251
|
Gao S, Tang AT, Wang M, Buchholz DW, Imbiakha B, Yang J, Chen X, Hewins P, Mericko-Ishizuka P, Leu NA, Sterling S, August A, Jurado KA, Morrisey EE, Aguilar-Carreno H, Kahn ML. Endothelial SARS-CoV-2 infection is not the underlying cause of COVID-19-associated vascular pathology in mice. Front Cardiovasc Med 2023; 10:1266276. [PMID: 37823176 PMCID: PMC10562591 DOI: 10.3389/fcvm.2023.1266276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 09/05/2023] [Indexed: 10/13/2023] Open
Abstract
Endothelial damage and vascular pathology have been recognized as major features of COVID-19 since the beginning of the pandemic. Two main theories regarding how severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) damages endothelial cells and causes vascular pathology have been proposed: direct viral infection of endothelial cells or indirect damage mediated by circulating inflammatory molecules and immune mechanisms. However, these proposed mechanisms remain largely untested in vivo. In the present study, we utilized a set of new mouse genetic tools developed in our lab to test both the necessity and sufficiency of endothelial human angiotensin-converting enzyme 2 (hACE2) in COVID-19 pathogenesis. Our results demonstrate that endothelial ACE2 and direct infection of vascular endothelial cells do not contribute significantly to the diverse vascular pathology associated with COVID-19.
Collapse
Affiliation(s)
- Siqi Gao
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - Alan T. Tang
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - Min Wang
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - David W. Buchholz
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Brian Imbiakha
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Jisheng Yang
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - Xiaowen Chen
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - Peter Hewins
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Patricia Mericko-Ishizuka
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - N. Adrian Leu
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Stephanie Sterling
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Avery August
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Kellie A. Jurado
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Edward E. Morrisey
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, United States
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Hector Aguilar-Carreno
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Mark L. Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
252
|
Pincavitch JD, Pisquiy JJ, Wen S, Bryan N, Ammons J, Makwana P, Dietz MJ. Thirty-Day Mortality and Complication Rates in Total Joint Arthroplasty After a Recent COVID-19 Diagnosis: A Retrospective Cohort in the National COVID Cohort Collaborative (N3C). J Bone Joint Surg Am 2023; 105:1362-1372. [PMID: 37352338 PMCID: PMC11317089 DOI: 10.2106/jbjs.22.01317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/25/2023]
Abstract
BACKGROUND The risk of postoperative complications in patients who had a positive COVID-19 test prior to a total joint arthroplasty (TJA) is unknown. The purpose of this investigation was to study the complications and mortality associated with a recent COVID-19 diagnosis prior to TJA. METHODS Patients undergoing primary and revision total hip arthroplasties (THAs) or total knee arthroplasties (TKAs) were identified using the National COVID Cohort Collaborative (N3C) Data Enclave. Patients were divided into a COVID-19-positive group (positive polymerase chain reaction [PCR] test, clinical diagnosis, or positive antibody test) and a COVID-19-negative group, and the time from diagnosis was noted. There was no differentiation between severity or acuity of illness available. The postoperative complications reviewed included venous thromboembolism, pneumonia, acute myocardial infarction, readmission rates, and 30-day mortality rates. RESULTS A total of 85,047 patients who underwent elective TJA were included in this study, and 3,516 patients (4.13%) had had a recent positive COVID-19 diagnosis. Patients diagnosed with COVID-19 at 2 weeks prior to TJA were at increased risk of pneumonia (odds ratio [OR], 2.46), acute myocardial infarction (OR, 2.90), sepsis within 90 days (OR, 2.63), and 30-day mortality (OR, 10.61). CONCLUSIONS Patients with a recent COVID-19 diagnosis prior to TJA are at greater risk of postoperative complications including 30-day mortality. Our analysis presents critical data that should be considered prior to TJA in patients recently diagnosed with COVID-19. LEVEL OF EVIDENCE Prognostic Level III . See Instructions for Authors for a complete description of levels of evidence.
Collapse
Affiliation(s)
- Jami D Pincavitch
- Department of Orthopaedics, West Virginia University, Morgantown, West Virginia
| | - John J Pisquiy
- Department of Orthopaedics, West Virginia University, Morgantown, West Virginia
| | - Sijin Wen
- Department of Epidemiology and Biostatistics, School of Public Health, West Virginia University, Morgantown, West Virginia
| | - Nicole Bryan
- Section of Infectious Diseases, Department of Medicine, West Virginia University, Morgantown, West Virginia
| | - Jeffrey Ammons
- West Virginia Clinical & Translational Science Institute, West Virginia University, Morgantown, West Virginia
| | - Priyal Makwana
- Department of Scientific Computing and Data, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Matthew J Dietz
- Department of Orthopaedics, West Virginia University, Morgantown, West Virginia
| |
Collapse
|
253
|
Forgács R, Bokrétás GP, Monori Z, Molnár Z, Ruszkai Z. Thromboelastometry-Guided Individualized Fibrinolytic Treatment for COVID-19-Associated Severe Coagulopathy Complicated by Portal Vein Thrombosis: A Case Report. Biomedicines 2023; 11:2463. [PMID: 37760902 PMCID: PMC10525483 DOI: 10.3390/biomedicines11092463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
COVID-19-associated coagulopathy (CAC), mainly characterized by hypercoagulability leading to micro- and macrovascular thrombotic events due to the fibrinolysis shutdown phenomenon, is a life-threatening complication of severe SARS-CoV-2 infection. However, optimal criteria to assess patients with the highest risk for progression of severe CAC are still unclear. Bedside point-of-care viscoelastic testing (VET) appears to be a promising tool to recognize CAC, to support the appropriate therapeutic decisions, and to monitor the efficacy of the treatment. The ClotPro VET has the potential to reveal fibrinolysis resistance indicated by a clot lysis time (LT) > 300 s on the TPA-test. We present a case of severe SARS-CoV-2 infection complicated by CAC-resulting portal vein thrombosis (PVT) and subsequent liver failure despite therapeutic anticoagulation. Since fibrinolysis shutdown (LT > 755 s) caused PVT, we performed a targeted systemic fibrinolytic therapy. We monitored the efficacy of the treatment with repeated TPA assays every three hours, while the dose of recombinant plasminogen activator (rtPA) was adjusted until fibrinolysis shutdown completely resolved and portal vein patency was confirmed by an ultrasound examination. Our case report highlights the importance of VET-guided personalized therapeutic approach during the care of severely ill COVID-19 patients, in order to appropriately treat CAC.
Collapse
Affiliation(s)
- Robin Forgács
- Department of Anesthesiology and Intensive Therapy, Flór Ferenc Hospital Kistarcsa, 2143 Kistarcsa, Hungary; (R.F.); (G.P.B.); (Z.M.); (Z.R.)
| | - Gergely Péter Bokrétás
- Department of Anesthesiology and Intensive Therapy, Flór Ferenc Hospital Kistarcsa, 2143 Kistarcsa, Hungary; (R.F.); (G.P.B.); (Z.M.); (Z.R.)
| | - Zoltán Monori
- Department of Anesthesiology and Intensive Therapy, Flór Ferenc Hospital Kistarcsa, 2143 Kistarcsa, Hungary; (R.F.); (G.P.B.); (Z.M.); (Z.R.)
| | - Zsolt Molnár
- Department of Anesthesiology and Intensive Therapy, Semmelweis University, 1082 Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, 1082 Budapest, Hungary
- Department of Anaesthesiology and Intensive Therapy, Faculty of Medicine, Poznan University of Medical Sciences, 60-005 Poznan, Poland
| | - Zoltán Ruszkai
- Department of Anesthesiology and Intensive Therapy, Flór Ferenc Hospital Kistarcsa, 2143 Kistarcsa, Hungary; (R.F.); (G.P.B.); (Z.M.); (Z.R.)
- Department of Anesthesiology and Intensive Therapy, Semmelweis University, 1082 Budapest, Hungary
| |
Collapse
|
254
|
Mohseni Afshar Z, Tavakoli Pirzaman A, Hosseinzadeh R, Babazadeh A, Taghizadeh Moghadam MA, Miri SR, Sio TT, Sullman MJM, Barary M, Ebrahimpour S. Anticoagulant therapy in COVID-19: A narrative review. Clin Transl Sci 2023; 16:1510-1525. [PMID: 37326220 PMCID: PMC10499427 DOI: 10.1111/cts.13569] [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: 04/12/2022] [Revised: 05/24/2023] [Accepted: 05/27/2023] [Indexed: 06/17/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), can manifest itself in several ways, including coagulopathy and thrombosis. These complications can be the first and sometimes only manifestations of SARS-CoV-2 infection and can occur early or late in the course of the disease. However, these symptoms are more prevalent in hospitalized patients with venous thromboembolism, particularly those admitted to intensive care units. Moreover, various forms of arterial and venous thrombosis, or micro- or macro-vasculature embolisms, have been reported during the current pandemic. They have led to harmful consequences, such as neurological and cardiac events, nearly all resulting from the hypercoagulable state caused by this viral infection. The severe hypercoagulability observed in patients with COVID-19 accounts for most cases of the disease that become critical. Therefore, anticoagulants seem to be one of the most vital therapeutics for treating this potentially life-threatening condition. In the current paper, we present a thorough review of the pathophysiology of COVID-19-induced hypercoagulable state and the use of anticoagulants to treat SARS-CoV-2 infections in different patient groups, as well as their pros and cons.
Collapse
Affiliation(s)
- Zeinab Mohseni Afshar
- Clinical Research Development Center, Imam Reza HospitalKermanshah University of Medical SciencesKermanshahIran
| | | | | | - Arefeh Babazadeh
- Infectious Diseases and Tropical Medicine Research CenterHealth Research Institute, Babol University of Medical SciencesBabolIran
| | | | - Seyed Rouhollah Miri
- Cancer Research CenterCancer Institute of Iran, Tehran University of Medical ScienceTehranIran
| | - Terence T. Sio
- Department of Radiation OncologyMayo ClinicPhoenixArizonaUSA
| | - Mark J. M. Sullman
- Department of Social SciencesUniversity of NicosiaNicosiaCyprus
- Department of Life and Health SciencesUniversity of NicosiaNicosiaCyprus
| | - Mohammad Barary
- Student Research Committee, Virtual School of Medical Education and ManagementShahid Beheshti University of Medical SciencesTehranIran
| | - Soheil Ebrahimpour
- Infectious Diseases and Tropical Medicine Research CenterHealth Research Institute, Babol University of Medical SciencesBabolIran
| |
Collapse
|
255
|
Mardani M, Mohammadshahi J, Teimourpour R. Outcomes of COVID-19 in immunocompromised patients: a single center experience. Virusdisease 2023; 34:373-382. [PMID: 37780900 PMCID: PMC10533436 DOI: 10.1007/s13337-023-00832-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/18/2023] [Indexed: 10/03/2023] Open
Abstract
Malignancy, bone marrow and organ transplantation are associated with deficient and defective immune systems. Immunocompromised patients are at risk for severe and chronic complication of COVID-19 infection. However, the pathogenesis, diagnosis and management of this comorbidity remain to be elucidated. The purpose of the present study was to describe key aspects of COVID-19 infection in immunocompromised patients. In this retrospective, cross-sectional study, lab findings and outcomes of 418 COVID-19 patients with secondary immunodeficiency disorders admitted to Taleghani Hospital in Tehran, from March 2020 to September 2022 were investigated. Of the 418 immunocompromised patients with COVID-19, 236 (56.5%) were male and the median age of all studied patients was 56.6 ± 16.4 with range of 14 to 92 years. Totally, 198 (47.4%) of the patients died during hospitalization. Remdesivir was used for treatment of all patients. Mortality rate among patients admitted to ICU ward (86.8%) was significantly higher than non ICU admission (p < 0.001). The death rate in patients with CKD was substantially higher than other underlying disease (p < 0.001). In terms of laboratory finding, there was a significant relationship between ICU admission and worse outcome with WBC count (HR = 1.94, 95% CI = 1. 46-2.59, p < 0.001), PMN count (HR = 1.93, 95% CI = 1.452.56, p < 0.001), Hb (HR = 1.49, 95% CI = 1.042.13, p = 0.028), AST (HR = 2.55, 95% CI = 1.913.41, p < 0.001), BUN (HR = 2.56, 95% CI = 2.063.69, p < 0.001), Cr (HR = 2.63, 95% CI = 1.89-3.64, p < 0.001), Comorbidities index (HR = 1.71, 95% CI = 1.29-2.27, p < 0.001) and aging (HR = 1.91, 95% CI = 1.4-2.54, p < 0.001). Immunocompromised status increased the risk of mortality or worse outcome in patients diagnosed with COVID-19. Our finding showed outcome predicting markers in whom the waned immune system encounter new emerging disease and improved our understanding of COVID-19 virus behavior in immunocompromised individuals.
Collapse
Affiliation(s)
- Masoud Mardani
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical, Tehran, Iran
| | - Jafar Mohammadshahi
- Ardabil University of medical science, Ardabil, Iran
- Department of Infectious Diseases, School of Medicine, Ardabil University of Medical Science, Ardabil, Iran
| | - Roghayeh Teimourpour
- Ardabil University of medical science, Ardabil, Iran
- Department of Microbiology, School of Medicine, Ardabil University of Medical Science, Ardabil, Iran
| |
Collapse
|
256
|
Liang Y, Liu L, Liang B. COVID-19 susceptibility and severity for dyslipidemia: A mendelian randomization investigation. Heliyon 2023; 9:e20247. [PMID: 37809746 PMCID: PMC10560011 DOI: 10.1016/j.heliyon.2023.e20247] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 09/14/2023] [Accepted: 09/14/2023] [Indexed: 10/10/2023] Open
Abstract
Background The severe acute respiratory syndrome coronavirus in 2019 (COVID-19) is still spreading and causing deaths worldwide, which further increased the burden of chronic diseases. Dyslipidemia is a common metabolic syndrome, which is a major risk factor for cardiovascular disease. However, studies on whether there is a direct causal relationship between COVID-19 and the exacerbation of hyperlipidemia are still scarce. Methods Two-sample Mendelian randomization was conducted using publicly available summary statistics from independent cohorts of European ancestry. For COVID-19 and hyperlipidemia, we used data from the ieu open GWAS project database. Inverse variance-weighted, mendelian randomization Egger, weighted median, simple mode, and weighted mode mendelian randomization analyses were performed, together with a range of sensitivity analyses. Results There is no direct causal relationship between COVID-19 and dyslipidemia, regardless of COVID-19 severity or either dyslipidemic outcome. In combination with previous studies, the reason for the clinical outcome that COVID-19 increased the burden of dyslipidemia may be due to the exacerbation of pre-existing disease caused by COVID-19. Conclusions COVID-19 has no direct causal relationship with dyslipidemia.
Collapse
Affiliation(s)
- Yi Liang
- Geriatric Medicine Department, The Second Hospital of Traditional Chinese Medicine in Sichuan Province, Chengdu, China
| | - Liang Liu
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Bo Liang
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
257
|
Hakami A, Altubayqi T, Qadah EA, Zogel B, Alfaifi SM, Refaei E, Sayed A, Alhazmi L, Sayegh M, Alamer A, Areeshi AS, Hakami D. Biochemical Analysis of Ferritin and D-dimer in COVID-19 Survivors and Non-survivors. Cureus 2023; 15:e45389. [PMID: 37854756 PMCID: PMC10579969 DOI: 10.7759/cureus.45389] [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: 09/17/2023] [Indexed: 10/20/2023] Open
Abstract
Background COVID-19 is a major cause of illness and mortality. The management of COVID-19-related illnesses might change if variables connected to their severity and the requirement for ICU admission could be found. The severity of COVID-19 might be efficiently predicted with several laboratory measures, such as ferritin levels and D-dimer analysis. Objectives This study aimed to evaluate the association between serum D-dimer and ferritin levels and their effects on mortality in patients with COVID-19. Methods This retrospective observational study included all patients with positive real-time polymerase chain reaction (RT-PCR) results for COVID-19 who were hospitalized in the Ministry of Health South Al-Qunfudah General Hospital between March and September 30, 2020. Their laboratory parameters, serum D-dimer, and ferritin levels were evaluated. IBM SPSS Statistics for Windows, Version 26.0 (released 2019; IBM Corp., Armonk, New York, United States) was used to analyze the data. Results A total of 318 COVID-19 patients were analyzed; 56.9% (n=181) were male and 43.1% (n=137) were female. Of these, 78.6% (n=250) survived, including 58% of men and 42% of women. The mean D-dimer was 2.1 mcg/mL (SD=3.16) and the mean ferritin was 698.59 ng/mL (SD=603.11). Non-recovered patients were substantially older (66.16 years old) and had higher D-dimer (5.46) mcg/mL and ferritin levels (992.96) ng/mL. Intubation length and gender did not affect survival. Of the non-survivors, 95.6% (n=239) were admitted to the ICU, and 50% (n=34) required mechanical ventilation. Conclusions COVID-19 infection mortality dramatically increased with older age and increased mean ferritin and plasma D-dimer values, which were significantly higher in COVID-19 non-survivors than in survivors. Therefore, assessing and monitoring these laboratory markers in the early stages of the disease may have a significant impact on preventing disease progression and death.
