/metabolomics research lies in the absence of reliable, cross-validated spectral libraries, leading to a considerable portion of LC-MS features being labeled as unknowns. Despite continuous advancement in annotation tools and libraries, it is important to safeguard, publish and share acquired data through public repositories. Embracing this trend of data sharing not only promotes efficient resource utilization but also paves the way for future repurposing and in-depth analysis; ultimately advancing our comprehension of Covid-19 and other diseases. In this work, we generated an extensive MS-dataset of 39 Covid-19 infected patients versus age- and gender-matched 39 healthy controls. We implemented state of the art acquisition techniques including IDA and SWATH-DIA to ensure a thorough insight in the lipidome and metabolome, ensuring a repurposable dataset.
Collapse
Affiliation(s)
- Ammar Tahir
- Department of Pharmaceutical Sciences, Division of Pharmacognosy, University of Vienna, Vienna, Austria.
- Section of Biomedical Sciences, Department of Health Sciences, FH Campus Wien, University of Applied Sciences, Vienna, Austria.
| | - Agnes Draxler
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
- Vienna Doctoral School for Pharmaceutical, Nutritional and Sport Sciences (PhaNuSpo), University of Vienna, Vienna, Austria
- Department of Health Sciences, FH Campus Wien, University of Applied Sciences, Vienna, Austria
| | - Tamara Stelzer
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
- Vienna Doctoral School for Pharmaceutical, Nutritional and Sport Sciences (PhaNuSpo), University of Vienna, Vienna, Austria
| | | | - Brenda Laky
- Medical University of Vienna, Vienna, Austria
- Austrian Society of Regenerative Medicine, Vienna, Austria
- Sigmund Freud University Vienna, Vienna, Austria
| | - Marton Széll
- Klinik Donaustadt, Emergency Department, Vienna, Austria
| | - Jessica Binar
- Section of Biomedical Sciences, Department of Health Sciences, FH Campus Wien, University of Applied Sciences, Vienna, Austria
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
| | - Viktoria Bartak
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
| | - Laura Bragagna
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
- Vienna Doctoral School for Pharmaceutical, Nutritional and Sport Sciences (PhaNuSpo), University of Vienna, Vienna, Austria
| | - Lina Maqboul
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
- Research Platform Active Ageing, University of Vienna, Vienna, Austria
| | - Theresa Herzog
- Klinik Donaustadt, Emergency Department, Vienna, Austria
| | - Rainer Thell
- Klinik Donaustadt, Emergency Department, Vienna, Austria
| | - Karl-Heinz Wagner
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
- Research Platform Active Ageing, University of Vienna, Vienna, Austria
| |
Collapse
|
44
|
Deora N, Venkatraman K. Potential use of plant-based therapeutics for the management of SARS-COV2 infection in diabetes mellitus – a review. J Herb Med 2024; 47:100923. [DOI: 10.1016/j.hermed.2024.100923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
45
|
Jerah AA, Farasani A, Abu-Tawil H, Kuriri H, Taha MME, Abdelwahab SI, Albasheer O. Unveiling coagulation dysfunction in patients with COVID-19: a retrospective analysis. J Med Life 2024; 17:886-891. [PMID: 39628975 PMCID: PMC11611055 DOI: 10.25122/jml-2024-0166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 08/01/2024] [Indexed: 12/06/2024] Open
Abstract
Coagulation dysfunction has emerged as a significant aspect of COVID-19 pathophysiology, with abnormal coagulation parameters observed in severe cases. This study aimed to investigate the predictive value of coagulation parameters, including prothrombin time (PT), activated partial thromboplastin time (PTT), and international normalized ratio (INR) for mortality in patients with COVID-19. A retrospective analysis was conducted on a cohort of patients diagnosed with COVID-19. Coagulation parameters, including PT, PTT, and INR, were measured upon admission. Receiver operating characteristic (ROC) curve analysis was performed to evaluate the predictive performance of these parameters. Sensitivity and specificity were calculated, and the area under the curve (AUC) values were determined. The analysis included 156 patients diagnosed with COVID-19. The t-test revealed a significant difference (P < 0.05) in PTT, PT, and INR. PTT demonstrated the highest predictive performance, with an AUC value of 0.68, indicating superior discrimination compared with PT and INR. PTT exhibited a sensitivity of 83% and a specificity of 46% for identifying deceased patients. These findings suggest that PTT may serve as a valuable prognostic marker of mortality risk in patients with COVID-19. Coagulation indicators, particularly PTT, predicted COVID-19 mortality. Monitoring coagulation markers may help stratify the risk and guide treatment. Further research and validation studies are needed to corroborate these findings and to establish the clinical importance of coagulation markers in COVID-19 therapy. COVID-19 coagulation dysfunction mechanisms must be understood in order to design targeted therapies to reduce thrombotic consequences.
