Your browser doesn't support javascript.
Montrer: 20 | 50 | 100
Résultats 1 - 16 de 16
Filtre
1.
Int J Mol Sci ; 23(18)2022 Sep 09.
Article Dans Anglais | MEDLINE | ID: covidwho-2071501

Résumé

In SARS-CoV-2-infected humans, disease progression is often associated with acute respiratory distress syndrome involving severe lung injury, coagulopathy, and thrombosis of the alveolar capillaries. The pathogenesis of these pulmonary complications in COVID-19 patients has not been elucidated. Autopsy study of these patients showed SARS-CoV-2 virions in pulmonary vessels and sequestrated leukocytes infiltrates associated with endotheliopathy and microvascular thrombosis. Since SARS-CoV-2 enters and infects target cells by binding its spike (S) protein to cellular angiotensin-converting enzyme 2 (ACE2), and there is evidence that vascular endothelial cells and neutrophils express ACE2, we investigated the effect of S-proteins and cell-cell communication on primary human lung microvascular endothelial cells (HLMEC) and neutrophils expression of thrombogenic factors and the potential mechanisms. Using S-proteins of two different SARS-CoV-2 variants (Wuhan and Delta), we demonstrate that exposure of HLMEC or neutrophils to S-proteins, co-culture of HLMEC exposed to S-proteins with non-exposed neutrophils, or co-culture of neutrophils exposed to S-proteins with non-exposed HLMEC induced transcriptional upregulation of tissue factor (TF), significantly increased the expression and secretion of factor (F)-V, thrombin, and fibrinogen and inhibited tissue factor pathway inhibitor (TFPI), the primary regulator of the extrinsic pathway of blood coagulation, in both cell types. Recombinant (r)TFPI and a thiol blocker (5,5'-dithio-bis-(2-nitrobenzoic acid)) prevented S-protein-induced expression and secretion of Factor-V, thrombin, and fibrinogen. Thrombomodulin blocked S-protein-induced expression and secretion of fibrinogen but had no effect on S-protein-induced expression of Factor-V or thrombin. These results suggests that following SARS-CoV-2 contact with the pulmonary endothelium or neutrophils and endothelial-neutrophil interactions, viral S-proteins induce coagulopathy via the TF pathway and mechanisms involving functional thiol groups. These findings suggest that using rTFPI and/or thiol-based drugs could be a viable therapeutic strategy against SARS-CoV-2-induced coagulopathy and thrombosis.


Sujets)
Troubles de l'hémostase et de la coagulation , COVID-19 , Thrombose , Angiotensin-converting enzyme 2 , Communication cellulaire , Cellules endothéliales/métabolisme , Endothélium/métabolisme , Fibrinogène , Humains , Lipoprotéines , Poumon/métabolisme , Granulocytes neutrophiles/métabolisme , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus/métabolisme , Thiols , Thrombine , Thrombomoduline , Thromboplastine , Thrombose/étiologie
2.
Mol Med ; 28(1): 122, 2022 10 10.
Article Dans Anglais | MEDLINE | ID: covidwho-2064734

Résumé

BACKGROUND: Long-COVID is characterized by prolonged, diffuse symptoms months after acute COVID-19. Accurate diagnosis and targeted therapies for Long-COVID are lacking. We investigated vascular transformation biomarkers in Long-COVID patients. METHODS: A case-control study utilizing Long-COVID patients, one to six months (median 98.5 days) post-infection, with multiplex immunoassay measurement of sixteen blood biomarkers of vascular transformation, including ANG-1, P-SEL, MMP-1, VE-Cad, Syn-1, Endoglin, PECAM-1, VEGF-A, ICAM-1, VLA-4, E-SEL, thrombomodulin, VEGF-R2, VEGF-R3, VCAM-1 and VEGF-D. RESULTS: Fourteen vasculature transformation blood biomarkers were significantly elevated in Long-COVID outpatients, versus acutely ill COVID-19 inpatients and healthy controls subjects (P < 0.05). A unique two biomarker profile consisting of ANG-1/P-SEL was developed with machine learning, providing a classification accuracy for Long-COVID status of 96%. Individually, ANG-1 and P-SEL had excellent sensitivity and specificity for Long-COVID status (AUC = 1.00, P < 0.0001; validated in a secondary cohort). Specific to Long-COVID, ANG-1 levels were associated with female sex and a lack of disease interventions at follow-up (P < 0.05). CONCLUSIONS: Long-COVID patients suffer prolonged, diffuse symptoms and poorer health. Vascular transformation blood biomarkers were significantly elevated in Long-COVID, with angiogenesis markers (ANG-1/P-SEL) providing classification accuracy of 96%. Vascular transformation blood biomarkers hold potential for diagnostics, and modulators of angiogenesis may have therapeutic efficacy.