Collapse
Affiliation(s)
- Abdulrahman Hakami
- Department of Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Tahani Altubayqi
- Department of Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Entsar A Qadah
- Department of Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Basem Zogel
- Department of Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Samar M Alfaifi
- Department of Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Eman Refaei
- Department of Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Ahmed Sayed
- Department of Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Luai Alhazmi
- Department of Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Maram Sayegh
- Department of Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Abdullah Alamer
- Department of Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Areej S Areeshi
- Department of Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Duaa Hakami
- Department of Medicine, Jazan General Hospital, Jazan Health Affairs, Ministry of Health, Jazan, SAU
| |
Collapse
|
258
|
Kumar Y, Kumari A, Kumar T, Jha K, Zabihullah M. Role of Hematological Parameters in the Grading of COVID-19 and a Model to Predict the Outcome in Inpatients. Cureus 2023; 15:e45276. [PMID: 37846240 PMCID: PMC10576849 DOI: 10.7759/cureus.45276] [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: 09/14/2023] [Indexed: 10/18/2023] Open
Abstract
Introduction Human coronaviruses, identified in the 1960s, are known culprits of respiratory infections. Classified into alpha, beta, gamma, and delta subgroups, these viruses have the capacity to transition from animal reservoirs to causing severe respiratory ailments in humans. Notable outbreaks like the 2003 severe acute respiratory distress syndrome (SARS) epidemic and the ongoing coronavirus disease 2019 (COVID-19) pandemic underscore the recurring emergence of novel coronaviruses with severe human infection potential. COVID-19, driven by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has rapidly become a leading global cause of severe acute respiratory syndrome. Immune system disruptions and cytokine imbalances contribute to severe cases, necessitating early diagnosis and precise severity assessment. Methodology This retrospective cross-sectional study encompassed 211 COVID-19 patients admitted to AIIMS Patna from May to July 2020. Clinical and hematological parameters, including neutrophils, eosinophils, basophils, lymphocytes, monocytes, red and white blood cell counts, platelet count, C-reactive protein (CRP), serum ferritin, and d-dimer, were meticulously recorded. Patients were categorized into non-severe and severe groups using the National Early Warning Score (NEWS) 2. Results Our findings underscore the pivotal role of hematological markers in gauging COVID-19 severity. Notably, markers such as neutrophil-to-lymphocyte ratio (NLR), derived NLR, lymphocyte monocyte ratio, platelet lymphocyte ratio, d-dimer, CRP, and serum ferritin exhibited notable elevation in severe cases. Survival analysis further established the predictive potential of these markers in assessing disease progression and mortality risk. We advocate for the integration of these markers into existing severity assessment frameworks to foster objective clinical evaluations. Conclusion In conclusion, our study unravels the intricate connection between COVID-19 severity and hematological parameters. We emphasize the early warning capabilities of NLR, derived NLR, platelet lymphocyte ratio, and other markers in predicting disease progression. This research underscores the imperative need to incorporate hematological markers into the evaluation of COVID-19 severity, thereby providing invaluable insights for enhancing clinical practice and patient outcomes.
Collapse
Affiliation(s)
- Yogesh Kumar
- Physiology, All India Institute of Medical Sciences, Patna, Patna, IND
| | - Amita Kumari
- Physiology, All India Institute of Medical Sciences, Deoghar, Deoghar, IND
| | - Tribhuwan Kumar
- Physiology, All India Institute of Medical Sciences, Patna, Patna, IND
| | - Kamlesh Jha
- Physiology, All India Institute of Medical Sciences, Patna, Patna, IND
| | - Md Zabihullah
- Physiology, All India Institute of Medical Sciences, Patna, Patna, IND
| |
Collapse
|
259
|
Shi Y, Zheng Z, Wang P, Wu Y, Liu Y, Liu J. Development and validation of a predicted nomogram for mortality of COVID-19: a multicenter retrospective cohort study of 4,711 cases in multiethnic. Front Med (Lausanne) 2023; 10:1136129. [PMID: 37724179 PMCID: PMC10505438 DOI: 10.3389/fmed.2023.1136129] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 08/22/2023] [Indexed: 09/20/2023] Open
Abstract
Background Coronavirus disease 2019 (COVID-19) is an infectious disease spreading rapidly worldwide. As it quickly spreads and can cause severe disease, early detection and treatment may reduce mortality. Therefore, the study aims to construct a risk model and a nomogram for predicting the mortality of COVID-19. Methods The original data of this study were from the article "Neurologic Syndromes Predict Higher In-Hospital Mortality in COVID-19." The database contained 4,711 multiethnic patients. In this secondary analysis, a statistical difference test was conducted for clinical demographics, clinical characteristics, and laboratory indexes. The least absolute shrinkage and selection operator (LASSO) and multivariate logistic regression analysis were applied to determine the independent predictors for the mortality of COVID-19. A nomogram was conducted and validated according to the independent predictors. The area under the curve (AUC), the calibration curve, and the decision curve analysis (DCA) were carried out to evaluate the nomogram. Results The mortality of COVID-19 is 24.4%. LASSO and multivariate logistic regression analysis suggested that risk factors for age, PCT, glucose, D-dimer, CRP, troponin, BUN, LOS, MAP, AST, temperature, O2Sats, platelets, Asian, and stroke were independent predictors of CTO. Using these independent predictors, a nomogram was constructed with good discrimination (0.860 in the C index) and internal validation (0.8479 in the C index), respectively. The calibration curves and the DCA showed a high degree of reliability and precision for this clinical prediction model. Conclusion An early warning model based on accessible variates from routine clinical tests to predict the mortality of COVID-19 were conducted. This nomogram can be conveniently used to facilitate identifying patients who might develop severe disease at an early stage of COVID-19. Further studies are warranted to validate the prognostic ability of the nomogram.
Collapse
Affiliation(s)
- Yuchen Shi
- Center for Coronary Artery Disease (CCAD), Beijing Anzhen Hospital, Capital Medical University, and Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Ze Zheng
- Center for Coronary Artery Disease (CCAD), Beijing Anzhen Hospital, Capital Medical University, and Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Ping Wang
- Center for Coronary Artery Disease (CCAD), Beijing Anzhen Hospital, Capital Medical University, and Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Yongxin Wu
- Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yanci Liu
- Center for Coronary Artery Disease (CCAD), Beijing Anzhen Hospital, Capital Medical University, and Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Jinghua Liu
- Center for Coronary Artery Disease (CCAD), Beijing Anzhen Hospital, Capital Medical University, and Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| |
Collapse
|
260
|
DE Vito A, Saderi L, Fiore V, Geremia N, Princic E, Fanelli C, Muredda AA, Panu Napodano C, Moi G, Maida I, Fois AG, Sotgiu G, Madeddu G, Babudieri S. Early treatment with low-molecular-weight heparin reduces mortality rate in SARS-CoV-2 patients. Panminerva Med 2023; 65:286-291. [PMID: 35622392 DOI: 10.23736/s0031-0808.22.04572-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Since the beginning of the SARS-CoV-2 pandemic, millions of people have been infected and died. Different therapeutic approaches have been recommended, but only a few have shown clinical advantages. Low-molecular-weight heparin (LMWH) has been recommended to prevent COVID-19-related thrombo-embolic events. We aimed to evaluate the impact of early treatment with LMWH on hospital admission and death in patients with SARS-CoV-2 infection. METHODS We conducted an observational monocentric retrospective study to evaluate the preventive role of LMWH on the mortality rate of COVID-19 patients. SARS-CoV-2 infected patients were recruited from the beginning of the Italian epidemic to March 31, 2021. We excluded patients with missing data and those chronically exposed to LMWH. Treatment prescription was based on international and national guidelines and modified depending on clinical presentation and drug-drug interactions. RESULTS Seven hundred thirty-four SARS-CoV-2 infected patients were recruited, with 357 (48.6%) males and a median (IQR) age of 77.9 (65-85.7) years. 77.5% of people developed SARS-CoV-2-related symptoms and 62.8% were admitted to the hospital, and 20.2% died. Four hundred ninety-two (67%) started LMWH. In particular, 296 (40.3%) were treated within five days since symptoms onset. At logistic regression, early LMWH therapy was associated with lower mortality. Furthermore, remdesivir treatment showed a lower risk of death. On the contrary, age, BMI>30 kg/m2, neurological diseases, fever or dyspnea were associated with an increased risk of death. CONCLUSIONS Early treatment with LMWH was associated with lower mortality in our cohort. Further studies are needed to better assess the role of wider LMWH administration in terms of timing and regimen dose.
Collapse
Affiliation(s)
- Andrea DE Vito
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy -
| | - Laura Saderi
- Unit of Clinical Epidemiology and Medical Statistics, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Vito Fiore
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Nicholas Geremia
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Elija Princic
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Chiara Fanelli
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Alberto A Muredda
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Catello Panu Napodano
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Giulia Moi
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Ivana Maida
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Alessandro G Fois
- Unit of Respiratory Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Giovanni Sotgiu
- Unit of Clinical Epidemiology and Medical Statistics, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Giordano Madeddu
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Sergio Babudieri
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
261
|
Kokkoris S, Kanavou A, Kremmydas P, Katsaros D, Karageorgiou S, Gkoufa A, Georgakopoulou VE, Spandidos DA, Giannopoulos C, Kardamitsi M, Routsi C. Temporal evolution of laboratory characteristics in patients critically ill with COVID‑19 admitted to the intensive care unit (Review). MEDICINE INTERNATIONAL 2023; 3:52. [PMID: 37810906 PMCID: PMC10557099 DOI: 10.3892/mi.2023.112] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/12/2023] [Indexed: 10/10/2023]
Abstract
In the context of coronavirus disease 2019 (COVID-19), laboratory medicine has played a crucial role in both diagnosis and severity assessment. Although the importance of baseline laboratory findings has been extensively reported, data regarding their evolution over the clinical course are limited. The aim of the present narrative review was to provide the dynamic changes of the routine laboratory variables reported in patients with severe COVID-19 over the course of their critical illness. A search was made of the literature for articles providing data on the time-course of routine laboratory tests in patients with severe COVID-19 during their stay in the intensive care unit (ICU). White blood cell, neutrophil and lymphocyte counts, neutrophil to lymphocyte ratio, platelet counts, as well as D-dimer, fibrinogen, C-reactive protein, lactate dehydrogenase and serum albumin levels were selected as disease characteristics and routine laboratory parameters. A total of 25 research articles reporting dynamic trends in the aforementioned laboratory parameters over the clinical course of severe COVID-19 were identified. During the follow-up period provided by each study, the majority of the laboratory values remained persistently abnormal in both survivors and non-survivors. Furthermore, in the majority of studies, the temporal trends of laboratory values distinctly differentiated patients between survivors and non-survivors. In conclusion, there are distinct temporal trends in selected routine laboratory parameters between survivors and non-survivors with severe COVID-19 admitted to the ICU, indicating their importance in the prognosis of clinical outcome.
Collapse
Affiliation(s)
- Stelios Kokkoris
- First Department of Intensive Care, Evangelismos Hospital, Medical School, National and Kapodistrian University of Athens, 10676 Athens, Greece
| | - Angeliki Kanavou
- First Department of Intensive Care, Evangelismos Hospital, Medical School, National and Kapodistrian University of Athens, 10676 Athens, Greece
| | - Panagiotis Kremmydas
- First Department of Intensive Care, Evangelismos Hospital, Medical School, National and Kapodistrian University of Athens, 10676 Athens, Greece
| | - Dimitrios Katsaros
- First Department of Intensive Care, Evangelismos Hospital, Medical School, National and Kapodistrian University of Athens, 10676 Athens, Greece
| | - Stavros Karageorgiou
- First Department of Intensive Care, Evangelismos Hospital, Medical School, National and Kapodistrian University of Athens, 10676 Athens, Greece
| | - Aikaterini Gkoufa
- First Department of Intensive Care, Evangelismos Hospital, Medical School, National and Kapodistrian University of Athens, 10676 Athens, Greece
- Department of Infectious Diseases, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | | | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Charalampos Giannopoulos
- First Department of Intensive Care, Evangelismos Hospital, Medical School, National and Kapodistrian University of Athens, 10676 Athens, Greece
| | - Marina Kardamitsi
- First Department of Intensive Care, Evangelismos Hospital, Medical School, National and Kapodistrian University of Athens, 10676 Athens, Greece
| | - Christina Routsi
- First Department of Intensive Care, Evangelismos Hospital, Medical School, National and Kapodistrian University of Athens, 10676 Athens, Greece
| |
Collapse
|
262
|
Simsek Yurt N, Ocak M. Ferritin/lymphocyte percentage ratio to predict the severity and mortality of COVID-19. Malawi Med J 2023; 35:183-189. [PMID: 38362294 PMCID: PMC10865067 DOI: 10.4314/mmj.v35i3.8] [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: 02/17/2024] Open
Abstract
Objective In this study, we aimed to investigate the relationship between ferritin/lymphocyte percentage ratio (FLPR) with clinical and radiological disease severity and mortality in COVID-19 patients. Methods This retrospective study was conducted with patients who had polymerase chain reaction positive results for COVID-19. We calculated FLPRs from laboratory tests taken during emergency clinic admission. The relationship between chest computed tomography (CT) scores, disease severity, and 30-day mortality with FLPR was evaluated. Results Our study included 309 patients. 30-day mortality occurred in 12.3% (n=38) of the patients. A statistically significant association was found between FLPR and clinical disease severity (p <0.001). In the post hoc analysis, the difference was caused by the critical and severe groups and FLPR was significantly higher in these groups. A significant correlation was found between CT scores and FLPR (r=0.496, p<0.001). Logistic regression analysis revealed that hypertension, smoking, C-reactive protein (CRP), and FLPR levels were independent risk factors for mortality in COVID-19 patients. In the receiver operating characteristics curve analysis, determined the predictive value and the optimal cut-off value of FLPR. The areas under the curve of WBC, lymphocyte, neutrophil, ferritin, CRP, FLPR were found 0.707, 0.233, 0.735, 0.878, 0.831, 0.924 (p<0.001), respectively. This analysis showed that the FLPR can predict 30-day mortality better than the other biomarkers in the comparison. When the optimal cut-off value of FLPR is 42.4, the sensitivity is 84.2% and specificity is 86.7%. Conclusion FLPR can be used as an independent biomarker of disease severity and mortality in COVID-19.
Collapse
Affiliation(s)
- Nur Simsek Yurt
- Clinic of Family Medicine, Samsun Training and Research Hospital, Samsun, Turkey
| | - Metin Ocak
- Clinic of Emergency Medicine, Samsun Gazi State Hospital, Samsun, Turkey
| |
Collapse
|
263
|
Lewczuk T, Kazimierczyk R, Sobkowicz B, Lisowska A. Pulmonary embolism in COVID-19 pneumonia patients admitted to temporary hospital - The follow-up study. Adv Med Sci 2023; 68:270-275. [PMID: 37633116 DOI: 10.1016/j.advms.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/04/2023] [Accepted: 08/18/2023] [Indexed: 08/28/2023]
Abstract
PURPOSE Coronavirus disease 2019 (COVID-19) is linked with major coagulation disorders, especially higher risk of developing pulmonary embolism (PE). Our study summarizes COVID-19 patients' management with concomitant PE during the first weeks of pandemic and underlines the importance of D-dimer concentration assessment at admission in terms of prognosis. MATERIAL AND METHODS Study group consisted of 107 outpatients (mean age 68.91 ± 12.83 years) admitted to the Temporary COVID-19 Hospital in Bialystok, Poland with confirmed severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and suspicion of PE based on elevated D-dimer concentration (>500 μg/l) and/or low saturation rate (<90%). The clinical follow-up lasted 6 months. Death or re-hospitalization were used as composite clinical endpoint (CEP). RESULTS Cumulative incidence of PE was 62.3% (73/107 patients). Most of the patients were in the intermediate PE risk group according to the pulmonary embolism severity index (PESI) score. The mean total computed tomography (CT) lung involvement of COVID-19 findings was 48.42 ± 27.71%. Neither D-dimers nor NT-proBNP concentrations correlated significantly with the percentage of lung abnormalities in CT. Patients with baseline D-dimer concentration higher than 1429 μg/l had worse prognosis in 6-months observation, log-rank test, p = 0.009. CONCLUSIONS Ongoing SARS-CoV-2 infection along with massive involvement of lung tissue and concomitant thrombi in pulmonary arteries are challenging for physicians. It seems that simple D-dimer concentration assessment at admission may be a helpful tool not only to predict PE but also to estimate the long-term prognosis.
Collapse
Affiliation(s)
- Tomasz Lewczuk
- Department of Cardiology, Medical University of Bialystok, Bialystok, Poland
| | | | - Bozena Sobkowicz
- Department of Cardiology, Medical University of Bialystok, Bialystok, Poland
| | - Anna Lisowska
- Department of Cardiology, Medical University of Bialystok, Bialystok, Poland.
| |
Collapse
|
264
|
Zhai K, Xu X, Zhang P, Wei S, Li J, Wu X, Gao B, Zhang Y, Li Y. Venovenous extracorporeal membrane oxygenation for coronavirus disease 2019 patients: A systematic review and meta-analysis. Perfusion 2023; 38:1107-1122. [PMID: 35608047 DOI: 10.1177/02676591221104302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Although the application of venovenous extracorporeal membrane oxygenation (VV-ECMO) in coronavirus disease 2019 (COVID-19) patients with acute respiratory distress syndrome (ARDS) is accumulating, the feasibility and safety of this therapy remain controversial. We aimed to evaluate the effect of VV-ECMO in the treatment of these patients. METHODS A comprehensive literature search was performed using PubMed, Embase, the Cochrane Library, and International Clinical Trials Registry Platform databases through November 2021. According to the inclusion and exclusion criteria, the included studies were screened, and meta-analysis was performed by R software (version 4.0.2). RESULTS Forty-two studies including 2037 COVID-19 patients supported with VV-ECMO due to ARDS were identified. The pooled analysis revealed that 30-, 60-, and 90-day mortality among patients were respectively 46% (95% CI 37%-57%, I2 = 66%), 46% (95% CI 30%-70%, I2 = 93%), and 49% (95% CI 43%-58%, I2 = 52%), and the pooled incidence rate of in-hospital mortality, major bleeding, hemorrhagic stroke, thrombosis, pulmonary embolism, deep venous thrombosis, and renal replacement therapy were respectively 35%, 39%, 11%, 40%, 15%, 21%, and 44%. CONCLUSION Although COVID-19 patients may have a higher risk of bleeding, hemorrhagic stroke, and acute kidney injury during ECMO therapy, the survival rate was more than half of the cases. Our data may support the application of VV-ECMO in COVID-19 patients.