Collapse
Affiliation(s)
- Ahmed Ali Jerah
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Abdullah Farasani
- Biomedical Research Unit, Medical Research Center, Jazan University, Jazan, Saudi Arabia
| | - Hisham Abu-Tawil
- Department of Clinical Laboratory and Blood Bank, King Faisal Medical City for Southern Regions, Abha, Saudi Arabia
| | - Hadi Kuriri
- Department of Clinical Laboratory and Blood Bank, Samtah General Hospital, Samtah, Saudi Arabia
| | | | | | - Osama Albasheer
- Family and Community Medicine Department, College of Medicine, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
46
|
Ward C, Schlichtholz B. Post-Acute Sequelae and Mitochondrial Aberration in SARS-CoV-2 Infection. Int J Mol Sci 2024; 25:9050. [PMID: 39201736 PMCID: PMC11354507 DOI: 10.3390/ijms25169050] [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: 06/12/2024] [Revised: 07/29/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024] Open
Abstract
This review investigates links between post-acute sequelae of SARS-CoV-2 infection (PASC), post-infection viral persistence, mitochondrial involvement and aberrant innate immune response and cellular metabolism during SARS-CoV-2 infection. Advancement of proteomic and metabolomic studies now allows deeper investigation of alterations to cellular metabolism, autophagic processes and mitochondrial dysfunction caused by SARS-CoV-2 infection, while computational biology and machine learning have advanced methodologies of predicting virus-host gene and protein interactions. Particular focus is given to the interaction between viral genes and proteins with mitochondrial function and that of the innate immune system. Finally, the authors hypothesise that viral persistence may be a function of mitochondrial involvement in the sequestration of viral genetic material. While further work is necessary to understand the mechanisms definitively, a number of studies now point to the resolution of questions regarding the pathogenesis of PASC.
Collapse
Affiliation(s)
| | - Beata Schlichtholz
- Department of Biochemistry, Gdańsk University of Medicine, 80-210 Gdańsk, Poland;
| |
Collapse
|
47
|
Mirza AF, Halim C, Sari MI. The relationship of age, sex and prothrombin time related to the severity of COVID-19 patients with diabetes mellitus: a systematic review and meta analysis. F1000Res 2024; 11:729. [PMID: 40061909 PMCID: PMC11889402 DOI: 10.12688/f1000research.107398.3] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/05/2024] [Indexed: 04/12/2025] Open
Abstract
Background SARS-CoV-2 first appeared in Wuhan, China, in December 2019. Looking at the prevalence data in the world and in Indonesia, the highest mortality rate due to COVID-19 involves age, gender and comorbidities such as diabetes mellitus. Severity of the condition also refers to coagulation abnormalities, such as abnormal prothrombin time values. Methods This systematic review study and meta-analysis used online literature sourced from PubMed, Science Direct, EBSCO, Cochrane and Google Scholar. The literature used here is literature that has data on age, sex and prothrombin time of COVID-19 patients with diabetes mellitus whose quality is assessed by the NOS (Newcastle-Ottawa Scale) criteria and processing data using Review Manager 5.4. Results Out of 8711 literatures that were traced from various search sources, there were 45 literatures that were included in this study. The results of the analysis on age showed the Standardized Mean Difference (SMD) value of 0.45 and P <0.0001 (95% CI: 0.23-0.68), the gender analysis showed an Odds Ratio (OR) value of 3.28 and P = 0.01 (95% CI: 1.26-8.52) and the prothrombin time analysis showed SMD values of 0.41 and P = 0.07 (95%CI = -0.03-0.85). Conclusion Patients with COVID-19 who have DM have a higher risk compared to those without DM. Among COVID-19 patients with DM admitted to hospitals, they were older compared to those without DM and prothrombin time values similar but slightly higher in COVID-19 patients with DM.