Sujets)
Marqueurs biologiques , COVID-19 , Marqueurs biologiques/sang , COVID-19/complications , Études cas-témoins , Endogline , Femelle , Humains , Intégrine alpha4bêta1 , Molécule-1 d'adhérence intercellulaire , Matrix metalloproteinase 1 , Néovascularisation pathologique , Antigènes CD31 , Thrombomoduline , Molécule-1 d'adhérence des cellules vasculaires , Facteur de croissance endothéliale vasculaire de type A , Facteur de croissance endothéliale vasculaire de type D ,
3.
Thromb Haemost ; 122(8): 1352-1360, 2022 Aug.
Article Dans Anglais | MEDLINE | ID: covidwho-1730359

Résumé

A large number of daily requests to exclude possible prothrombotic risk factors for coronavirus disease 2019 (COVID-19) vaccines were received. Our aim was to longitudinally evaluate coagulation profiles in a series of healthy subjects who received COVID-19 vaccination and assess hypercoagulability thereafter. Volunteers awaiting a first or second dose of either the ChAdOx1 or BNT162b2 vaccine were enrolled. Venous samples were obtained at baseline (before the vaccine) and longitudinally 3 ± 2 days (T1) and 10 ± 2 days after the vaccine (T2). Global coagulation monitoring was assessed via platelet count, whole blood thromboelastometry and impedance aggregometry, plasma thrombin generation, and anti-platelet factor 4 (PF4)/heparin immunoglobulin G antibodies. One hundred and twenty-two subjects were enrolled (61 [50%] ChAdOx1 and 61 BNT162b2). The ChAdOx1 cohort showed a slight but transient increase in thrombin generation (mainly endogenous thrombin potential [ETP] with thrombomodulin and ETP ratio) at T1, which promptly decreased at T2. In addition, the second dose of either vaccine was associated with increased thrombin peak, ETP with thrombomodulin, and ETP ratio. At baseline, 3.2% of the ChAdOx1 cohort and 1.6% BNT162b2 cohort were positive for PF4/heparin antibodies with a stable titer through T1 and T2. No relevant differences were detected in platelet count and aggregation, or thromboelastometry parameters. No thrombotic or hemorrhagic events occurred. We can confirm that no clinically meaningful hypercoagulability occurred after either vaccine, albeit keeping in mind that thrombin generation may increase in the first days after the second dose of either vaccine and after the first dose of the ChAdOx1 vaccine.


Sujets)
Vaccins contre la COVID-19 , COVID-19 , Thrombophilie , Vaccin BNT162 , COVID-19/prévention et contrôle , Vaccins contre la COVID-19/effets indésirables , Héparine/effets indésirables , Humains , Études longitudinales , Thrombine , Thrombomoduline , Thrombophilie/diagnostic , Thrombophilie/étiologie , Vaccination
4.
Am J Respir Crit Care Med ; 205(3): 324-329, 2022 02 01.
Article Dans Anglais | MEDLINE | ID: covidwho-1673593