Collapse
Affiliation(s)
- Kerong Zhai
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Laboratory of Extracorporeal Life Support, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Xu Xu
- Department of Anesthesiology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Pengbin Zhang
- Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Shilin Wei
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Laboratory of Extracorporeal Life Support, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Jian Li
- Laboratory of Extracorporeal Life Support, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Xiangyang Wu
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Bingren Gao
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Yanhua Zhang
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Yongnan Li
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Laboratory of Extracorporeal Life Support, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| |
Collapse
|
265
|
Filippi L, Turcato G, Milan M, Barbar S, Miozzo E, Zaboli A, Tonello D, Milazzo D, Marchetti M, Cuppini S, Prandoni P. Long term follow-up of a multicentre cohort of COVID-19 patients with pulmonary embolism: Anticoagulation management and outcomes. Thromb Res 2023; 229:73-76. [PMID: 37419005 PMCID: PMC10289817 DOI: 10.1016/j.thromres.2023.06.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 06/13/2023] [Accepted: 06/16/2023] [Indexed: 07/09/2023]
Abstract
BACKGROUND AND AIMS Pulmonary embolism (PE) is a frequent complication in COVID19 hospitalized patients. Inflammatory storm and endothelial dysfunction due to the virus seem to be the two major risk factors for PE. Consequently, PE related to COVID19 could be consider as triggered by a transient inflammatory acute phase and treated for no longer than 3 months. However, few data are available on management of anticoagulation and risk of venous thromboembolic (VTE) recurrences in these patients and guidelines are still undefined. Aim of the present study is to evaluate the long-term follow-up of a cohort of covid-19 patients with PE. METHODS We conducted a retrospective multicenter study in four Italian hospitals between March 1st, 2020, and May 31st, 2021 in patients who experienced a PE during hospitalization for a COVID-19 pneumonia, excluding patients who died during hospitalization. Baseline characteristics were collected and patients were grouped according to duration of anticoagulant treatment (< 3 months or > 3 months). The primary outcome was incidence of VTE recurrence while secondary outcome was the composite of deaths, major hemorrhages and VTE recurrence during follow-up. RESULTS 106 patients with PE were discharged, of these 95 (89.6 %) had follow up longer than 3 months (seven patients were lost to follow up and four died within three months). The median follow-up was 13 months (IQR 1-19). Overall, 23 % of subjects (22/95) were treated for 3 months or less and 76.8 % (73/95) received anticoagulation for >3 months. Of patients in the short treatment group, 4.5 % died, compared with 5.5 % of those in the longer treatment group (p = NS); no difference was shown in risk of VTE recurrence (0 % vs 4.1 %, p = NS), major bleeding (4.5 % vs 4.1 %, p = NS) or in composite outcome (9.1 % vs 11 %, p = NS). No difference was found between the two treatment groups for composite outcome using the Kaplan-Meier analysis (Log Rank Test p = 0.387). CONCLUSION In our retrospective multi-center cohort, prolongation of duration of anticoagulation seems not to affect risk of VTE recurrences, deaths and bleeding after a PE related to COVID-19.
Collapse
Affiliation(s)
- Lucia Filippi
- Medical Department Altovicentino Hospital, Santorso, Vicenza, Italy.
| | - Gianni Turcato
- Medical Department Altovicentino Hospital, Santorso, Vicenza, Italy
| | - Marta Milan
- Medical Department Santa Maria della Misericordia Hospital, Rovigo, Italy
| | - Sofia Barbar
- Medical Department Cittadella Hospital, Cittadella, Padova, Italy
| | - Eliana Miozzo
- Medical Department Santa Maria della Misericordia Hospital, Rovigo, Italy
| | - Arian Zaboli
- Emergency Department Tappeiner Hospital, Merano, Bolzano, Italy
| | - Diego Tonello
- Medical Department Altovicentino Hospital, Santorso, Vicenza, Italy
| | - Daniela Milazzo
- Medical Department Altovicentino Hospital, Santorso, Vicenza, Italy
| | | | - Stefano Cuppini
- Medical Department Santa Maria della Misericordia Hospital, Rovigo, Italy
| | | |
Collapse
|
266
|
Baruah P, Patra A, Barge S, Khan MR, Mukherjee AK. Therapeutic Potential of Bioactive Compounds from Edible Mushrooms to Attenuate SARS-CoV-2 Infection and Some Complications of Coronavirus Disease (COVID-19). J Fungi (Basel) 2023; 9:897. [PMID: 37755005 PMCID: PMC10532592 DOI: 10.3390/jof9090897] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/09/2023] [Accepted: 08/14/2023] [Indexed: 09/28/2023] Open
Abstract
The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a highly infectious positive RNA virus, has spread from its epicenter to other countries with increased mortality and morbidity. Its expansion has hampered humankind's social, economic, and health realms to a large extent. Globally, investigations are underway to understand the complex pathophysiology of coronavirus disease (COVID-19) induced by SARS-CoV-2. Though numerous therapeutic strategies have been introduced to combat COVID-19, none are fully proven or comprehensive, as several key issues and challenges remain unresolved. At present, natural products have gained significant momentum in treating metabolic disorders. Mushrooms have often proved to be the precursor of various therapeutic molecules or drug prototypes. The plentiful bioactive macromolecules in edible mushrooms, like polysaccharides, proteins, and other secondary metabolites (such as flavonoids, polyphenols, etc.), have been used to treat multiple diseases, including viral infections, by traditional healers and the medical fraternity. Some edible mushrooms with a high proportion of therapeutic molecules are known as medicinal mushrooms. In this review, an attempt has been made to highlight the exploration of bioactive molecules in mushrooms to combat the various pathophysiological complications of COVID-19. This review presents an in-depth and critical analysis of the current therapies against COVID-19 versus the potential of natural anti-infective, antiviral, anti-inflammatory, and antithrombotic products derived from a wide range of easily sourced mushrooms and their bioactive molecules.
Collapse
Affiliation(s)
- Paran Baruah
- Life Sciences Division, Institute of Advanced Study in Science and Technology, Paschim Boragaon, Garchuk, Guwahati 781035, Assam, India; (P.B.); (A.P.); (S.B.); (M.R.K.)
- Faculty of Science, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Aparup Patra
- Life Sciences Division, Institute of Advanced Study in Science and Technology, Paschim Boragaon, Garchuk, Guwahati 781035, Assam, India; (P.B.); (A.P.); (S.B.); (M.R.K.)
| | - Sagar Barge
- Life Sciences Division, Institute of Advanced Study in Science and Technology, Paschim Boragaon, Garchuk, Guwahati 781035, Assam, India; (P.B.); (A.P.); (S.B.); (M.R.K.)
| | - Mojibur R. Khan
- Life Sciences Division, Institute of Advanced Study in Science and Technology, Paschim Boragaon, Garchuk, Guwahati 781035, Assam, India; (P.B.); (A.P.); (S.B.); (M.R.K.)
| | - Ashis K. Mukherjee
- Life Sciences Division, Institute of Advanced Study in Science and Technology, Paschim Boragaon, Garchuk, Guwahati 781035, Assam, India; (P.B.); (A.P.); (S.B.); (M.R.K.)
| |
Collapse
|
267
|
Németh M, Mühl D, Csontos C, Nagy Á, Alizadeh H, Szakács Z. Acquired Hemophilia A after SARS-CoV-2 Infection: A Case Report and an Updated Systematic Review. Biomedicines 2023; 11:2400. [PMID: 37760842 PMCID: PMC10526109 DOI: 10.3390/biomedicines11092400] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/04/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
The role of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has been implicated in the pathogenesis of acquired hemophilia A (AHA). The aim of this study is to report our case and to summarize clinical studies on de novo AHA after SARS-CoV-2 infection. We performed a systematic search on the association of SARS-CoV-2 with AHA in four medical databases up to 28 May 2023. Eligible studies should include de novo AHA patients who had SARS-CoV-2 infection before or concomitant with the diagnosis of AHA. Findings were synthesized narratively. In addition, we report the case of a 62-year-old female patient, who presented to our clinic with left flank pain 2 weeks after SARS-CoV-2 infection. Clinical investigations confirmed AHA and imaging studies revealed retroperitoneal bleeding. Her hemostasis was successfully secured with bypassing agents; however, despite immunosuppressive therapy, high inhibitor titer persisted. In the systematic review, we identified only 12 relevant cases with a questionable cause-effect relationship between SARS-CoV-2 infection and AHA. Based on the qualitative analysis of the relevant publications, current clinical evidence is insufficient to support a cause-effect relationship. The analysis of data from ongoing AHA registries can serve further evidence.
Collapse
Affiliation(s)
- Márton Németh
- Department of Anesthesiology and Intensive Therapy, Medical School, University of Pécs, 7624 Pécs, Hungary; (M.N.); (D.M.); (C.C.)
| | - Diána Mühl
- Department of Anesthesiology and Intensive Therapy, Medical School, University of Pécs, 7624 Pécs, Hungary; (M.N.); (D.M.); (C.C.)
| | - Csaba Csontos
- Department of Anesthesiology and Intensive Therapy, Medical School, University of Pécs, 7624 Pécs, Hungary; (M.N.); (D.M.); (C.C.)
| | - Ágnes Nagy
- First Department of Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary; (Á.N.); (Z.S.)
| | - Hussain Alizadeh
- First Department of Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary; (Á.N.); (Z.S.)
| | - Zsolt Szakács
- First Department of Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary; (Á.N.); (Z.S.)
| |
Collapse
|
268
|
Shama, Mahmood A, Mehmood S, Zhang W. Pathological Effects of SARS-CoV-2 Associated with Hematological Abnormalities. Curr Issues Mol Biol 2023; 45:7161-7182. [PMID: 37754237 PMCID: PMC10528388 DOI: 10.3390/cimb45090453] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/09/2023] [Accepted: 08/22/2023] [Indexed: 09/28/2023] Open
Abstract
The SARS coronavirus 2 (SARS-CoV-2) is the causative agent of the 2019 coronavirus disease (COVID-19) pandemic that has claimed the lives of 6.9 million people and infected over 765 million. It has become a major worldwide health problem and is also known to cause abnormalities in various systems, including the hematologic system. COVID-19 infection primarily affects the lower respiratory tract and can lead to a cascade of events, including a cytokine storm, intravascular thrombosis, and subsequent complications such as arterial and venous thromboses. COVID-19 can cause thrombocytopenia, lymphopenia, and neutrophilia, which are associated with worse outcomes. Prophylactic anticoagulation is essential to prevent complications and death rates associated with the virus's effect on the coagulation system. It is crucial to recognize these complications early and promptly start therapeutic anticoagulation to improve patient outcomes. While rare, COVID-19-induced disseminated intravascular coagulation (DIC) exhibits some similarities to DIC induced by sepsis. Lactate dehydrogenase (LDH), D-dimer, ferritin, and C-reactive protein (CRP) biomarkers often increase in serious COVID-19 cases and poor prognosis. Understanding the pathophysiology of the disease and identifying risk factors for adverse outcomes is critical for effective management of COVID-19.
Collapse
Affiliation(s)
- Shama
- Department of Microbiology, School of Medicine, Jiangsu University, Zhenjiang 212013, China (A.M.)
| | - Asif Mahmood
- Department of Microbiology, School of Medicine, Jiangsu University, Zhenjiang 212013, China (A.M.)
- School of Material Science and Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Shahid Mehmood
- Institute of Life Sciences, Jiangsu University, Zhenjiang 212013, China;
| | - Wen Zhang
- Department of Microbiology, School of Medicine, Jiangsu University, Zhenjiang 212013, China (A.M.)
| |
Collapse
|
269
|
Koc I, Deniz O, Ozmen SU, Bulut S. Predicting acute pulmonary embolism in COVID-19. Medicine (Baltimore) 2023; 102:e34916. [PMID: 37653759 PMCID: PMC10470735 DOI: 10.1097/md.0000000000034916] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/10/2023] [Accepted: 08/03/2023] [Indexed: 09/02/2023] Open
Abstract
Acute pulmonary embolism (PE) is a life-threatening condition in patients with Coronavirus disease-2019 (COVID-19). Computed tomography pulmonary angiography is the preferred test to confirm the diagnosis. However, computed tomography pulmonary angiography is expensive and is not available in every clinic. This study aimed to determine whether clinical findings, symptoms, and parameters that are cost-effective and available in many clinics such as C-reactive protein (CRP) lymphocyte ratio (CLR), and ferritin CRP ratio (FCR) can be used in the diagnosis of PE in patients with COVID-19. Out of the reviewed files, 127 patients were diagnosed with PE, whereas 105 patients had no PE. At the first admission, laboratory parameters, complaints, respiratory rate, and percent oxygen saturation in the blood (SpO2) with a pulse oximeter were recorded for each patient. Eosinophil levels remained lower, whereas ferritin lymphocyte ratio and CLR were higher in the PE group. Patients with more elevated ferritin, CRP, and CLR had an increased mortality risk. Shortness of breath and tiredness was more common in the PE group. A decrease in eosinophil levels, whereas an increase in CLR, D-dimer, and CRP may predict PE. Elevated CLR is highly predictive of PE and is associated with increased mortality risk. COVID-19 patients with a CLR level above 81 should be investigated for PE.
Collapse
Affiliation(s)
- Ibrahim Koc
- Bursa City Hospital Pulmonary Medicine, Bursa, Turkey
| | - Olgun Deniz
- Bursa City Hospital, Palliative Care Unit, Geriatric Medicine Clinic, Bursa, Turkey
| | | | - Sertan Bulut
- Ankara Atatürk Sanatoryum Educational Research Hospital, Bursa, Turkey
| |
Collapse
|
270
|
Klein A, Bastard M, Hemat H, Singh S, Muniz B, Manangama G, Alayyan A, Tamanna AH, Barakzaie B, Popal N, Kakar MAZ, Poulet E, Finger F. Factors associated with adverse outcomes among patients hospitalized at a COVID-19 treatment center in Herat, Afghanistan. PLOS GLOBAL PUBLIC HEALTH 2023; 3:e0001687. [PMID: 37619213 PMCID: PMC10449473 DOI: 10.1371/journal.pgph.0001687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 06/30/2023] [Indexed: 08/26/2023]
Abstract
Though many studies on COVID have been published to date, data on COVID-19 epidemiology, symptoms, risk factors and severity in low- and middle-income countries (LMICS), such as Afghanistan are sparse. To describe clinical characteristics, severity, and outcomes of patients hospitalized in the MSF COVID-19 treatment center (CTC) in Herat, Afghanistan and to assess risk factors associated with severe outcomes. 1113 patients were included in this observational study between June 2020 and April 2022. Descriptive analysis was performed on clinical characteristics, complications, and outcomes of patients. Univariate description by Cox regression to identify risk factors for an adverse outcome was performed. Adverse outcome was defined as death or transfer to a level 3 intensive care located at another health facility. Finally, factors identified were included in a multivariate Cox survival analysis. A total of 165 patients (14.8%) suffered from a severe disease course, with a median time of 6 days (interquartile range: 2-11 days) from admission to adverse outcome. In our multivariate model, we identified male gender, age over 50, high O2 flow administered during admission, lymphopenia, anemia and O2 saturation < = 93% during the first three days of admission as predictors for a severe disease course (p<0.05). Our analysis concluded in a relatively low rate of adverse outcomes of 14.8%. This is possibly related to the fact that the resources at an MSF-led facility are higher, in terms of human resources as well as supply of drugs and biomedical equipment, including oxygen therapy devices, compared to local hospitals. Predictors for severe disease outcomes were found to be comparable to other settings.
Collapse
|
271
|
Tang Z, Xu Y, Tan Y, Shi H, Jin P, Li Y, Teng J, Liu H, Pan H, Hu Q, Cheng X, Ye J, Su Y, Sun Y, Meng J, Zhou Z, Chi H, Wang X, Liu J, Lu Y, Liu F, Dai J, Yang C, Chen S, Liu T. CD36 mediates SARS-CoV-2-envelope-protein-induced platelet activation and thrombosis. Nat Commun 2023; 14:5077. [PMID: 37604832 PMCID: PMC10442425 DOI: 10.1038/s41467-023-40824-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 08/10/2023] [Indexed: 08/23/2023] Open
Abstract
Aberrant coagulation and thrombosis are associated with severe COVID-19 post-SARS-CoV-2 infection, yet the underlying mechanism remains obscure. Here we show that serum levels of SARS-CoV-2 envelope (E) protein are associated with coagulation disorders of COVID-19 patients, and intravenous administration of the E protein is able to potentiate thrombosis in mice. Through protein pull-down and mass spectrometry, we find that CD36, a transmembrane glycoprotein, directly binds with E protein and mediates hyperactivation of human and mouse platelets through the p38 MAPK-NF-κB signaling pathway. Conversely, the pharmacological blockade of CD36 or p38 notably attenuates human platelet activation induced by the E protein. Similarly, the genetic deficiency of CD36, as well as the pharmacological inhibition of p38 in mice, significantly diminishes E protein-induced platelet activation and thrombotic events. Together, our study reveals a critical role for the CD36-p38 axis in E protein-induced platelet hyperactivity, which could serve as an actionable target for developing therapies against aberrant thrombotic events related to the severity and mortality of COVID-19.