Collapse
Affiliation(s)
- Audrey Fabianisa Mirza
- Faculty of Medicine, Universitas Sumatera Utara, Medan, Sumatera Utara, 20155, Indonesia
| | - Ceria Halim
- Faculty of Medicine, Universitas Sumatera Utara, Medan, Sumatera Utara, 20155, Indonesia
| | - Mutiara Indah Sari
- Department of Biochemistry, Universitas Sumatera Utara, Medan, Sumatera Utara, 20155, Indonesia
| |
Collapse
|
48
|
Castro-Varela A, Martinez-Magallanes DM, Reyes-Chavez MF, Gonzalez-Rayas JM, Paredes-Vazquez JG, Vazquez-Garza E, Castillo-Perez M, Flores-Sayavedra YZ, Martinez A, Ramos Cazares RE, Guajardo J, Lopez-de la Garza H, Salinas-Casanova JA, Betancourt H, Molina-Rodriguez AM, Panneflek J, Fabiani MA, Jerjes-Sanchez C. Risk Factors, Clinical Presentation, Therapeutic Trends, and Outcomes in Arterial Thrombosis Complicating Unvaccinated COVID-19 Patients: A Systematic Review. Angiology 2024; 75:625-634. [PMID: 37005343 PMCID: PMC10083125 DOI: 10.1177/00033197231167055] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
Abstract
Data on characteristics and outcomes of coronavirus (COVID)-19 patients complicated with arterial thrombosis (AT) are scarce. Therefore, we carried out a systematic review (PRISMA, PROSPERO statements; PubMed, Scopus, and Web of Science) to identify risk factors, clinical presentation, treatment, and outcomes. We included publications from December 2019 to October 2020. Groups: (a) ischemic stroke, (b) thrombotic storm, (c) peripheral vascular thrombosis, (d) myocardial infarction, and (e) left cardiac thrombus or in-transit thrombus (venous system thrombus floating or attaching to the right heart). We considered 131 studies. The most frequent cardiovascular risk factors were: hypertension, diabetes, and dyslipidemia. A high proportion presented with asymptomatic, mild, or moderate COVID-19 (n = 91, 41.4%). We identified a high percentage of isolated ischemic stroke and thrombotic storm. Groups with higher mortality rate: intracardiac thrombus (1/2, 50.0%), thrombotic storm (18/49, 36.7%), and ischemic stroke (48/131, 36.6%). A small number received thromboprophylaxis. Most patients received antithrombotic treatment. The most frequent bleeding complication was intracranial hemorrhage, primarily with isolated stroke. Overall mortality was 33.6% (74/220). Despite a wide range of COVID-19 severity, a high proportion had AT as a complication of non-severe disease. AT can affect different vascular territories; mortality is associated with stroke, intensive care unit stay, and severe COVID-19.
Collapse
Affiliation(s)
- Alejandra Castro-Varela
- Tecnologico de
Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey,
Nuevo Leon, Mexico
| | | | - Maria Fernanda Reyes-Chavez
- Tecnologico de
Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey,
Nuevo Leon, Mexico
- Unidad Experimental de Terapias
Avanzadas del Hospital Zambrano Hellion, TecSalud, San Pedro Garza Garcia, Nuevo Leon, Mexico
| | | | - Jose Gildardo Paredes-Vazquez
- Tecnologico de
Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey,
Nuevo Leon, Mexico
- Instituto de Cardiologia y Medicina
Vascular, TecSalud, Escuela de Medicina y Ciencias de la Salud,
Tecnologico de Monterrey, San Pedro Garza Garcia, Nuevo Leon, Mexico
| | - Eduardo Vazquez-Garza
- Tecnologico de
Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey,
Nuevo Leon, Mexico
- Unidad Experimental de Terapias
Avanzadas del Hospital Zambrano Hellion, TecSalud, San Pedro Garza Garcia, Nuevo Leon, Mexico
| | - Mauricio Castillo-Perez
- Tecnologico de
Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey,
Nuevo Leon, Mexico
| | | | - Arturo Martinez
- Tecnologico de
Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey,
Nuevo Leon, Mexico
| | - Ray Erick Ramos Cazares
- Tecnologico de
Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey,
Nuevo Leon, Mexico
| | - Jaime Guajardo
- Tecnologico de
Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey,
Nuevo Leon, Mexico
| | - Hector Lopez-de la Garza
- Tecnologico de
Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey,
Nuevo Leon, Mexico
| | | | - Hector Betancourt
- Tecnologico de
Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey,
Nuevo Leon, Mexico
| | | | - Jathniel Panneflek
- Unidad Experimental de Terapias
Avanzadas del Hospital Zambrano Hellion, TecSalud, San Pedro Garza Garcia, Nuevo Leon, Mexico
| | - Mario Alejandro Fabiani
- Instituto de Cardiologia y Medicina
Vascular, TecSalud, Escuela de Medicina y Ciencias de la Salud,
Tecnologico de Monterrey, San Pedro Garza Garcia, Nuevo Leon, Mexico
| | - Carlos Jerjes-Sanchez
- Tecnologico de
Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey,
Nuevo Leon, Mexico
- Unidad Experimental de Terapias
Avanzadas del Hospital Zambrano Hellion, TecSalud, San Pedro Garza Garcia, Nuevo Leon, Mexico
- Instituto de Cardiologia y Medicina
Vascular, TecSalud, Escuela de Medicina y Ciencias de la Salud,
Tecnologico de Monterrey, San Pedro Garza Garcia, Nuevo Leon, Mexico
| |
Collapse
|
49
|
Rettew A, Garrahy I, Rahimian S, Brown R, Sangha N. COVID-19 Coagulopathy. Life (Basel) 2024; 14:953. [PMID: 39202695 PMCID: PMC11355811 DOI: 10.3390/life14080953] [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: 07/03/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 09/03/2024] Open
Abstract
Coronavirus disease of 2019 (COVID-19) is the respiratory viral infection caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Despite being a primary respiratory illness, it is commonly complicated by systemic involvement of the vasculature leading to arterial and venous thrombosis. In this review, we will focus on the association between COVID-19 and thrombosis. We will highlight the pathophysiology of COVID-19 coagulopathy. The clinical manifestations of COVID-19 vasculopathy will be discussed with a focus on venous and arterial thromboembolic events. COVID-19 vasculopathy and disseminated intravascular coagulation (DIC) are distinguished within, as well as areas of controversy, such as "long COVID". Finally, the current professional guidelines on prevention and treatment of thrombosis associated with SARS-CoV-2 infection will be discussed.