Résumé

Rationale: The mortality in patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) who require mechanical ventilation remains high, and endotheliopathy has been implicated. Objectives: To determine the effect of prostacyclin infusion in mechanically ventilated patients infected with SARS-CoV-2 with severe endotheliopathy. Methods: We conducted a multicenter, randomized clinical trial in adults infected with coronavirus disease (COVID-19) who required mechanical ventilation and had a plasma level of thrombomodulin >4 ng/ml; patients were randomized to 72-hour infusion of prostacyclin 1 ng/kg/min or placebo. Measurements and Main Results: The main outcome was the number of days alive and without mechanical ventilation within 28 days. Key secondary outcomes were 28-day mortality and serious adverse events within 7 days. Eighty patients were randomized (41 prostacyclin and 39 placebo). The median number of days alive without mechanical ventilation at 28 days was 16.0 days (SD, 12) versus 5.0 days (SD, 10) (difference of the medians, 10.96 days; 95% confidence interval [CI], -5 to 21; P = 0.07) in the prostacyclin and the placebo groups, respectively. The 28-day mortality was 21.9% versus 43.6% in the prostacyclin and the placebo groups, respectively (risk ratio, 0.50; 95% CI, 0.24 to 0.96; P = 0.06). The incidence of serious adverse events within 7 days was 2.4% versus 12.8% (risk ratio, 0.19; 95% CI, 0.001 to 1.11; P = 0.10) in the prostacyclin and the placebo groups, respectively. Conclusions: Prostacyclin was not associated with a significant reduction in the number of days alive and without mechanical ventilation within 28 days. The point estimates, however, favored the prostacyclin group in all analyses, including 28-day mortality, warranting further investigation in larger trials. Clinical trial registered with www.clinicaltrials.gov (NCT04420741); EudraCT Identifier: 2020-001296-33.


Sujets)
, COVID-19/thérapie , Endothélium vasculaire/anatomopathologie , Prostacycline/administration et posologie , Antiagrégants plaquettaires/administration et posologie , Ventilation artificielle , Sujet âgé , COVID-19/sang , COVID-19/complications , Danemark , Femelle , Humains , Perfusions veineuses , Intubation trachéale , Mâle , Adulte d'âge moyen , Taux de survie , Thrombomoduline/sang , Résultat thérapeutique
5.
J Cell Mol Med ; 26(5): 1445-1455, 2022 03.
Article Dans Anglais | MEDLINE | ID: covidwho-1642687

Résumé

There is an unmet need of models for early prediction of morbidity and mortality of Coronavirus disease-19 (COVID-19). We aimed to a) identify complement-related genetic variants associated with the clinical outcomes of ICU hospitalization and death, b) develop an artificial neural network (ANN) predicting these outcomes and c) validate whether complement-related variants are associated with an impaired complement phenotype. We prospectively recruited consecutive adult patients of Caucasian origin, hospitalized due to COVID-19. Through targeted next-generation sequencing, we identified variants in complement factor H/CFH, CFB, CFH-related, CFD, CD55, C3, C5, CFI, CD46, thrombomodulin/THBD, and A Disintegrin and Metalloproteinase with Thrombospondin motifs (ADAMTS13). Among 381 variants in 133 patients, we identified 5 critical variants associated with severe COVID-19: rs2547438 (C3), rs2250656 (C3), rs1042580 (THBD), rs800292 (CFH) and rs414628 (CFHR1). Using age, gender and presence or absence of each variant, we developed an ANN predicting morbidity and mortality in 89.47% of the examined population. Furthermore, THBD and C3a levels were significantly increased in severe COVID-19 patients and those harbouring relevant variants. Thus, we reveal for the first time an ANN accurately predicting ICU hospitalization and death in COVID-19 patients, based on genetic variants in complement genes, age and gender. Importantly, we confirm that genetic dysregulation is associated with impaired complement phenotype.


Sujets)
COVID-19/génétique , COVID-19/mortalité , , COVID-19/épidémiologie , Activation du complément/génétique , Facteur H du complément/génétique , Protéines du système du complément/génétique , Femelle , Grèce/épidémiologie , Hospitalisation/statistiques et données numériques , Humains , Unités de soins intensifs/statistiques et données numériques , Mâle , Adulte d'âge moyen , Modèles génétiques , Morbidité , Polymorphisme de nucléotide simple , Thrombomoduline/génétique
6.
EBioMedicine ; 75: 103812, 2022 Jan.
Article Dans Anglais | MEDLINE | ID: covidwho-1639102