Collapse
Affiliation(s)
- Zihan Tang
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Second Road, Shanghai, 200025, China
| | - Yanyan Xu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Yun Tan
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hui Shi
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Second Road, Shanghai, 200025, China
| | - Peipei Jin
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yunqi Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jialin Teng
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Second Road, Shanghai, 200025, China
| | - Honglei Liu
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Second Road, Shanghai, 200025, China
| | - Haoyu Pan
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Second Road, Shanghai, 200025, China
| | - Qiongyi Hu
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Second Road, Shanghai, 200025, China
| | - Xiaobing Cheng
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Second Road, Shanghai, 200025, China
| | - Junna Ye
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Second Road, Shanghai, 200025, China
| | - Yutong Su
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Second Road, Shanghai, 200025, China
| | - Yue Sun
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Second Road, Shanghai, 200025, China
| | - Jianfen Meng
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Second Road, Shanghai, 200025, China
| | - Zhuochao Zhou
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Second Road, Shanghai, 200025, China
| | - Huihui Chi
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Second Road, Shanghai, 200025, China
| | - Xuefeng Wang
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Yong Lu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Feng Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Jing Dai
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Chengde Yang
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Second Road, Shanghai, 200025, China.
| | - Saijuan Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Tingting Liu
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Second Road, Shanghai, 200025, China.
| |
Collapse
|
272
|
Alijotas-Reig J, Anunciación-Llunell A, Morales-Pérez S, Trapé J, Esteve-Valverde E, Miro-Mur F. Thrombosis and Hyperinflammation in COVID-19 Acute Phase Are Related to Anti-Phosphatidylserine and Anti-Phosphatidylinositol Antibody Positivity. Biomedicines 2023; 11:2301. [PMID: 37626797 PMCID: PMC10452204 DOI: 10.3390/biomedicines11082301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/11/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
Antiphospholipid antibodies (APLA) are strongly associated with thrombosis seen in patients with antiphospholipid syndrome. In COVID-19, thrombosis has been observed as one of the main comorbidities. In patients hospitalised for COVID-19, we want to check whether APLA positivity is associated with COVID-19-related thrombosis, inflammation, severity of disease, or long COVID-19. We enrolled 92 hospitalised patients with COVID-19 between March and April 2020 who were tested for 18 different APLAs (IgG and IgM) with a single line-immunoassay test. A total of 30 healthy blood donors were used to set the cut-off for each APLA positivity. Of the 92 COVID-19 inpatients, 30 (32.61%; 95% CI [23.41-43.29]) tested positive for APLA, of whom 10 (33.3%; 95% CI [17.94-52.86]) had more than one APLA positivity. Anti-phosphatidylserine IgM positivity was described in 5.4% of inpatients (n = 5) and was associated with the occurrence of COVID-19-related thrombosis (p = 0.046). Anti-cardiolipin IgM positivity was the most prevalent among the inpatients (n = 12, 13.0%) and was associated with a recorded thrombosis in their clinical history (p = 0.044); however, its positivity was not associated with the occurrence of thrombosis during their hospitalisation for COVID-19. Anti-phosphatidylinositol IgM positivity, with a prevalence of 5.4% (n = 5), was associated with higher levels of interleukin (IL)-6 (p = 0.007) and ferritin (p = 0.034). Neither of these APLA positivities was a risk factor for COVID-19 severity or a predictive marker for long COVID-19. In conclusion, almost a third of COVID-19 inpatients tested positive for at least one APLA. Anti-phosphatidylserine positivity in IgM class was associated with thrombosis, and anti-phosphatidylinositol positivity in IgM class was associated with inflammation, as noticed by elevated levels of IL-6. Thus, testing for non-criteria APLA to assess the risk of clinical complications in hospitalised COVID-19 patients might be beneficial. However, they were not related to disease severity or long COVID-19.
Collapse
Affiliation(s)
- Jaume Alijotas-Reig
- Systemic Autoimmune Diseases Research Unit, Vall d’Hebron Institut de Recerca (VHIR), 08035 Barcelona, Catalonia, Spain;
- Systemic Autoimmune Diseases Unit, Department of Internal Medicine, Hospital Universitari Vall d’Hebron (HUVH), 08035 Barcelona, Catalonia, Spain
- Department of Medicine, Faculty of Medicine, Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Catalonia, Spain
| | - Ariadna Anunciación-Llunell
- Systemic Autoimmune Diseases Research Unit, Vall d’Hebron Institut de Recerca (VHIR), 08035 Barcelona, Catalonia, Spain;
| | - Stephanie Morales-Pérez
- Systemic Autoimmune Disease Unit, Internal Medicine Department, Althaia Healthcare University Network of Manresa, 08243 Manresa, Catalonia, Spain (J.T.)
| | - Jaume Trapé
- Systemic Autoimmune Disease Unit, Internal Medicine Department, Althaia Healthcare University Network of Manresa, 08243 Manresa, Catalonia, Spain (J.T.)
| | - Enrique Esteve-Valverde
- Systemic Autoimmune Diseases Unit, Department of Internal Medicine, Hospital Universitari Parc Taulí, 08208 Sabadell, Catalonia, Spain
| | - Francesc Miro-Mur
- Systemic Autoimmune Diseases Research Unit, Vall d’Hebron Institut de Recerca (VHIR), 08035 Barcelona, Catalonia, Spain;
| |
Collapse
|
273
|
Zheng LL, Wang S, Li ZG, Han L, Zhu CD, Li CY, Zhang XX, Deng X. Correlation of Coagulation Dysfunction with Infection and Hypercapnia in Acute Exacerbation of COPD Patients. Infect Drug Resist 2023; 16:5387-5394. [PMID: 37614682 PMCID: PMC10443683 DOI: 10.2147/idr.s421925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 08/02/2023] [Indexed: 08/25/2023] Open
Abstract
Background This study aims to explore the factors influencing the coagulation function of patients with chronic obstructive pulmonary disease (COPD) and its effects on thrombosis. Methods A total of 155 COPD patients, including 118 patients with acute exacerbation of COPD (AECOPD) and 37 patients with stable COPD (SCOPD), were enrolled in this study. Meanwhile, 50 patients with gastrointestinal polyps found during physical examination and treated with surgery in the same period were enrolled as the control group. The basic data, routine blood tests, C-reactive protein (CRP), procalcitonin (PCT), and coagulation indexes of the three groups were collected, as well as arterial blood gas indexes of AECOPD patients. Results The differences in erythrocyte count and hemoglobin among groups were not statistically significant. Compared with the SCOPD group and control group, white blood cell (WBC), neutrophil percentage, PCT, CRP, prothrombin time (PT), and fibrinogen (FIB) in the AECOPD group increased significantly, while the international normalized ratio (INR) decreased (P < 0.05). The differences in activated partial thromboplastin time (APTT) and D-dimer among groups were not statistically significant (P > 0.05). Thrombin time (TT) in the AECOPD group was shorter than that of the control group, and PT was longer than that of the SCOPD group (P < 0.05). Five patients with AECOPD and one patient with SCOPD had venous thrombosis. Conclusion The abnormal coagulation function in AECOPD patients is related to the degree of infection and hypercapnia, which may be a risk factor for thrombosis.
Collapse
Affiliation(s)
- Li-Li Zheng
- Department of Respiratory Medicine of Geriatrics Center, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, 230000, People’s Republic of China
| | - Sheng Wang
- Department of Respiratory Medicine of Geriatrics Center, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, 230000, People’s Republic of China
| | - Ze-Geng Li
- Anhui University of Traditional Chinese Medicine, Hefei, 230000, People’s Republic of China
| | - Lei Han
- Anhui University of Traditional Chinese Medicine, Hefei, 230000, People’s Republic of China
| | - Chun-Dong Zhu
- Department of Respiratory Medicine of Geriatrics Center, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, 230000, People’s Republic of China
| | - Chun-Ying Li
- Department of Respiratory Medicine of Geriatrics Center, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, 230000, People’s Republic of China
| | - Xing-Xing Zhang
- Department of Respiratory Medicine of Geriatrics Center, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, 230000, People’s Republic of China
| | - Xue Deng
- Department of Respiratory Medicine of Geriatrics Center, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, 230000, People’s Republic of China
| |
Collapse
|
274
|
Santos BC, Flumignan RL, Civile VT, Atallah ÁN, Nakano LC. Prophylactic anticoagulants for non-hospitalised people with COVID-19. Cochrane Database Syst Rev 2023; 8:CD015102. [PMID: 37591523 PMCID: PMC10428666 DOI: 10.1002/14651858.cd015102.pub2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) pandemic has impacted healthcare systems worldwide. Multiple reports on thromboembolic complications related to COVID-19 have been published, and researchers have described that people with COVID-19 are at high risk for developing venous thromboembolism (VTE). Anticoagulants have been used as pharmacological interventions to prevent arterial and venous thrombosis, and their use in the outpatient setting could potentially reduce the prevalence of vascular thrombosis and associated mortality in people with COVID-19. However, even lower doses used for a prophylactic purpose may result in adverse events such as bleeding. It is important to consider the evidence for anticoagulant use in non-hospitalised people with COVID-19. OBJECTIVES To evaluate the benefits and harms of prophylactic anticoagulants versus active comparators, placebo or no intervention, or non-pharmacological interventions in non-hospitalised people with COVID-19. SEARCH METHODS We used standard, extensive Cochrane search methods. The latest search date was 18 April 2022. SELECTION CRITERIA We included randomised controlled trials (RCTs) comparing prophylactic anticoagulants with placebo or no treatment, another active comparator, or non-pharmacological interventions in non-hospitalised people with COVID-19. We included studies that compared anticoagulants with a different dose of the same anticoagulant. We excluded studies with a duration of under two weeks. DATA COLLECTION AND ANALYSIS We used standard Cochrane methodological procedures. Our primary outcomes were all-cause mortality, VTE (deep vein thrombosis (DVT) or pulmonary embolism (PE)), and major bleeding. Our secondary outcomes were DVT, PE, need for hospitalisation, minor bleeding, adverse events, and quality of life. We used GRADE to assess the certainty of the evidence. MAIN RESULTS We included five RCTs with up to 90 days of follow-up (short term). Data were available for meta-analysis from 1777 participants. Anticoagulant compared to placebo or no treatment Five studies compared anticoagulants with placebo or no treatment and provided data for three of our outcomes of interest (all-cause mortality, major bleeding, and adverse events). The evidence suggests that prophylactic anticoagulants may lead to little or no difference in all-cause mortality (risk ratio (RR) 0.36, 95% confidence interval (CI) 0.04 to 3.61; 5 studies; 1777 participants; low-certainty evidence) and probably reduce VTE from 3% in the placebo group to 1% in the anticoagulant group (RR 0.36, 95% CI 0.16 to 0.85; 4 studies; 1259 participants; number needed to treat for an additional beneficial outcome (NNTB) = 50; moderate-certainty evidence). There may be little to no difference in major bleeding (RR 0.36, 95% CI 0.01 to 8.78; 5 studies; 1777 participants; low-certainty evidence). Anticoagulants probably result in little or no difference in DVT (RR 1.02, 95% CI 0.30 to 3.46; 3 studies; 1009 participants; moderate-certainty evidence), but probably reduce the risk of PE from 2.7% in the placebo group to 0.7% in the anticoagulant group (RR 0.25, 95% CI 0.08 to 0.79; 3 studies; 1009 participants; NNTB 50; moderate-certainty evidence). Anticoagulants probably lead to little or no difference in reducing hospitalisation (RR 1.01, 95% CI 0.59 to 1.75; 4 studies; 1459 participants; moderate-certainty evidence) and may lead to little or no difference in adverse events (minor bleeding, RR 2.46, 95% CI 0.90 to 6.72; 5 studies, 1777 participants; low-certainty evidence). Anticoagulant compared to a different dose of the same anticoagulant One study compared anticoagulant (higher-dose apixaban) with a different (standard) dose of the same anticoagulant and reported five relevant outcomes. No cases of all-cause mortality, VTE, or major bleeding occurred in either group during the 45-day follow-up (moderate-certainty evidence). Higher-dose apixaban compared to standard-dose apixaban may lead to little or no difference in reducing the need for hospitalisation (RR 1.89, 95% CI 0.17 to 20.58; 1 study; 278 participants; low-certainty evidence) or in the number of adverse events (minor bleeding, RR 0.47, 95% CI 0.09 to 2.54; 1 study; 278 participants; low-certainty evidence). Anticoagulant compared to antiplatelet agent One study compared anticoagulant (apixaban) with antiplatelet agent (aspirin) and reported five relevant outcomes. No cases of all-cause mortality or major bleeding occurred during the 45-day follow-up (moderate-certainty evidence). Apixaban may lead to little or no difference in VTE (RR 0.36, 95% CI 0.01 to 8.65; 1 study; 279 participants; low-certainty evidence), need for hospitalisation (RR 3.20, 95% CI 0.13 to 77.85; 1 study; 279 participants; low-certainty evidence), or adverse events (minor bleeding, RR 2.13, 95% CI 0.40 to 11.46; 1 study; 279 participants; low-certainty evidence). No included studies reported on quality of life or investigated anticoagulants compared to a different anticoagulant, or anticoagulants compared to non-pharmacological interventions. AUTHORS' CONCLUSIONS We found low- to moderate-certainty evidence from five RCTs that prophylactic anticoagulants result in little or no difference in major bleeding, DVT, need for hospitalisation, or adverse events when compared with placebo or no treatment in non-hospitalised people with COVID-19. Low-certainty evidence indicates that prophylactic anticoagulants may result in little or no difference in all-cause mortality when compared with placebo or no treatment, but moderate-certainty evidence indicates that prophylactic anticoagulants probably reduce the incidence of VTE and PE. Low-certainty evidence suggests that comparing different doses of the same prophylactic anticoagulant may result in little or no difference in need for hospitalisation or adverse events. Prophylactic anticoagulants may result in little or no difference in risk of VTE, hospitalisation, or adverse events when compared with antiplatelet agents (low-certainty evidence). Given that there were only short-term data from one study, these results should be interpreted with caution. Additional trials of sufficient duration are needed to clearly determine any effect on clinical outcomes.
Collapse
Affiliation(s)
- Brena C Santos
- Department of Surgery, Division of Vascular and Endovascular Surgery, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ronald Lg Flumignan
- Department of Surgery, Division of Vascular and Endovascular Surgery, Universidade Federal de São Paulo, São Paulo, Brazil
- Cochrane Brazil, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Vinicius T Civile
- Cochrane Brazil, Universidade Federal de São Paulo, São Paulo, Brazil
- Department of Physiotherapy, Universidade Paulista, São Paulo, Brazil
| | - Álvaro N Atallah
- Cochrane Brazil, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Luis Cu Nakano
- Department of Surgery, Division of Vascular and Endovascular Surgery, Universidade Federal de São Paulo, São Paulo, Brazil
- Cochrane Brazil, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
275
|
Szilveszter M, Pál S, Simon-Szabó Z, Akácsos-Szász OZ, Moldován M, Réger B, Dénes L, Faust Z, Tilinca MC, Nemes-Nagy E. The Management of COVID-19-Related Coagulopathy: A Focus on the Challenges of Metabolic and Vascular Diseases. Int J Mol Sci 2023; 24:12782. [PMID: 37628963 PMCID: PMC10454092 DOI: 10.3390/ijms241612782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/07/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
The course of COVID-19 is highly dependent on the associated cardiometabolic comorbidities of the patient, which worsen the prognosis of coronavirus infection, mainly due to systemic inflammation, endothelium dysfunction, and thrombosis. A search on the recent medical literature was performed in five languages, using the PubMed, Embase, Cochrane, and Google Scholar databases, for the review of data regarding the management of patients with a high risk for severe COVID-19, focusing on the associated coagulopathy. Special features of COVID-19 management are presented, based on the underlying conditions (obesity, diabetes mellitus, and cardiovascular diseases), emphasizing the necessity of a modern, holistic approach to thromboembolic states. The latest findings regarding the most efficient therapeutic approaches are included in the article, offering guidance for medical professionals in severe, complicated cases of SARS-CoV-2 infection. We can conclude that severe COVID-19 is closely related to vascular inflammation and intense cytokine release leading to hemostasis disorders. Overweight, hyperglycemia, cardiovascular diseases, and old age are important risk factors for severe outcomes of coronavirus infection, involving a hypercoagulable state. Early diagnosis and proper therapy in complicated SARS-CoV-2-infected cases could reduce mortality and the need for intensive care during hospitalization in patients with cardiometabolic comorbidities.
Collapse
Affiliation(s)
- Mónika Szilveszter
- Clinic of Plastic Surgery, Mureș County Emergency Hospital, 540136 Târgu-Mureș, Romania;
| | - Sándor Pál
- Department of Transfusion Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary;
| | - Zsuzsánna Simon-Szabó
- Department of Pathophysiology, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu-Mureș, 540142 Târgu-Mureș, Romania
| | - Orsolya-Zsuzsa Akácsos-Szász
- Doctoral School, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu-Mureș, 540142 Târgu-Mureș, Romania;
| | - Mihály Moldován
- Klinik für Suchttherapie, ZtP Winnenden-Haus der Gesundheit, 73525 Schwäbisch Gümund, Germany;
| | - Barbara Réger
- Department of Laboratory Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary;
| | - Lóránd Dénes
- Department of Anatomy and Embryology, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu-Mureș, 540142 Târgu-Mureș, Romania;
| | - Zsuzsanna Faust
- Department of Transfusion Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary;
| | - Mariana Cornelia Tilinca
- Department of Internal Medicine I, Faculty of Medicine in English, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu-Mureș, 540142 Târgu-Mureș, Romania;
| | - Enikő Nemes-Nagy
- Department of Chemistry and Medical Biochemistry, Faculty of Medicine in English, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu-Mureș, 540142 Târgu-Mureș, Romania;
| |
Collapse
|
276
|
Babkina AS, Yadgarov MY, Volkov AV, Kuzovlev AN, Grechko AV, Golubev AM. Spectrum of Thrombotic Complications in Fatal Cases of COVID-19: Focus on Pulmonary Artery Thrombosis In Situ. Viruses 2023; 15:1681. [PMID: 37632023 PMCID: PMC10458612 DOI: 10.3390/v15081681] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/24/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023] Open
Abstract
COVID-19-related thrombosis affects the venous and arterial systems. Data from 156 autopsies of COVID-19 patients were retrospectively analyzed to investigate the pattern of thrombotic complications and factors associated with pulmonary artery thrombosis and thromboembolism. Thrombotic complications were observed in a significant proportion (n = 68, 44%), with pulmonary artery thrombosis the most frequently identified thrombotic event (42, 27%). Multivariate analysis revealed that the length of hospital stay (OR 1.1, p = 0.004), neutrophil infiltration in the alveolar spaces (OR 3.6, p = 0.002), and the absence of hyaline membranes (OR 0.1, p = 0.01) were associated with thrombotic complications. Neutrophil infiltration in the alveolar spaces (OR 8, p < 0.001) and the absence of hyaline membranes (OR 0.1, p = 0.003) were also independent predictors of pulmonary artery thrombosis. The association of pulmonary artery thrombosis with an absence of hyaline membranes suggests it occurs later in the course of COVID-19 infection. As neutrophil infiltration in the alveolar spaces may indicate bacterial infection, our studies suggest the consideration of bacterial infections in these critically ill patients.