Collapse
Affiliation(s)
| | - Ian Garrahy
- Tower Health System, Reading Hospital, West Reading, PA 19611, USA; (A.R.); (S.R.); (R.B.); (N.S.)
| | | | | | | |
Collapse
|
50
|
Yao Y, Sun L, Luo J, Qi W, Zuo X, Yang Z. The effect of long-term COVID-19 infection on maternal and fetal complications: a retrospective cohort study conducted at a single center in China. Sci Rep 2024; 14:17273. [PMID: 39068277 PMCID: PMC11283478 DOI: 10.1038/s41598-024-68184-2] [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: 01/27/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024] Open
Abstract
Investigate the effect of long-term COVID-19 on maternal and fetal complications. A retrospective cohort study was conducted. A total of 623 pregnant women who delivered in Kunming First People's Hospital from November 1, 2022 to July 31, 2023 were selected. By employing statistical methods, we compared the associations between maternal and fetal complications in pregnant women with acute COVID-19 during pregnancy, long-term COVID-19, and non-COVID-19 pregnant women. In the final 623 samples, there were 209 pregnant women with acute COVID-19, 72 pregnant women with long-term COVID-19, and 342 pregnant women without COVID-19. The epidemiological and clinical characteristics of all subjects were similar. Pregnant individuals who developed long-term COVID-19 during their pregnancy had an increased risk of experiencing gestational hypertension (OR 3.344, 95% CI 1.544-7.243), gestational diabetes mellitus (OR 2.301, 95% CI 1.290-4.102), and fetal intrauterine growth restriction (OR 2.817, 95% CI 1.385-5.952). Multivariate binary logistic regression analysis showed that this association remained consistent even after adjusting for confounders and performing subgroup analyses. Other maternal and fetal complications, such as premature rupture of membranes, preterm delivery, neonatal asphyxia, and transfer of neonates to NICU, did not exhibit statistically significant associations. After linear regression analysis, the platelet count (β: - 0.127, 95% CI - 0.001-0.000) of pregnant women with long-term COVID-19 was slightly lower than that of non-COVID-19 pregnant women, and the other coagulation parameters were not statistically significant. The incidence of gestational hypertension, gestational diabetes mellitus and fetal intrauterine growth restriction in pregnant women with long-term COVID-19 is significantly increased, but it does not further increase the coagulation status.
Collapse
Affiliation(s)
- Yang Yao
- Gynecology and Obstetrics, Kunming First People's Hospital, Kunming City, 650000, Yunnan Province, China
| | - Lanxu Sun
- Gynecology and Obstetrics, Kunming First People's Hospital, Kunming City, 650000, Yunnan Province, China
| | - Jing Luo
- Gynecology and Obstetrics, Kunming First People's Hospital, Kunming City, 650000, Yunnan Province, China
| | - Wenjin Qi
- Gynecology and Obstetrics, The First Affiliated Hospital of Kunming Medical University, Kunming City, 650000, Yunnan Province, China.
| | - Xin Zuo
- Gynecology and Obstetrics, Kunming First People's Hospital, Kunming City, 650000, Yunnan Province, China
| | - Zenglin Yang
- Gynecology and Obstetrics, Kunming First People's Hospital, Kunming City, 650000, Yunnan Province, China
| |
Collapse
|