Résumé

BACKGROUND: Thromboembolism is a life-threatening manifestation of coronavirus disease 2019 (COVID-19). We investigated a dysfunctional phenotype of vascular endothelial cells in the lungs during COVID-19. METHODS: We obtained the lung specimens from the patients who died of COVID-19. The phenotype of endothelial cells and immune cells was examined by flow cytometry and immunohistochemistry (IHC) analysis. We tested the presence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in the endothelium using IHC and electron microscopy. FINDINGS: The autopsy lungs of COVID-19 patients exhibited severe coagulation abnormalities, immune cell infiltration, and platelet activation. Pulmonary endothelial cells of COVID-19 patients showed increased expression of procoagulant von Willebrand factor (VWF) and decreased expression of anticoagulants thrombomodulin and endothelial protein C receptor (EPCR). In the autopsy lungs of COVID-19 patients, the number of macrophages, monocytes, and T cells was increased, showing an activated phenotype. Despite increased immune cells, adhesion molecules such as ICAM-1, VCAM-1, E-selectin, and P-selectin were downregulated in pulmonary endothelial cells of COVID-19 patients. Notably, decreased thrombomodulin expression in endothelial cells was associated with increased immune cell infiltration in the COVID-19 patient lungs. There were no SARS-CoV-2 particles detected in the lung endothelium of COVID-19 patients despite their dysfunctional phenotype. Meanwhile, the autopsy lungs of COVID-19 patients showed SARS-CoV-2 virions in damaged alveolar epithelium and evidence of hypoxic injury. INTERPRETATION: Pulmonary endothelial cells become dysfunctional during COVID-19, showing a loss of thrombomodulin expression related to severe thrombosis and infiltration, and endothelial cell dysfunction might be caused by a pathologic condition in COVID-19 patient lungs rather than a direct infection with SARS-CoV-2. FUNDING: This work was supported by the Johns Hopkins University, the American Heart Association, and the National Institutes of Health.


Sujets)
Troubles de l'hémostase et de la coagulation/métabolisme , COVID-19/métabolisme , Régulation négative , Endothélium vasculaire/métabolisme , Hypoxie/métabolisme , Poumon/métabolisme , SARS-CoV-2/métabolisme , Thrombomoduline/biosynthèse , Sujet âgé , Sujet âgé de 80 ans ou plus , Troubles de l'hémostase et de la coagulation/anatomopathologie , COVID-19/anatomopathologie , Cellules endothéliales/métabolisme , Cellules endothéliales/ultrastructure , Endothélium vasculaire/ultrastructure , Femelle , Humains , Hypoxie/anatomopathologie , Poumon/ultrastructure , Mâle , Adulte d'âge moyen
7.
Shock ; 57(1): 95-105, 2022 01 01.
Article Dans Anglais | MEDLINE | ID: covidwho-1574295