Collapse
Affiliation(s)
- Anastasiya S. Babkina
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia; (M.Y.Y.); (A.N.K.); (A.V.G.); (A.M.G.)
| | - Mikhail Y. Yadgarov
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia; (M.Y.Y.); (A.N.K.); (A.V.G.); (A.M.G.)
| | - Alexey V. Volkov
- Department of Pathological Anatomy, Institute of Medicine, Peoples’ Friendship University of Russia Named after Patrice Lumumba, Moscow 117198, Russia;
| | - Artem N. Kuzovlev
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia; (M.Y.Y.); (A.N.K.); (A.V.G.); (A.M.G.)
| | - Andrey V. Grechko
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia; (M.Y.Y.); (A.N.K.); (A.V.G.); (A.M.G.)
| | - Arkady M. Golubev
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia; (M.Y.Y.); (A.N.K.); (A.V.G.); (A.M.G.)
| |
Collapse
|
277
|
Fang C, Yan W, Chen Y, Dou Z, Liu T, Luo F, Chen W, Li X, Chen Y, Wu W, Yuan Z, Niu Y, Wang P, Zhu W, Luo X, Chen T, Bai X, Wang X, Ning Q. Long-term SARS-CoV-2 neutralizing antibody level prediction using multimodal deep learning: A prospective cohort study on longitudinal data in Wuhan, China. J Med Virol 2023; 95:e29036. [PMID: 37621210 DOI: 10.1002/jmv.29036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 07/07/2023] [Accepted: 08/02/2023] [Indexed: 08/26/2023]
Abstract
The ongoing epidemic of SARS-CoV-2 is taking a substantial financial and health toll on people worldwide. Assessing the level and duration of SARS-CoV-2 neutralizing antibody (Nab) would provide key information for government to make sound healthcare policies. Assessed at 3-, 6-, 12-, and 18-month postdischarge, we described the temporal change of IgG levels in 450 individuals with moderate to critical COVID-19 infection. Moreover, a data imputation framework combined with a novel deep learning model was implemented to predict the long-term Nab and IgG levels in these patients. Demographic characteristics, inspection reports, and CT scans during hospitalization were used in this model. Interpretability of the model was further validated with Shapely Additive exPlanation (SHAP) and Gradient-weighted Class Activation Mapping (GradCAM). IgG levels peaked at 3 months and remained stable in 12 months postdischarge, followed by a significant decline in 18 months postdischarge. However, the Nab levels declined from 6 months postdischarge. By training on the cohort of 450 patients, our long-term antibody prediction (LTAP) model could predict long-term IgG levels with relatively high area under the receiver operating characteristic curve (AUC), accuracy, precision, recall, and F1-score, which far exceeds the performance achievable by commonly used models. Several prognostic factors including FDP levels, the percentages of T cells, B cells and natural killer cells, older age, sex, underlying diseases, and so forth, served as important indicators for IgG prediction. Based on these top 15 prognostic factors identified in IgG prediction, a simplified LTAP model for Nab level prediction was established and achieved an AUC of 0.828, which was 8.9% higher than MLP and 6.6% higher than LSTM. The close correlation between IgG and Nab levels making it possible to predict long-term Nab levels based on the factors selected by our LTAP model. Furthermore, our model identified that coagulation disorders and excessive immune response, which indicate disease severity, are closely related to the production of IgG and Nab. This universal model can be used as routine discharge tests to identify virus-infected individuals at risk for recurrent infection and determine the optimal timing of vaccination for general populations.
Collapse
Affiliation(s)
- Cong Fang
- School of Electronic Information and Communications, Huazhong University of Science and Technology, Wuhan, China
| | - Weiming Yan
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuying Chen
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiyong Dou
- School of Electronic Information and Communications, Huazhong University of Science and Technology, Wuhan, China
| | - Tingting Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengning Luo
- Department of Computer Science, University of Toronto, Toronto, Canada
| | - Weiwei Chen
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xitang Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yajie Chen
- School of Electronic Information and Communications, Huazhong University of Science and Technology, Wuhan, China
| | - Wenhui Wu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhize Yuan
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuxin Niu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenzhen Zhu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Luo
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Chen
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Bai
- School of Artificial Intelligence and Automation, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaojing Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Ning
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
278
|
Li S, Wang H, Shao Q. The central role of neutrophil extracellular traps (NETs) and by-products in COVID-19 related pulmonary thrombosis. Immun Inflamm Dis 2023; 11:e949. [PMID: 37647446 PMCID: PMC10461423 DOI: 10.1002/iid3.949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/26/2023] [Accepted: 07/08/2023] [Indexed: 09/01/2023] Open
Abstract
Extracellular trap networks (neutrophil extracellular traps [NETs]) of polymorphonuclear neutrophils are mesh-like substances that prevent the spread of pathogens. They primarily consist of DNA skeletons, histones, granule components, and cytoplasmic proteins. NETs formation requires a certain environment and there are different pathways for NETs production. However, it is still not clear how severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) promotes NETs. NETs exert antiinflammatory effects through immune response, while they can also lead to certain adverse outcomes, such as the development of immunothrombosis. Coronavirus disease 2019 (COVID-19) is an inflammatory reaction affecting various organs caused by SARS-CoV-2, especially the lungs. NETs production and disease severity are linked with unique neutrophil clusters by single-cell RNA sequencing. NETs might exert an anti-inflammatory role in the initial stage of lung tissue inflammation. Nevertheless, numerous studies and cases have shown that they can also result in pulmonary thrombosis. There is mounting evidence that NETs are tightly related with COVID-19 pulmonary thrombosis, and many studies on the mechanisms are involved. The role and mechanism of NETs in the development of pulmonary thrombosis will be the main topics of this manuscript. Additionally, we address the potential targeting of NETs in COVID-19 patients.
Collapse
Affiliation(s)
- Shi Li
- Department of ImmunologySchool of Medicine, Jiangsu UniversityZhenjiangJiangsuChina
| | - Hui Wang
- Department of ImmunologySchool of Medicine, Jiangsu UniversityZhenjiangJiangsuChina
| | - Qixiang Shao
- Department of ImmunologySchool of Medicine, Jiangsu UniversityZhenjiangJiangsuChina
- Department of Medical Microbiology and Immunology, Institute of Medical Genetics and Reproductive Immunity, School of Medical Science and Laboratory MedicineJiangsu College of NursingHuai'anJiangsuChina
| |
Collapse
|
279
|
Kovačević M, Rizvanović N, Šabanović Adilović A, Čaušević S. COVID-19 associated coagulopathy in septic critically ill patients - a retrospective cohort study. MEDICINSKI GLASNIK : OFFICIAL PUBLICATION OF THE MEDICAL ASSOCIATION OF ZENICA-DOBOJ CANTON, BOSNIA AND HERZEGOVINA 2023; 20:142-147. [PMID: 37585306 DOI: 10.17392/1592-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/15/2023] [Accepted: 03/31/2023] [Indexed: 08/18/2023]
Abstract
Aim To determine the relationship between coagulation disorders and septic condition in COVID-19 critically ill patients. Methods Data from 99 patients who presented with COVID-19 acute hypoxemic respiratory failure (CAHRF) were divided into two groups: Group 1- patients who developed sepsis, and Group 2 - patients who developed septic shock. Age, sex, comorbidities, quick Sequential Organ Failure Assessment (qSOFA) score, vasopressor and inotrope requirement, laboratory findings (platelets, neutrophils, lymphocytes, procalcitonin - PCT, C-reactive protein, fibrinogen, D-dimer, sepsis-induced coagulopathy - SIC, and disseminated intravascular coagulation - DIC score) were recorded on the day of admission and on the day of starting invasive mechanical ventilation. The primary outcome was to establish COVID-19 associated coagulopathy with sepsis and septic shock; the secondary outcome measure was incidence of coagulopathy in septic COVID-19 critically ill patients. Results The most common coagulation abnormality was international normalized ratio (INR) (p=0.019) for Group 2, followed by the values of inflammatory parameters PCT (p=0.002) and lymphocytes (p=0.011) also for Group 2. The statistical significance of SIC score was observed for both groups (p=0.007) and p=0.012, respectively. Norepinephrine (p=0.000) and dobutamine (p=0.000) for Group 2, qSOFA for both groups (p = 0.000) were statistically significant. Conclusion The observed coagulation abnormalities met the criteria for a SIC diagnosis, therefore, the management of coagulation disorders at this stage of the disease should follow the management of a septic condition.
Collapse
Affiliation(s)
- Mirza Kovačević
- Department of Anaesthesiology, Resuscitation and Intensive Care, Cantonal Hospital Zenica, Zenica, Bosnia and Herzegovina
- School of Medicine, University of Zenica, Zenica, Bosnia and Herzegovina
| | - Nermina Rizvanović
- Department of Anaesthesiology, Resuscitation and Intensive Care, Cantonal Hospital Zenica, Zenica, Bosnia and Herzegovina
- School of Medicine, University of Zenica, Zenica, Bosnia and Herzegovina
| | - Adisa Šabanović Adilović
- Department of Anaesthesiology, Resuscitation and Intensive Care, Cantonal Hospital Zenica, Zenica, Bosnia and Herzegovina
- School of Medicine, University of Zenica, Zenica, Bosnia and Herzegovina
| | - Senada Čaušević
- Department of Anaesthesiology, Resuscitation and Intensive Care, Cantonal Hospital Zenica, Zenica, Bosnia and Herzegovina
- School of Medicine, University of Zenica, Zenica, Bosnia and Herzegovina
| |
Collapse
|
280
|
Zhang Z, Dai W, Zhu W, Rodriguez M, Lund H, Xia Y, Chen Y, Rau M, Schneider EA, Graham MB, Jobe S, Wang D, Cui W, Wen R, Whiteheart SW, Wood JP, Silverstein R, Berger JS, Kreuziger LB, Barrett TJ, Zheng Z. Plasma tissue-type plasminogen activator is associated with lipoprotein(a) and clinical outcomes in hospitalized patients with COVID-19. Res Pract Thromb Haemost 2023; 7:102164. [PMID: 37680312 PMCID: PMC10480648 DOI: 10.1016/j.rpth.2023.102164] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 09/09/2023] Open
Abstract
Background Patients with COVID-19 have a higher risk of thrombosis and thromboembolism, but the underlying mechanism(s) remain to be fully elucidated. In patients with COVID-19, high lipoprotein(a) (Lp(a)) is positively associated with the risk of ischemic heart disease. Lp(a), composed of an apoB-containing particle and apolipoprotein(a) (apo(a)), inhibits the key fibrinolytic enzyme, tissue-type plasminogen activator (tPA). However, whether the higher Lp(a) associates with lower tPA activity, the longitudinal changes of these parameters in hospitalized patients with COVID-19, and their correlation with clinical outcomes are unknown. Objectives To assess if Lp(a) associates with lower tPA activity in COVID-19 patients, and how in COVID-19 populations Lp(a) and tPA change post infection. Methods Endogenous tPA enzymatic activity, tPA or Lp(a) concentration were measured in plasma from hospitalized patients with and without COVID-19. The association between plasma tPA and adverse clinical outcomes was assessed. Results In hospitalized patients with COVID-19, we found lower tPA enzymatic activity and higher plasma Lp(a) than that in non-COVID-19 controls. During hospitalization, Lp(a) increased and tPA activity decreased, which associates with mortality. Among those who survived, Lp(a) decreased and tPA enzymatic activity increased during recovery. In patients with COVID-19, tPA activity is inversely correlated with tPA concentrations, thus, in another larger COVID-19 cohort, we utilized plasma tPA concentration as a surrogate to inversely reflect tPA activity. The tPA concentration was positively associated with death, disease severity, plasma inflammatory, and prothrombotic markers, and with length of hospitalization among those who were discharged. Conclusion High Lp(a) concentration provides a possible explanation for low endogenous tPA enzymatic activity, and poor clinical outcomes in patients with COVID-19.
Collapse
Affiliation(s)
- Ziyu Zhang
- Versiti Blood Research Institute, Milwaukee, Wisconsin, USA
| | - Wen Dai
- Versiti Blood Research Institute, Milwaukee, Wisconsin, USA
| | - Wen Zhu
- Versiti Blood Research Institute, Milwaukee, Wisconsin, USA
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Maya Rodriguez
- Versiti Blood Research Institute, Milwaukee, Wisconsin, USA
- Diversity Summer Health-Related Research Education Program (DSHREP), Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- College of Arts and Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Hayley Lund
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Yuhe Xia
- Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Yiliang Chen
- Versiti Blood Research Institute, Milwaukee, Wisconsin, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Mary Rau
- Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Ellen Anje Schneider
- Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Mary Beth Graham
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Shawn Jobe
- Versiti Blood Research Institute, Milwaukee, Wisconsin, USA
- Center for Bleeding and Clotting Disorders, Michigan State University, Lansing, Michigan, USA
| | - Demin Wang
- Versiti Blood Research Institute, Milwaukee, Wisconsin, USA
| | - Weiguo Cui
- Versiti Blood Research Institute, Milwaukee, Wisconsin, USA
| | - Renren Wen
- Versiti Blood Research Institute, Milwaukee, Wisconsin, USA
| | - Sidney W. Whiteheart
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, Kentucky, USA
- Divison of Cardiovascular Medicine, Gill Heart and Vascular Institute, University of Kentucky, Lexington, Lexington, Kentucky, USA
| | - Jeremy P. Wood
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, Kentucky, USA
- Divison of Cardiovascular Medicine, Gill Heart and Vascular Institute, University of Kentucky, Lexington, Lexington, Kentucky, USA
| | - Roy Silverstein
- Versiti Blood Research Institute, Milwaukee, Wisconsin, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jeffery S. Berger
- Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
- Department of Surgery, New York University Langone Health, New York, New York, USA
| | - Lisa Baumann Kreuziger
- Versiti Blood Research Institute, Milwaukee, Wisconsin, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Tessa J. Barrett
- Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Ze Zheng
- Versiti Blood Research Institute, Milwaukee, Wisconsin, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
281
|
Gaspar V, Silva B, Ambrioso I, Alves C, Alçada M. Right Ventricle Thrombus in a Massive Pulmonary Embolism COVID-19 Patient. Cureus 2023; 15:e43937. [PMID: 37746420 PMCID: PMC10513353 DOI: 10.7759/cureus.43937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/22/2023] [Indexed: 09/26/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) infection presents with a wild range of clinical manifestations. Increased inflammatory response and thrombotic risk have been described, being pulmonary embolism a potential cause of death in these patients. Pulmonary embolisms with right ventricle thrombus are rare and have higher mortality rates. This case report concerns a rare clinical presentation of a 75-year-old male with a medical history of right renal transplantation 36 years ago, that presented with a ten-day history of asthenia, followed by fever, shortness of breath, and cough since the day before. He was admitted with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) pneumonia and respiratory insufficiency. The next morning the patient worsened, he presented with hypotension, tachycardia, severe refractory hypoxemia, and chest pain. Contrast CT showed a massive pulmonary embolism with a right ventricle thrombus, confirmed by an echocardiogram. Anticoagulation and IV fluids were started, and the patient was transferred to the ICU. He developed obstructive shock, so thrombolysis was performed with a full dose of alteplase. The outcome was good with complete recovery. Posterior investigation excluded other causes for pulmonary embolism. The severity of pulmonary parenchymal disease secondary to COVID-19 correlates with thromboembolic complications, which demand a swift response to avoid death. An abrupt deterioration in oxygenation should raise suspicion for PE in COVID-19 patients, and mostly in the presence of hypotension and tachycardia. In our case report, there was a massive pulmonary embolism with a rare right ventricle thrombus that had a good outcome with medical treatment.
Collapse
Affiliation(s)
- Vasco Gaspar
- Internal Medicine Department, Hospital Distrital de Santarém, Santarém, PRT
| | - Bernardo Silva
- Internal Medicine Department, Hospital Distrital de Santarém, Santarém, PRT
| | - Inês Ambrioso
- Internal Medicine Department, Hospital Distrital de Santarém, Santarém, PRT
| | - Cláudia Alves
- Internal Medicine Department, Hospital Distrital de Santarém, Santarém, PRT
| | - Martim Alçada
- Internal Medicine Department, Hospital Distrital de Santarém, Santarém, PRT
| |
Collapse
|
282
|
Zhu J, Yan H, Shi M, Zhang M, Lu J, Wang J, Chen L, Wang Y, Li L, Miao L, Zhang H. Luteolin inhibits spike protein of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) binding to angiotensin-converting enzyme 2. Phytother Res 2023; 37:3508-3521. [PMID: 37166054 DOI: 10.1002/ptr.7826] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 03/16/2023] [Accepted: 03/24/2023] [Indexed: 05/12/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19), a respiratory illness that poses a serious threat to global public health. In an essential step during infection, SARS-CoV-2 uses the receptor-binding domain (RBD) of the spike (S) protein to engage with angiotensin-converting enzyme 2 (ACE2) in host cells. Chinese herbal medicines and their active components exhibit antiviral activity, with luteolin being a flavonoid that can significantly inhibit SARS-CoV infection. However, whether it can block the interaction between the S-protein RBD of SARS-CoV-2 and ACE2 has not yet been elucidated. Here, we investigated the effects of luteolin on the binding of the S-protein RBD to ACE2. By employing a competitive binding assay in vitro, we found that luteolin significantly blocked the binding of S-protein RBD to ACE2 with IC50 values of 0.61 mM, which was confirmed by the neutralized infection with SARS-CoV-2 pseudovirus in vivo. A surface plasmon resonance-based competition assay revealed that luteolin significantly affects the binding of the S-protein RBD to the ACE2 receptor. Molecular docking was performed to predict the binding sites of luteolin to the S-protein RBD-ACE2 complex. The active binding sites were defined based on published literature, and we found that luteolin might interfere with the mixture at residues including LYS353, ASP30, and TYR83 in the cellular ACE2 receptor and GLY496, GLN498, TYR505, LEU455, GLN493, and GLU484 in the S-protein RBD. These residues may together form attractive charges and destroy the stable interaction of S-protein RBD-ACE2. Luteolin also inhibits SARS-CoV-2 spike protein-induced platelet spreading, thereby inhibiting the binding of the spike protein to ACE2. Our results are the first to provide evidence that luteolin is an anti-SARS-CoV-2 agent associated with interference between viral S-protein RBD-ACE2 interactions.