Résumé

BACKGROUND: Endotheliopathy is a key element in COVID-19 pathophysiology, contributing to both morbidity and mortality. Biomarkers distinguishing different COVID-19 phenotypes from sepsis syndrome remain poorly understood. OBJECTIVE: To characterize circulating biomarkers of endothelial damage in different COVID-19 clinical disease stages compared with sepsis syndrome and normal volunteers. METHODS: Patients with COVID-19 pneumonia (n = 49) were classified into moderate, severe, or critical (life-threatening) disease. Plasma samples were collected within 48 to 72 h of hospitalization to analyze endothelial activation markers, including soluble Vascular Cell Adhesion Molecule-1 (sVCAM-1), von Willebrand Factor (VWF), A disintegrin-like and metalloprotease with thrombospondin type 1 motif no. 13 (ADAMTS-13) activity, thrombomodulin (TM), and soluble TNF receptor I (sTNFRI); heparan sulfate (HS) for endothelial glycocalyx degradation; C5b9 deposits on endothelial cells in culture and soluble C5b9 for complement activation; circulating dsDNA for neutrophil extracellular traps (NETs) presence, and α2-antiplasmin and PAI-1 as parameters of fibrinolysis. We compared the level of each biomarker in all three COVID-19 groups and healthy donors as controls (n = 45). Results in critically ill COVID-19 patients were compared with other intensive care unit (ICU) patients with septic shock (SS, n = 14), sepsis (S, n = 7), and noninfectious systemic inflammatory response syndrome (NI-SIRS, n = 7). RESULTS: All analyzed biomarkers were increased in COVID-19 patients versus controls (P < 0.001), except for ADAMTS-13 activity that was normal in both groups. The increased expression of sVCAM-1, VWF, sTNFRI, and HS was related to COVID-19 disease severity (P < 0.05). Several differences in these parameters were found between ICU groups: SS patients showed significantly higher levels of VWF, TM, sTNFRI, and NETS compared with critical COVID-19 patients and ADAMTS-13 activity was significantly lover in SS, S, and NI-SIRS versus critical COVID-19 (P < 0.001). Furthermore, α2-antiplasmin activity was higher in critical COVID-19 versus NI-SIRS (P < 0.01) and SS (P < 0.001), whereas PAI-1 levels were significantly lower in COVID-19 patients compared with NI-SIRS, S, and SS patients (P < 0.01). CONCLUSIONS: COVID-19 patients present with increased circulating endothelial stress products, complement activation, and fibrinolytic dysregulation, associated with disease severity. COVID-19 endotheliopathy differs from SS, in which endothelial damage is also a critical feature of pathobiology. These biomarkers could help to stratify the severity of COVID-19 disease and may also provide information to guide specific therapeutic strategies to mitigate endotheliopathy progression.


Sujets)
COVID-19/sang , Protéine ADAMTS13/sang , Sujet âgé , Marqueurs biologiques/sang , Complexe d'attaque membranaire du complément/analyse , ADN/sang , Femelle , Héparitine sulfate/sang , Humains , Mâle , Adulte d'âge moyen , Acuité des besoins du patient , Inhibiteur-1 d'activateur du plasminogène/sang , Études prospectives , Récepteur au facteur de nécrose tumorale de type I/sang , Sepsie/sang , Thrombomoduline/sang , Molécule-1 d'adhérence des cellules vasculaires/sang , alpha-2-Antiplasmine/analyse , Facteur de von Willebrand/analyse
8.
Mol Med ; 27(1): 151, 2021 12 03.
Article Dans Anglais | MEDLINE | ID: covidwho-1551198

Résumé

BACKGROUND: We investigated the feasibility of two biomarkers of endothelial damage (Syndecan-1 and thrombomodulin) in coronavirus disease 2019 (COVID-19), and their association with inflammation, coagulopathy, and mortality. METHODS: The records of 49 COVID-19 patients who were admitted to an intensive care unit (ICU) in Wuhan, China between February and April 2020 were examined. Demographic, clinical, and laboratory data, and outcomes were compared between survivors and non-survivors COVID-19 patients, and between patients with high and low serum Syndecan-1 levels. The dynamics of serum Syndecan-1 levels were also analyzed. RESULTS: The levels of Syndecan-1 were significantly higher in non-survivor group compared with survivor group (median 1031.4 versus 504.0 ng/mL, P = 0.002), and the levels of thrombomodulin were not significantly different between these two groups (median 4534.0 versus 3780.0 ng/mL, P = 0.070). Kaplan-Meier survival analysis showed that the group with high Syndecan-1 levels had worse overall survival (log-rank test: P = 0.023). Patients with high Syndecan-1 levels also had significantly higher levels of thrombomodulin, interleukin-6, and tumor necrosis factor-α. Data on the dynamics of Syndecan-1 levels indicated much greater variations in non-survivors than survivors. CONCLUSIONS: COVID-19 patients with high levels of Syndecan-1 develop more serious endothelial damage and inflammatory reactions, and have increased mortality. Syndecan-1 has potential for use as a marker for progression or severity of COVID-19. Protecting the glycocalyx from destruction is a potential treatment for COVID-19.