Collapse
Affiliation(s)
- Junjie Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Huimin Yan
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Mengyao Shi
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Min Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jia Lu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiabao Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lu Chen
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yu Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lin Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lin Miao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Han Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
283
|
Gloeck N, Jaca A, Kredo T, Calligaro G. Cochrane Corner: The use of anticoagulants in patients hospitalised with COVID-19. SOUTHERN AFRICAN JOURNAL OF CRITICAL CARE 2023; 39:10.7196/SAJCC.2023.v39i2.450. [PMID: 37547768 PMCID: PMC10399546 DOI: 10.7196/sajcc.2023.v39i2.450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 05/28/2023] [Indexed: 08/08/2023] Open
Abstract
Background In this Cochrane Corner, we highlight the main findings of a Cochrane Review by Flumignan et al. entitled 'Anticoagulants for people hospitalised with COVID-19' and discuss the implications of these findings for research and practice in South Africa. In particular, we underscore the need for additional, high-quality, randomised controlled trials comparing different intensities of anticoagulation in patients with COVID-19 illness. Individuals in the intensive care unit and those hospitalised with another illness who are incidentally found to be infected with SARS-CoV-2 should still only be treated with prophylactic-dose low-molecular-weight heparin. Contributions of the study This Cochrane Corner summarises findings in a recent systematic review on the use of anticoagulation in people hospitalised with COVID-19, and provides insights on the implications of these findings for implementation by clinicians in South Africa. It highlights the need for clinicians to balance the benefits and harms of providing an anticoagulant, while considering the patients underlying risk for bleeding and thromboembolism.
Collapse
Affiliation(s)
- N Gloeck
- Cochrane South Africa, South African Medical Research Council, Parow Valley, South Africa
- Health Systems Research Unit, South African Medical Research Council, Parow Valley, South Africa
| | - A Jaca
- Cochrane South Africa, South African Medical Research Council, Parow Valley, South Africa
| | - T Kredo
- Cochrane South Africa, South African Medical Research Council, Parow Valley, South Africa
- Health Systems Research Unit, South African Medical Research Council, Parow Valley, South Africa
| | - G Calligaro
- Division of Pulmonology, Department of Medicine, University of Cape Town, South Africa
| |
Collapse
|
284
|
Aldali JA, Aldali HJ, Aljohani R, Algahtani M, Meo SA, Alharbi S, Al-Afghani H, Aldabaseh LN, Al Rubai EH, Fallata A, Zahrani SA, Al Zahrani MA. Implications of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Infected Hospitalised Patients with Co-Infections and Clinical Outcomes. Microorganisms 2023; 11:1921. [PMID: 37630481 PMCID: PMC10458585 DOI: 10.3390/microorganisms11081921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
The clinical severity of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection may rise because of acquiring a co-infection during the hospital stay of the patients. The rate of hospital co-infection alongside COVID-19 infection remains low. However, the mortality rates and intensive care unit (ICU) admission remains ambiguous. The present study investigates the implications of COVID-19 hospitalised infected patients with co-infection and the clinical outcomes. In this study, 142 patients were included. The eligible patients who tested positive for COVID-19 infection were hospitalised for more than two days. Each patient's characteristics and laboratory results were collected, such as who was admitted to the intensive care unit and who was discharged or expired. The results revealed that out of the 142 hospitalised patients, 25 (17.6%) were co-infection positive, and 12 identified types of co-infection: two Gram-positive bacterial infections, one fungal infection and nine Gram-negative bacterial infections. In addition, 33 (23.2%) were ICU admitted, 21 were co-infection negative and 12 were co-infection positive. Among the 12 ICU admitted with co-infection, 33.4% were discharged. The death rate and ICU admission had a p-value < 0.05, indicating statistical significance for co-infected patients compared to non-co-infected patients. It was concluded that co-infection remains very low within hospitalised COVID-19-infected patients but can have severe outcomes with increased ICU admission and increased mortality rates. Thus, implementing infection preventive measures to minimize the spread of hospital-acquired infections among COVID-19 hospitalised patients.
Collapse
Affiliation(s)
- Jehad A. Aldali
- Department of Pathology, College of Medicine, Imam Mohammad Ibn Saud Islamic University, Riyadh 13317, Saudi Arabia;
| | - Hamzah J. Aldali
- Cellular and Molecular Medicine, College of Biomedical Science, University of Bristol, Bristol City BS8 1QU, UK
| | - Razan Aljohani
- Hematology and Immunology, Faculty of Applied Medical Sciences, Tabuk University, Tabuk City 47512, Saudi Arabia
| | - Mohammad Algahtani
- Department of Laboratory and Blood Bank, Security Forces Hospital, Makkah 24251, Saudi Arabia (H.A.-A.)
| | - Sultan Ayoub Meo
- Department of Physiology, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| | - Saad Alharbi
- Department of Laboratory, Comprehensive Specialized Clinics, Security Forces Hospital, Jeddah 11481, Saudi Arabia
| | - Hani Al-Afghani
- Department of Laboratory and Blood Bank, Security Forces Hospital, Makkah 24251, Saudi Arabia (H.A.-A.)
| | | | | | - Abdulaziz Fallata
- Department of Medicine, Security Forces Hospital, Makkah 24251, Saudi Arabia
| | | | | |
Collapse
|
285
|
Pagliarin LG, de Oliveira LM, dos Anjos VNF, de Souza CDBT, Peiter GC, Façanha Wendel C, Dillmann Groto A, Freire de Melo F, Teixeira KN. In silico evidence of Remdesivir action in blood coagulation cascade modulation in COVID-19 treatment. World J Biol Chem 2023; 14:72-83. [PMID: 37547340 PMCID: PMC10401403 DOI: 10.4331/wjbc.v14.i4.72] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 04/30/2023] [Accepted: 07/03/2023] [Indexed: 07/25/2023] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) has demonstrated several clinical manifestations which include not only respiratory system issues but also liver, kidney, and other organ injuries. One of these abnormalities is coagulopathies, including thrombosis and disseminated intravascular coagulation. Because of this, the administration of low molecular weight heparin is required for patients that need to be hospitalized. In addition, Remdesivir is an antiviral that was used against Middle East Acute Respiratory Syndrome, Ebola, Acute Respiratory Syndrome, and other diseases, showing satisfactory results on recovery. Besides, there is evidence suggesting that this medication can provide a better prognosis for patients with COVID-19. AIM To investigate in silico the interaction between Remdesivir and clotting factors, pursuing a possibility of using it as medicine. METHODS In this in silico study, the 3D structures of angiotensin-converting enzyme 2 (ACE2), Factor I (fibrinogen), Factor II (prothrombin), Factor III (thromboplastin), Factor V (proaccelerin), Factor VII (proconvertin), Factor VIII (antihemophilic factor A), Factor IX (antihemophilic factor B), Factor X (Stuart-Prower factor), and Factor XI (precursor of thromboplastin (these structures are technically called receptors) were selected from the Protein Data Bank. The structures of the antivirals Remdesivir and Osetalmivir (these structures are called ligands) were selected from the PubChem database, while the structure of Atazanavir was selected from the ZINC database. The software AutoDock Tools (ADT) was used to prepare the receptors for molecular docking. Ions, peptides, water molecules, and other ones were removed from each ligand, and then, hydrogen atoms were added to the structures. The grid box was delimited and calculated using the same software ADT. A physiological environment with pH 7.4 is needed to make the ligands interact with the receptors, and still the software Marvin sketch® (ChemAxon®) was used to forecast the protonation state. To perform molecular docking, ADT and Vina software was connected. Using PyMol® software and Discovery studio® software from BIOVIA, it was possible to analyze the amino acid residues from receptors that were involved in the interactions with the ligands. Ligand tortions, atoms that participated in the interactions, and the type, strength, and duration of the interactions were also analyzed using those software. RESULTS Molecular docking analysis showed that Remdesivir and ACE2 had an affinity energy of -8.8 kcal/moL, forming a complex with eight hydrogen bonds involving seven atoms of Remdesivir and five amino acid residues of ACE2. Remdesivir and prothrombin had an interaction with six hydrogen bonds involving atoms of the drug and five amino acid residues of the clotting factor. Similar to that, Remdesivir and thromboplastin presented interactions via seven hydrogen bonds involving five atoms of the drug and four residues of the clotting factor. While Remdesivir and Factor V established a complex with seven hydrogen bonds between six antiviral atoms and six amino acid residues from the factor, and Factor VII connected with the drug by four hydrogen bonds, which involved three atoms of the drug and three residues of amino acids of the factor. The complex between Remdesivir and Factor IX formed an interaction via 11 hydrophilic bonds with seven atoms of the drug and seven residues of the clotting factor, plus one electrostatic bond and three hydrophobic interactions. Factor X and Remdesivir had an affinity energy of -9.6 kcal/moL, and the complex presented 10 hydrogen bonds and 14 different hydrophobic interactions which involved nine atoms of the drug and 16 amino acid residues of the clotting factor. The interaction between Remdesivir and Factor XI formed five hydrogen bonds involving five amino acid residues of the clotting factor and five of the antiviral atoms. CONCLUSION Because of the in silico significant affinity, Remdesivir possibly could act in the severe acute respiratory syndrome coronavirus 2 infection blockade by interacting with ACE2 and concomitantly act in the modulation of the coagulation cascade preventing the hypercoagulable state.
Collapse
Affiliation(s)
| | | | | | | | - Gabrielle Caroline Peiter
- Programa Multicêntrico de Pós-graduação em Bioquímica e Biologia Molecular - Setor Palotina, Universidade Federal do Paraná, Palotina 85.950-000, Paraná, Brazil
| | | | | | - Fabrício Freire de Melo
- Instituto Multidisciplinar em Saúde - Campus Anísio Teixeira, Universidade Federal da Bahia, Vitória da Conquista 45.029-094, Bahia, Brazil
| | - Kádima Nayara Teixeira
- Campus Toledo, Universidade Federal do Paraná, Toledo 85.919-899, Paraná, Brazil
- Programa Multicêntrico de Pós-graduação em Bioquímica e Biologia Molecular - Setor Palotina, Universidade Federal do Paraná, Palotina 85.950-000, Paraná, Brazil
| |
Collapse
|
286
|
Gao S, Tang AT, Wang M, Buchholz DW, Imbiakha B, Yang J, Chen X, Hewins P, Mericko-Ishizuka P, Leu NA, Sterling S, August A, Jurado KA, Morrisey EE, Aguilar-Carreno H, Kahn ML. Endothelial SARS-CoV-2 infection is not the underlying cause of COVID19-associated vascular pathology in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.24.550352. [PMID: 37546961 PMCID: PMC10402014 DOI: 10.1101/2023.07.24.550352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Endothelial damage and vascular pathology have been recognized as major features of COVID-19 since the beginning of the pandemic. Two main theories regarding how Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) damages endothelial cells and causes vascular pathology have been proposed: direct viral infection of endothelial cells or indirect damage mediated by circulating inflammatory molecules and immune mechanisms. However, these proposed mechanisms remain largely untested in vivo. Here, we utilized a set of new mouse genetic tools 1 developed in our lab to test both the necessity and sufficiency of endothelial human angiotensin-converting enzyme 2 (hACE2) in COVID19 pathogenesis. Our results demonstrate that endothelial ACE2 and direct infection of vascular endothelial cells does not contribute significantly to the diverse vascular pathology associated with COVID-19.
Collapse
|
287
|
Pacheco V, Cuber Guimarães R, Corrêa-Moreira D, Magalhães CE, Figueiredo D, Guttmann P, Trindade GF, da Silva JFA, Ano Bom APD, de Lourdes Maia M, Melgaço JG, da Costa Barros TA, da Silva AMV, Group C, Oliveira MME. Clinical Profile of SARS-CoV-2 Infection: Mechanisms of the Cellular Immune Response and Immunogenetic Markers in Patients from Brazil. Viruses 2023; 15:1609. [PMID: 37515295 PMCID: PMC10385121 DOI: 10.3390/v15071609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/13/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
OBJECTIVES The aim of this study is to evaluate some mechanisms of the immune response of people infected with SARS-CoV-2 in both acute infection and early and late convalescence phases. METHODS This is a cohort study of 70 cases of COVID-19, confirmed by RT-PCR, followed up to 60 days. Plasma Samples and clinical data were. Viral load, blood count, indicators inflammation were the parameters evaluated. Cellular immune response was evaluated by flow cytometry and Luminex immunoassays. RESULTS In the severe group, hypertension was the only reported comorbidity. Non severe patients have activated memory naive CD4+ T cells. Critically ill patients have central memory CD4+ T cell activation. Severe COVID-19 patients have both central memory and activated effector CD8+ T cells. Non-severe COVID-19 cases showed an increase in IL1β, IL-6, IL-10 and TNF and severely ill patients had higher levels of the cytokines IL-6, IL-10 and CXCL8. CONCLUSIONS The present work showed that different cellular responses are observed according to the COVID-19 severity in patients from Brazil an epicenter the pandemic in South America. Also, we notice that some cytokines can be used as predictive markers for the disease outcome, possibility implementation of strategies effective by health managers.
Collapse
Affiliation(s)
- Vanessa Pacheco
- Quality Assurance Department, Bio-Manguinhos-Fiocruz, Rio de Janeiro 21040-900, Brazil
| | | | - Danielly Corrêa-Moreira
- Laboratory of Taxonomy, Biochemistry and Bioprospecting of Fungi, Oswaldo Cruz Institute-Fiocruz, Rio de Janeiro 21041-250, Brazil
| | - Carlos Eduardo Magalhães
- UERJ-Universitary Hospital Pedro Ernesto, Outpatient Vascular Surgery, Rio de Janeiro 20950-003, Brazil
| | - Douglas Figueiredo
- UERJ-Universitary Hospital Pedro Ernesto, Outpatient Vascular Surgery, Rio de Janeiro 20950-003, Brazil
| | - Patricia Guttmann
- Municial Health Secretary of Rio de Janeiro, Rio de Janeiro 20211-110, Brazil
| | | | | | - Ana Paula Dinis Ano Bom
- Laboratory of Imunologic Tecnology, Bio-Manguinhos-Fiocruz, Rio de Janeiro 21040-900, Brazil
| | | | - Juliana Gil Melgaço
- Laboratory of Imunologic Tecnology, Bio-Manguinhos-Fiocruz, Rio de Janeiro 21040-900, Brazil
| | | | | | | | | |
Collapse
|
288
|
Mavropoulou X, Psoma E, Papachristodoulou A, Pyrrou N, Spanou E, Alexandratou M, Sidiropoulou M, Theocharidou A, Rafailidis V, Chrysanthidis T, Prassopoulos P. Gastrointestinal Imaging Findings in the Era of COVID-19: A Pictorial Review. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1332. [PMID: 37512143 PMCID: PMC10385728 DOI: 10.3390/medicina59071332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023]
Abstract
The potentially fatal COVID-19 pandemic has been associated with a largespectrum of clinical presentations. Beyond the classical pulmonary manifestations, gastrointestinal tract-related symptoms suchas nausea, diarrhea, abdominal distention and pain have been observed in patients, as a consequence of the binding of SARS-CoV-19 to Angiotensin-converting Enzyme 2 (ACE2) receptors in the gastrointestinal (GI) tract. The early recognition ofspecific imaging features, including hepatobiliary involvement, pancreatic involvement, development of solid organ infarcts, ischemic bowel changes and vascular occlusion, plays a key role through the course of the disease. Also, suspicious symptoms, especially in critically ill patients with clinical and biochemical markers of hypovolemia, necessitate timely imaging for bleeding complications. The aim of this pictorial review is to illustrate the spectrum of the GIimaging findings in patients with COVID-19. Awareness of diagnostic imaging hallmarks is crucial to optimize the management of these patients.
Collapse
Affiliation(s)
- Xanthippi Mavropoulou
- Department of Clinical Radiology, AHEPA University Hospital of Thessaloniki, Aristotle University of Thessaloniki, 54634 Thessaloniki, Greece
| | - Elisavet Psoma
- Department of Clinical Radiology, AHEPA University Hospital of Thessaloniki, Aristotle University of Thessaloniki, 54634 Thessaloniki, Greece
| | - Angeliki Papachristodoulou
- Department of Clinical Radiology, AHEPA University Hospital of Thessaloniki, Aristotle University of Thessaloniki, 54634 Thessaloniki, Greece
| | - Nikoletta Pyrrou
- Department of Clinical Radiology, AHEPA University Hospital of Thessaloniki, Aristotle University of Thessaloniki, 54634 Thessaloniki, Greece
| | - Ekaterini Spanou
- Department of Clinical Radiology, AHEPA University Hospital of Thessaloniki, Aristotle University of Thessaloniki, 54634 Thessaloniki, Greece
| | - Maria Alexandratou
- Department of Clinical Radiology, AHEPA University Hospital of Thessaloniki, Aristotle University of Thessaloniki, 54634 Thessaloniki, Greece
| | - Maria Sidiropoulou
- Department of Clinical Radiology, AHEPA University Hospital of Thessaloniki, Aristotle University of Thessaloniki, 54634 Thessaloniki, Greece
| | - Anastasia Theocharidou
- Department of Clinical Radiology, AHEPA University Hospital of Thessaloniki, Aristotle University of Thessaloniki, 54634 Thessaloniki, Greece
| | - Vasileios Rafailidis
- Department of Clinical Radiology, AHEPA University Hospital of Thessaloniki, Aristotle University of Thessaloniki, 54634 Thessaloniki, Greece
| | - Theofilos Chrysanthidis
- Infectious Diseases Division, First Internal Medicine Department, School of Medicine, Aristotle University of Thessaloniki, 54634 Thessaloniki, Greece
| | - Panos Prassopoulos
- Department of Clinical Radiology, AHEPA University Hospital of Thessaloniki, Aristotle University of Thessaloniki, 54634 Thessaloniki, Greece
| |
Collapse
|
289
|
Khorasani Esmaili P, Dabiri S, Movahedinia S, Shojaeepour S, Bagheri F, Ranjbar H, Shamsi Meymandi M, Mohebbi E, Farrokhnia M. Evaluation of Laboratory Findings of Patients with Coronavirus Disease 2019 in Kerman, Iran. IRANIAN JOURNAL OF PATHOLOGY 2023; 18:347-355. [PMID: 37942197 PMCID: PMC10628381 DOI: 10.30699/ijp.2023.1971332.3031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 05/30/2023] [Indexed: 11/10/2023]
Abstract
Background & Objective Since December 2019 in Wuhan, China there is a new form of pneumonia and after expansion in other countries, World Health Organization (WHO) called it Coronavirus Disease 2019 (COVID-19). Since the clinical laboratory findings have played an important role in the progression of the disease, this study aimed to evaluate the laboratory findings in COVID-19 patients (before vaccination). Methods In this case-control study that was conducted from February to August 2020; the laboratory test status in 101 positive COVID-19 patients was evaluated and compared with 101 healthy individuals. Results The results of our study showed that 21% of patients had low WBC, 24.75% low RBC, 37.62%, low Hb, 18.81% with low HCT, 29.7%, low Plt, 41.58% had High PT, 71.29% high CRP, 17.82% high urea, 11.88% high CR, 15.84% high LDH, 10.89% low sodium, 14.75% low potassium (K). The quantitative examination of blood factors showed that lymph%, mixed%, PLT, HCT, Hb, and RBC were higher in the control group than in the case group. While Neu%, WBC, PTT, CRP, UREA, LDH, K in the patient group were higher than in the control group. Conclusion According to the results of the study, it can be concluded that in the clinical treatment of COVID-19 patients, much attention should be paid to the laboratory indicators to identify and intervene early in critically ill patients.
Collapse
Affiliation(s)
- Parisa Khorasani Esmaili
- Department of Pathology, Pathology and Stem Cells Research Center, Afzali Pour Medical Faculty, Kerman University of Medical Sciences, Kerman, Iran
| | - Shahriar Dabiri
- Department of Pathology, Pathology and Stem Cells Research Center, Afzali Pour Medical Faculty, Kerman University of Medical Sciences, Kerman, Iran
| | - Sajjadeh Movahedinia
- Department of Pathology, Pathology and Stem Cells Research Center, Afzali Pour Medical Faculty, Kerman University of Medical Sciences, Kerman, Iran
| | - Saeedeh Shojaeepour
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Fatemeh Bagheri
- Legal Medicine Research Center, Legal Medicine Organization, Kerman, Iran
| | - Hanieh Ranjbar
- Department of Pathology, Pathology and Stem Cells Research Center, Afzali Pour Medical Faculty, Kerman University of Medical Sciences, Kerman, Iran
| | - Manzumeh Shamsi Meymandi
- Department of Pathology, Pathology and Stem Cells Research Center, Afzali Pour Medical Faculty, Kerman University of Medical Sciences, Kerman, Iran
| | - Elham Mohebbi
- Research Center for Modeling in Health, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Iran
| | - Mehrdad Farrokhnia
- Infectious and Internal Medicine Department, Afzalipour Hospital, Kerman University of Medical Science, Kerman, Iran
| |
Collapse
|
290
|
Yang K, Liu S, Yan H, Lu W, Shan X, Chen H, Bao C, Feng H, Liao J, Liang S, Xu L, Tang H, Yuan JXJ, Zhong N, Wang J. SARS-CoV-2 spike protein receptor-binding domain perturbates intracellular calcium homeostasis and impairs pulmonary vascular endothelial cells. Signal Transduct Target Ther 2023; 8:276. [PMID: 37452066 PMCID: PMC10349149 DOI: 10.1038/s41392-023-01556-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/09/2023] [Accepted: 06/28/2023] [Indexed: 07/18/2023] Open
Abstract
Exposure to the spike protein or receptor-binding domain (S-RBD) of SARS-CoV-2 significantly influences endothelial cells and induces pulmonary vascular endotheliopathy. In this study, angiotensin-converting enzyme 2 humanized inbred (hACE2 Tg) mice and cultured pulmonary vascular endothelial cells were used to investigate how spike protein/S-RBD impacts pulmonary vascular endothelium. Results show that S-RBD leads to acute-to-prolonged induction of the intracellular free calcium concentration ([Ca2+]i) via acute activation of TRPV4, and prolonged upregulation of mechanosensitive channel Piezo1 and store-operated calcium channel (SOCC) key component Orai1 in cultured human pulmonary arterial endothelial cells (PAECs). In mechanism, S-RBD interacts with ACE2 to induce formation of clusters involving Orai1, Piezo1 and TRPC1, facilitate the channel activation of Piezo1 and SOCC, and lead to elevated apoptosis. These effects are blocked by Kobophenol A, which inhibits the binding between S-RBD and ACE2, or intracellular calcium chelator, BAPTA-AM. Blockade of Piezo1 and SOCC by GsMTx4 effectively protects the S-RBD-induced pulmonary microvascular endothelial damage in hACE2 Tg mice via normalizing the elevated [Ca2+]i. Comparing to prototypic strain, Omicron variants (BA.5.2 and XBB) of S-RBD induces significantly less severe cell apoptosis. Transcriptomic analysis indicates that prototypic S-RBD confers more severe acute impacts than Delta or Lambda S-RBD. In summary, this study provides compelling evidence that S-RBD could induce persistent pulmonary vascular endothelial damage by binding to ACE2 and triggering [Ca2+]i through upregulation of Piezo1 and Orai1. Targeted inhibition of ACE2-Piezo1/SOCC-[Ca2+]i axis proves a powerful strategy to treat S-RBD-induced pulmonary vascular diseases.
Collapse
Affiliation(s)
- Kai Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Shiyun Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Han Yan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| | - Wenju Lu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaoqian Shan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Haixia Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Pathology, The Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Changlei Bao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Huazhuo Feng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jing Liao
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Shuxin Liang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lei Xu
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Haiyang Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China.
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China.
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
291
|
Zhong M, Gao Y, Hu H, Zhu X, Gan L, Li L, Xiang C, Yan Y, Dai Z. Transient low T3 syndrome in patients with COVID-19: a new window for prediction of disease severity. Front Endocrinol (Lausanne) 2023; 14:1154007. [PMID: 37522120 PMCID: PMC10374310 DOI: 10.3389/fendo.2023.1154007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
Objective To investigate the relationship of low T3 syndrome with disease severity in patients with COVID-19. Methods The clinical data of 145 patients with COVID-19 were retrospectively collected, and patients were divided into a low T3 group and a normal T3 group. Logistic regression models were used to assess predictive performance of FT3. Receiver operating characteristic (ROC) analysis was used to evaluate the use of low T3 syndrome in predicting critical disease. Kaplan-Meier analysis was used to analyze the impact of low T3 syndrome on mortality. Results The prevalence of low T3 level among COVID-19 patients was 34.48%. The low T3 group was older, and had lower levels of hemoglobin, lymphocytes, prealbumin, and albumin, but higher levels of white blood cells, neutrophils, CRP, ESR, and D-dimer (all p<0.05). The low T3 group had greater prevalences of critical disease and mortality (all p <0.05). Multivariate logistic regression analysis showed that the Lymphocytes, free T3 (FT3), and D-dimer were independent risk factors for disease severity in patients with COVID-19. ROC analysis showed that FT3, lymphocyte count, and D-dimer, and all three parameters together provided reliable predictions of critical disease. Kaplan-Meier analysis showed the low T3 group had increased mortality (p<0.001). Six patients in the low T3 group and one patient in the normal T3 group died. All 42 patients whose T3 levels were measured after recovery had normal levels after discharge. Conclusion Patients with COVID-19 may have transient low T3 syndrome at admission, and this may be useful for predicting critical illness.
Collapse
Affiliation(s)
- Mingyao Zhong
- Department of Endocrinology, Xiaogan Hospital Affiliated to Wuhan University of Science and Technology, The Central Hospital of Xiaogan, Xiaogan, Hubei, China
- Department of Internal Medicine, Medical College of Wuhan University of Science and Technology, Wuhan, China
| | - Yue Gao
- Department of Internal Medicine, Medical College of Wuhan University of Science and Technology, Wuhan, China
| | - Hongling Hu
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xuan Zhu
- Department of Internal Medicine, Medical College of Wuhan University of Science and Technology, Wuhan, China
| | - Lulu Gan
- Department of Internal Medicine, Medical College of Wuhan University of Science and Technology, Wuhan, China
| | - Ling Li
- Department of Endocrinology, Xiaogan Hospital Affiliated to Wuhan University of Science and Technology, The Central Hospital of Xiaogan, Xiaogan, Hubei, China
| | - Cheng Xiang
- Department of Endocrinology, Xiaogan Hospital Affiliated to Wuhan University of Science and Technology, The Central Hospital of Xiaogan, Xiaogan, Hubei, China
| | - Yimin Yan
- Department of Endocrinology, Xiaogan Hospital Affiliated to Wuhan University of Science and Technology, The Central Hospital of Xiaogan, Xiaogan, Hubei, China
- Department of Internal Medicine, Medical College of Wuhan University of Science and Technology, Wuhan, China
| | - Zhe Dai
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
292
|
Niculae CM, Gorea ME, Tirlescu LG, Constantin RA, Moroti R, Hristea A. Pulmonary Thrombosis despite Therapeutic Anticoagulation in COVID-19 Pneumonia: A Case Report and Literature Review. Viruses 2023; 15:1535. [PMID: 37515221 PMCID: PMC10386232 DOI: 10.3390/v15071535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
The rate of thrombotic complications in COVID-19 patients is high and could be associated with the risk of unfavourable outcomes. Moreover, pulmonary thrombotic events can occur even in patients already on anticoagulant treatment. We present the case of a patient with severe COVID-19 pneumonia, without traditional risk factors for thrombosis, who developed massive pulmonary thrombosis (PT) despite therapeutic anticoagulation. The diagnosis was challenging, and the case raised concerns about the protective role of conventional anticoagulant treatment in COVID-19 pneumonia. Thus, we searched for literature reports on COVID-19 patients who developed PT despite being under anticoagulation therapy. We identified 13 cohort studies including 4058 patients of which 346 (8.5%) developed PT and nine case reports/series enrolling 14 patients. Four cohorts were further analysed, which reported data on risk factors for thrombosis, outcomes and biological characteristics. We found that there were no differences between patients with and without PT regarding the classical risk factors for thrombosis. PT occurred regardless of the anticoagulation regimen, and the risk factor identified was severe COVID-19 pneumonia and a stay in an intensive care unit (ICU). Pulmonary thrombotic events in patients with COVID-19 are rather inflammation-related than correlated with traditional thromboembolic risk factors, and the therapeutic approach must take into consideration this aspect.
Collapse
Affiliation(s)
- Cristian-Mihail Niculae
- Infectious Diseases Department, Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, 37 Dionisie Lupu Street, 050474 Bucharest, Romania
- National Institute for Infectious Diseases "Prof. Dr. Matei Bals", 1 Calistrat Grozovici Street, 021105 Bucharest, Romania
| | - Maria-Evelina Gorea
- National Institute for Infectious Diseases "Prof. Dr. Matei Bals", 1 Calistrat Grozovici Street, 021105 Bucharest, Romania
| | - Laura-Georgiana Tirlescu
- National Institute for Infectious Diseases "Prof. Dr. Matei Bals", 1 Calistrat Grozovici Street, 021105 Bucharest, Romania
| | - Rares-Alexandru Constantin
- National Institute for Infectious Diseases "Prof. Dr. Matei Bals", 1 Calistrat Grozovici Street, 021105 Bucharest, Romania
| | - Ruxandra Moroti
- Infectious Diseases Department, Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, 37 Dionisie Lupu Street, 050474 Bucharest, Romania
- National Institute for Infectious Diseases "Prof. Dr. Matei Bals", 1 Calistrat Grozovici Street, 021105 Bucharest, Romania
| | - Adriana Hristea
- Infectious Diseases Department, Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, 37 Dionisie Lupu Street, 050474 Bucharest, Romania
- National Institute for Infectious Diseases "Prof. Dr. Matei Bals", 1 Calistrat Grozovici Street, 021105 Bucharest, Romania
| |
Collapse
|
293
|
Xing Y, Li Y, Feng L, Huo R, Ma X, Dong Y, Liu D, Niu Y, Tian X, Chen E. Predictors of COVID-19 Severity in Elderly Patients Infected by Omicron in China, 18 December 2022-5 February 2023. Infect Drug Resist 2023; 16:4505-4518. [PMID: 37457796 PMCID: PMC10349581 DOI: 10.2147/idr.s418622] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/06/2023] [Indexed: 07/18/2023] Open
Abstract
Purpose To analyze the clinical characteristics and prognosis of patients hospitalized with non-severe, severe pneumonia and death in Omicron COVID-19. Patients and Methods We collected clinical data from 118 patients with COVID-19 in China from 18 December, 2022 and 5 February, 2023. According to the outcome, the patients were divided into non-severe group, severe group and death group. Subsequently, we statistically analyzed the general condition, clinical manifestations, laboratory parameters, NLR, MLR, PLR and HALP of these groups. We also retrospectively analyzed the possible factors affecting the prognostic regression of patients with COVID-19. Results A total of 118 COVID-19 patients were enrolled in this study, including 64 non-severe patients, 38 severe patients and 16 death patients. Compared with the non-severe group, T lymphocytes, B lymphocytes, Th1, Th2, Th17, Treg cells, IgA, IgG, IgM in the severe and death groups decreased more significantly (P<0.05). The levels of myocardial markers, ALT, AST, BUN, Cr, D-dimer, fibrinogen, NLR, MLR and PLR in the severe and death groups were significantly higher than those in the non-severe group (P<0.05). The level of HALP was significantly lower than that of non-severe group (P<0.05). MLR is not only an independent risk factor for the transition from non-severe to severe disease, but also an independent risk factor for predicting the possibility of death in COVID-19 patients. Conclusion The analysis of COVID-19 patients in China showed that severe patients were older, more likely to have related complications, lower lymphocyte count, liver and kidney function disorder, glucose and lipid metabolism disorders, myocardial injury, and abnormal coagulation function, suggesting the need for early anticoagulant therapy. In addition, NLR, MLR, PLR and HALP can be used as biomarkers to evaluate the severity and prognosis of COVID-19 patients.
Collapse
Affiliation(s)
- Yanqing Xing
- The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Yupeng Li
- The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Liting Feng
- The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Rujie Huo
- The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Xinkai Ma
- The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Yanting Dong
- The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Dai Liu
- The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Yuheng Niu
- The First Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Xinrui Tian
- The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Erjing Chen
- The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| |
Collapse
|
294
|
Zaira B, Yulianti T, Levita J. Correlation between Hepatocyte Growth Factor (HGF) with D-Dimer and Interleukin-6 as Prognostic Markers of Coagulation and Inflammation in Long COVID-19 Survivors. Curr Issues Mol Biol 2023; 45:5725-5740. [PMID: 37504277 PMCID: PMC10377933 DOI: 10.3390/cimb45070361] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/03/2023] [Accepted: 07/07/2023] [Indexed: 07/29/2023] Open
Abstract
In general, an individual who experiences the symptoms of Severe Acute Respiratory Syndrome Coronavirus 2 or SARS-CoV-2 infection is declared as recovered after 2 weeks. However, approximately 10-20% of these survivors have been reported to encounter long-term health problems, defined as 'long COVID-19', e.g., blood coagulation which leads to stroke with an estimated incidence of 3%, and pulmonary embolism with 5% incidence. At the time of infection, the immune response produces pro-inflammatory cytokines that stimulate stromal cells to produce pro-hepatocyte growth factor (pro-HGF) and eventually is activated into hepatocyte growth factor (HGF), which helps the coagulation process in endothelial and epithelial cells. HGF is a marker that appears as an inflammatory response that leads to coagulation. Currently, there is no information on the effect of SARS-CoV-2 infection on serum HGF concentrations as a marker of the prognosis of coagulation in long COVID-19 survivors. This review discusses the pathophysiology between COVID-19 and HGF, IL-6, and D-dimer.
Collapse
Affiliation(s)
- Bena Zaira
- Student at Master Program in Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia
- Prodia Education and Research Institute, Jakarta 10430, Indonesia
| | - Trilis Yulianti
- Prodia Education and Research Institute, Jakarta 10430, Indonesia
| | - Jutti Levita
- Department of Pharmacology and Clinical Pharmacy, Padjadjaran University, Sumedang 45363, Indonesia
| |
Collapse
|
295
|
Hogea T, Suciu BA, Chinezu L, Brinzaniuc K, Arbănași EM, Ungureanu A, Kaller R, Carașca C, Arbănași EM, Vunvulea V, Hălmaciu I, Mureșan AV, Russu E, Ciucanu CC, Radu CM, Radu CC. Pregnancy-Associated Spontaneous Coronary Acute Dissection as a Cause of Sudden Cardiac Death-Autopsy Findings and Literature Review: Is COVID-19 Related? MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1257. [PMID: 37512074 PMCID: PMC10386237 DOI: 10.3390/medicina59071257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/25/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023]
Abstract
Sudden cardiac death (SCD) is the leading cause of mortality globally (violent or non-violent), with few to no feasible remedies. The etiopathogenesis of SCD involves a complex and multilayered substrate in which dynamic factors interact with a preexistent cardiovascular pathology, which is often undiagnosed and untreated, leading to the rapid development of cardiac rhythm disorders and cardiac arrest. Cardiovascular disease is a rare but emerging factor in maternal mortality that can be justified by an upward trend in the mean age of pregnant individuals. Spontaneous coronary artery dissection (SCAD) is defined as a non-traumatic and non-iatrogenic separation of the coronary arterial wall by intramural hemorrhage with or without an intimal tear. The resulting intramural hematoma compresses the coronary arteries, reducing blood flow and causing myocardial ischemia. SCAD continues to be misdiagnosed, underdiagnosed, and managed as an atherosclerotic acute coronary syndrome, which may harm patients with SCAD. The latest research shows that individuals who have or have had coronavirus disease 2019 (COVID-19) may also present coagulation abnormalities, so infection with COVID-19 during pregnancy can increase this hypercoagulable condition, thus increasing the risk of SCAD and SCD. This present study reports two cases over 35 years, one being infected with SARS-COV2 one month before the event and the other being tested positive during admission, both asymptomatic, who were declared healthy on periodic clinical evaluations, with pregnancies over 35 weeks, with normal fetal development, which suddenly caused chest pain, dyspnea, and loss of consciousness, required emergency c-sections, and died suddenly after they were performed. In both cases, the cause of death was SCAD on the anterior-descending artery. In both cases, emergency percutaneous coronary intervention was performed. The second part of the study represents a literature overview of SCAD during COVID-19. In addition to pregnancy hormonal changes, other potential hormone-mediated SCAD triggers are still under discussion.
Collapse
Affiliation(s)
- Timur Hogea
- Department of Forensic Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania
- Institute of Forensic Medicine, 540141 Targu Mures, Romania
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania
| | - Bogdan Andrei Suciu
- Department of Anatomy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania
| | - Laura Chinezu
- Institute of Forensic Medicine, 540141 Targu Mures, Romania
- Department of Histology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania
| | - Klara Brinzaniuc
- Department of Anatomy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania
| | - Emil Marian Arbănași
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania
- Department of Anatomy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania
- Clinic of Vascular Surgery, Mures County Emergency Hospital, 540136 Targu Mures, Romania
- Department of Vascular Surgery, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania
| | - Ancuța Ungureanu
- Epidemiology Clinic, Mures County Hospital, 540072 Targu Mures, Romania
| | - Réka Kaller
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania
- Clinic of Vascular Surgery, Mures County Emergency Hospital, 540136 Targu Mures, Romania
| | - Cosmin Carașca
- Department of Forensic Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania
- Institute of Forensic Medicine, 540141 Targu Mures, Romania
| | - Eliza Mihaela Arbănași
- Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania
| | - Vlad Vunvulea
- Department of Anatomy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania
- Department of Radiology, Mures County Emergency Hospital, 540136 Targu Mures, Romania
| | - Ioana Hălmaciu
- Institute of Forensic Medicine, 540141 Targu Mures, Romania
- Department of Radiology, Mures County Emergency Hospital, 540136 Targu Mures, Romania
| | - Adrian Vasile Mureșan
- Clinic of Vascular Surgery, Mures County Emergency Hospital, 540136 Targu Mures, Romania
- Department of Vascular Surgery, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania
| | - Eliza Russu
- Clinic of Vascular Surgery, Mures County Emergency Hospital, 540136 Targu Mures, Romania
- Department of Vascular Surgery, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania
| | | | - Casandra Maria Radu
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 1 University Street, 410087 Oradea, Romania
| | - Carmen Corina Radu
- Department of Forensic Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania
| |
Collapse
|
296
|
Niculae CM, Hristea A, Albulescu AS, Petre VB, Anghel AMJ, Damalan AC, Bel AA, Lazar M. Quantitative chest CT imaging characteristics and outcome of patients with COVID-19 associated pulmonary artery thrombosis: A single-center retrospective cohort study. Medicine (Baltimore) 2023; 102:e34250. [PMID: 37417640 PMCID: PMC10328685 DOI: 10.1097/md.0000000000034250] [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: 03/09/2023] [Revised: 05/20/2023] [Accepted: 06/16/2023] [Indexed: 07/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19)-associated pulmonary thrombotic events occur frequently and are associated with disease severity and worse clinical outcomes. We aimed to describe the clinical and quantitative chest computed tomography (CT) imaging characteristics based on density ranges (Hounsfield units) and the outcomes of patients with COVID-19 associated pulmonary artery thrombosis. This retrospective cohort study included all patients with COVID-19 hospitalized in a tertiary care hospital between March 2020 and June 2022 who underwent a CT pulmonary angiography. We included 73 patients: 36 (49.3%) with and 37 (50.7%) without pulmonary artery thrombosis. The in-hospital all-cause mortality was 22.2 versus 18.9% ( P = .7), and the intensive care unit admission rates were 30.5 versus 8.1% ( P = .01) at the time of diagnosis of pulmonary artery thrombosis. Except for D-dimers (median of 3142 vs 533, P = .002), the other clinical, coagulopathy, and inflammatory markers were similar. Logistic regression analysis revealed that only D-dimers were associated with pulmonary artery thrombosis ( P = .012). ROC curve analysis of D-dimers showed that a value greater than 1716 ng/mL predicted pulmonary artery thrombosis with an area under the curve of 0.779, 72.2% sensitivity, and 73% specificity (95% CI 0.672-0.885). Peripheral distribution of pulmonary artery thrombosis was recorded in 94.5% of cases. In the lower lobes of the lungs, the incidence of pulmonary artery thrombosis was 6 times higher than that in the upper lobes (58-64%), with a percentage of lung injury of 80% to 90%. Analysis of the distribution of arterial branches with filling defects revealed that 91.6% occurred in lung areas with inflammatory lesions. Quantitative chest CT imaging provides valuable information regarding the extent of COVID-19 associated lung damage and can be used to anticipate the co-location of pulmonary immunothrombotic events. In patients with severe COVID-19, in-hospital all-cause mortality was similar regardless of the presence of associated distal pulmonary thrombosis.
Collapse
Affiliation(s)
- Cristian-Mihail Niculae
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
- National Institute for Infectious Diseases “Prof. Dr. Matei Bals”, Bucharest, Romania
| | - Adriana Hristea
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
- National Institute for Infectious Diseases “Prof. Dr. Matei Bals”, Bucharest, Romania
| | | | - Vladimir Bogdan Petre
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
| | | | - Anca-Cristina Damalan
- National Institute for Infectious Diseases “Prof. Dr. Matei Bals”, Bucharest, Romania
| | - Adela-Abigaela Bel
- National Institute for Infectious Diseases “Prof. Dr. Matei Bals”, Bucharest, Romania
| | - Mihai Lazar
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
- National Institute for Infectious Diseases “Prof. Dr. Matei Bals”, Bucharest, Romania
| |
Collapse
|
297
|
Smolyakov YN, Kuznik BI, Fefelova EV, Kazantseva LS, Shapovalov YK, Lukyanchuk MS, Lukyanov SA, Shapovalov KG. [Predictive role of erythrocytes in assessment of COVID-19 outcomes]. Vopr Virusol 2023; 68:198-204. [PMID: 37436411 DOI: 10.36233/0507-4088-166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Indexed: 07/13/2023]
Abstract
INTRODUCTION The search for affordable and accurate predictors of the outcome of COVID-19 is extremely important, as it provides the possibility to effectively correct the patient treatment tactics. AIM OF THE STUDY To develop simple and accurate criteria based on the dynamics of red blood counts that predict the outcome of COVID-19. MATERIALS AND METHODS Observations were carried out in 125 patients with severe and extremely severe COVID-19, in whom indicators characterizing the state of red blood were determined in dynamics on days 1, 5, 7, 10, 14 and 21 after the hospitalization. ROC analysis was performed to calculate the threshold predictive values for survival and mortality. RESULTS The total number of erythrocytes and the level of hemoglobin in severe and extremely severe patients did not go beyond the acceptable limits, although showed a tendency to decrease in the group of fatal cases. On the 1st and 21st days, the number of MacroR in the deceased patients was reduced compared to those in group of survivors. It has been established that the RDW-CV test can predict the outcome of the COVID-19 with a high degree of probability at a relatively early stage of disease. RDW-SD test can be an additional predictive criterion of COVID-19 outcome. CONCLUSION The RDW-CV test can be used as an effective predictor of disease outcome in patients with severe COVID-19.
Collapse
|
298
|
Advani S, Hosseini SMM, Bozorgmehr R, Khameneh-Bagheri A, Mohammadzadeh S, Hasanzadeh T, Jalilian L, Vahidi M, Nofeli AH, Hooshyari Z. Abnormalities of brain imaging in COVID-19 patients with neurological symptoms. CURRENT JOURNAL OF NEUROLOGY 2023; 22:162-169. [PMID: 38011453 PMCID: PMC10626144 DOI: 10.18502/cjn.v22i3.13796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/09/2023] [Indexed: 11/29/2023]
Abstract
Background: Coronavirus disease 2019 (COVID-19) is a multisystem disease, manifested by several symptoms of various degrees. Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) can affect the central nervous system (CNS) through several mechanisms and brain imaging plays an essential role in the diagnosis and evaluation of the neurological involvement of COVID-19. Moreover, brain imaging of patients with COVID-19 would result in a better understanding of SARS-CoV-2 neuro-pathophysiology. In this study, we evaluated the brain imaging findings of patients with COVID-19 in Shohada-e Tajrish Hospital, Tehran, Iran. Methods: This was a single-center, retrospective, and observational study. The hospital records and chest and brain computed tomography (CT) scans of patients with confirmed COVID-19 were reviewed. Results: 161 patients were included in this study (39.1% women, mean age: 60.84). Thirteen patients (8%) had ischemic strokes identified by brain CT. Subdural hematoma, subdural effusion, and subarachnoid hemorrhage were confirmed in three patients. Furthermore, there were four cases of intracranial hemorrhage (ICH) and intraventricular hemorrhage (IVH). Patients with and without abnormal brain CTs had similar average ages. The rate of brain CT abnormalities in both genders did not differ significantly. Moreover, abnormal brain CT was not associated with increased death rate. There was no significant difference in lung involvement (according to lung CT scan) between the two groups. Conclusion: Our experience revealed a wide range of imaging findings in patients with COVID-19 and these findings were not associated with a more severe lung involvement or increased rate of mortality.
Collapse
Affiliation(s)
- Soroor Advani
- Men’s Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyyed Mohammad Mahdi Hosseini
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rama Bozorgmehr
- Clinical Research Development Unit, Shohada-e Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arash Khameneh-Bagheri
- Department of Radiology, Shohada-e Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sevda Mohammadzadeh
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Taha Hasanzadeh
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Laya Jalilian
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Vahidi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Hasan Nofeli
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Hooshyari
- Psychiatry and Psychology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
299
|
Bellanti F, Kasperczyk S, Kasperczyk A, Dobrakowski M, Pacilli G, Vurchio G, Maddalena A, Quiete S, Lo Buglio A, Capurso C, Serviddio G, Vendemiale G. Alteration of circulating redox balance in coronavirus disease-19-induced acute respiratory distress syndrome. J Intensive Care 2023; 11:30. [PMID: 37408073 PMCID: PMC10320967 DOI: 10.1186/s40560-023-00679-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/27/2023] [Indexed: 07/07/2023] Open
Abstract
BACKGROUND Mechanisms underpinning ARDS induced by COVID-19 are mostly immune-mediated, but need to be completely clarified. This study aimed to investigate redox balance in COVID-19 patients with ARDS, trying to recognize possible differences from typical ARDS related to the pathophysiology of severe disease. METHODS Patients affected by ARDS and positive for the SARS-CoV-2 virus (N = 40, COVID-19) were compared to ARDS patients negative to the molecular test (N = 42, No COVID-19). Circulating markers of redox balance were measured in serum and erythrocytes, and related to markers of inflammation and coagulability. RESULTS No differences in serum markers of oxidative damage were found between both groups, but a reduction in total antioxidant status and serum ceruloplasmin level was observed in COVID-19 rather than No COVID-19 patients. Redox balance alterations were described in erythrocytes from COVID-19 with respect to No COVID-19 group, characterized by increased lipofuscin and malondialdehyde concentration, and reduced glutathione S-transferase and glutathione reductase activity. These markers were associated with circulating indexes of respiratory disease severity (Horowitz index and alveolar-to-arterial oxygen gradient), inflammation (interleukin-6 and interleukin-10), and hypercoagulability (D-dimer) in COVID-19 patients with ARDS. CONCLUSIONS ARDS caused by COVID-19 is sustained by impairment of redox balance, particularly in erythrocytes. This alteration is associated with the pro-inflammatory and pro-coagulant status which characterizes severe COVID-19.
Collapse
Affiliation(s)
- Francesco Bellanti
- Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto, 1, 71122, Foggia, Italy.
| | - Sławomir Kasperczyk
- Department of Biochemistry, School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia, 41-808, Zabrze, Poland
| | - Aleksandra Kasperczyk
- Department of Biochemistry, School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia, 41-808, Zabrze, Poland
| | - Michał Dobrakowski
- Department of Biochemistry, School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia, 41-808, Zabrze, Poland
| | - Gabriella Pacilli
- Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto, 1, 71122, Foggia, Italy
| | - Giuseppina Vurchio
- Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto, 1, 71122, Foggia, Italy
| | - Alessandro Maddalena
- Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto, 1, 71122, Foggia, Italy
| | - Stefano Quiete
- Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto, 1, 71122, Foggia, Italy
| | - Aurelio Lo Buglio
- Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto, 1, 71122, Foggia, Italy
| | - Cristiano Capurso
- Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto, 1, 71122, Foggia, Italy
| | - Gaetano Serviddio
- Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto, 1, 71122, Foggia, Italy
| | - Gianluigi Vendemiale
- Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto, 1, 71122, Foggia, Italy
| |
Collapse
|
300
|
Abate V, Vergatti A, Casoria A, Zarrella AF, Muscariello R, Nuzzo V, Vargas M, Servillo G, Conca P, D'Elia L, Iannuzzo G, Venetucci P, De Filippo G, Rendina D, Tufano A. Venous thromboembolism and major bleeding in severe and critical COVID-19 hospitalized patients. Monaldi Arch Chest Dis 2023; 94. [PMID: 37403940 DOI: 10.4081/monaldi.2023.2617] [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: 04/24/2023] [Accepted: 06/07/2023] [Indexed: 07/06/2023] Open
Abstract
Venous thromboembolism (VTE) and major bleeding (MB) are life-threatening complications described in COVID-19 hospitalized patients, and they can be considered two sides of the same coin. This retrospective study aims to evaluate the risk factors for VTE and MB in COVID-19 patients admitted to two Italian hospitals. The medical records of all COVID-19 patients (males 139; 62.3%, mean age 67.2±13.6 years, body weight 88.2±20.6 kg) hospitalized from March 11th to July 31st, 2020, at the Federico II University Hospital and the Sea Hospital, Naples, Italy, were analyzed. The COVID-19 patients were classified into four groups: COVID-19 patients developing VTE and/or MB, COVID-19 patients developing only VTE, COVID-19 patients developing only MB, and COVID-19 patients not developing either VTE or MB. During the hospitalization, 53 COVID-19 patients (24.7%; males 40; 75.5%, mean age 67.2±13.6 years, weight 88.2±20.6 kg) developed VTE, 33 COVID-19 patients (15.3%; males 17; 51.5, mean age 67.3±14.9 years, weight 74.1±14.3 kg) developed MB, and 129 COVID-19 patients did not develop either TVP or MB. No parameters to identify severe COVID-19 complicated by VTE and/or MB were found. However, some clinical and biochemical parameters can be evaluated to predict the risk of MB in order to modify the treatment and take prompt action to reduce mortality.
Collapse
Affiliation(s)
- Veronica Abate
- Department of Clinical Medicine and Surgery, Federico II University of Naples.
| | - Anita Vergatti
- Department of Clinical Medicine and Surgery, Federico II University of Naples.
| | - Aniello Casoria
- Department of Clinical Medicine and Surgery, Federico II University of Naples.
| | | | | | - Vincenzo Nuzzo
- Endocrinology and Nutrition Department, Ospedale del Mare, Naples.
| | - Maria Vargas
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University of Naples.
| | - Giuseppe Servillo
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University of Naples.
| | - Paolo Conca
- Department of Clinical Medicine and Surgery, Federico II University of Naples.
| | - Lanfranco D'Elia
- Department of Clinical Medicine and Surgery, Federico II University of Naples.
| | - Gabriella Iannuzzo
- Department of Clinical Medicine and Surgery, Federico II University of Naples.
| | - Pietro Venetucci
- Department of Morphological and Functional Diagnostics, Radiotherapy, Forensic Medicine, Federico II University of Naples.
| | - Gianpaolo De Filippo
- Department of Pediatric Endocrinology and Diabetology, Robert Debré University Hospital, Assistance Publique-Hôpitaux de Paris.
| | - Domenico Rendina
- Department of Clinical Medicine and Surgery, Federico II University of Naples.
| | - Antonella Tufano
- Department of Clinical Medicine and Surgery, Federico II University of Naples.
| |
Collapse
|