Sujets)
COVID-19/sang , COVID-19/thérapie , Endothélium/métabolisme , Glycocalyx/métabolisme , Syndécane-1/sang , Sujet âgé , Marqueurs biologiques/sang , Coagulation sanguine , COVID-19/mortalité , Chine/épidémiologie , Cytokines/métabolisme , Endothélium vasculaire/anatomopathologie , Femelle , Humains , Inflammation , Unités de soins intensifs , Interleukine-6/sang , Estimation de Kaplan-Meier , Mâle , Adulte d'âge moyen , Oxygène , Courbe ROC , SARS-CoV-2 , Thrombomoduline/sang , Résultat thérapeutique , Facteur de nécrose tumorale alpha/sang
9.
Curr Med Chem ; 29(21): 3790-3805, 2022.
Article Dans Anglais | MEDLINE | ID: covidwho-1496773

Résumé

BACKGROUND: Several studies have revealed the link between Coronavirus Disease 2019 (COVID-19) and endothelial dysfunction. To better understand the global pattern of this relationship, we conducted a meta-analysis on endothelial biomarkers related to COVID-19 severity. METHODS: We systematically searched the literature up to March 10, 2021, for studies investigating the association between COVID-19 severity and the following endothelial biomarkers: Intercellular Adhesion Molecule 1 (ICAM-1), Vascular Cell Adhesion Molecule 1 (VCAM-1), E-selectin, P-selectin, Von Willebrand Factor Antigen (VWFAg), soluble Thrombomodulin (sTM), Mid-regional pro-adrenomedullin (MR-proADM), and Angiopoietin-2 (Ang-2). Pooled estimates and mean differences (PMD) for each biomarker were reported. RESULTS: A total of 27 studies (n=2213 patients) were included. Critically ill patients presented with higher levels of MR-proADM (PMD: 0.71 nmol/L, 95% CI: 0.22 to 1.20 nmol/L, p=0.02), E-selectin (PMD: 13,32 pg/ml, 95% CI: 4,89 to 21,75 pg/ml, p=0.008), VCAM-1 (PMD: 479 ng/ml, 95% CI: 64 to 896 ng/ml, p=0.03), VWF-Ag (PMD: 110.5 IU/dl, 95% CI: 44.8 to 176.1 IU/dl, p=0.04) and Ang-2 (PMD: 2388 pg/ml, 95% CI: 1121 to 3655 pg/ml, p=0.003), as compared to non-critically ill ones. ICAM-1, P-selectin and thrombomodulin did not differ between the two groups (p>0.05). CONCLUSION: Endothelial biomarkers display significant heterogeneity in COVID-19 patients, with higher MR-proADM, E-selectin, VCAM-1, VWF-Ag, and Ang-2 levels being associated with increased severity. These findings strengthen the evidence on the key role of endothelial dysfunction in disease progress.


Sujets)
COVID-19 , Maladies vasculaires , Marqueurs biologiques/métabolisme , COVID-19/diagnostic , Sélectine E/métabolisme , Endothélium vasculaire/métabolisme , Humains , Molécule-1 d'adhérence intercellulaire/métabolisme , Thrombomoduline/métabolisme , Molécule-1 d'adhérence des cellules vasculaires/métabolisme , Maladies vasculaires/métabolisme , Facteur de von Willebrand/analyse , Facteur de von Willebrand/métabolisme
10.
Microvasc Res ; 138: 104224, 2021 11.
Article Dans Anglais | MEDLINE | ID: covidwho-1309345

Résumé

BACKGROUND: Several studies have reported that the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can directly infect endothelial cells, and endothelial dysfunction is often found in severe cases of coronavirus disease 2019 (COVID-19). To better understand the prognostic values of endothelial dysfunction in COVID-19-associated coagulopathy, we conducted a systematic review and meta-analysis to assess biomarkers of endothelial cells in patients with COVID-19. METHODS: A literature search was conducted on online databases for observational studies evaluating biomarkers of endothelial dysfunction and composite poor outcomes in COVID-19 patients. RESULTS: A total of 1187 patients from 17 studies were included in this analysis. The estimated pooled means for von Willebrand Factor (VWF) antigen levels in COVID-19 patients was higher compared to healthy control (306.42 [95% confidence interval (CI) 291.37-321.48], p < 0.001; I2:86%), with the highest VWF antigen levels was found in deceased COVID-19 patients (448.57 [95% CI 407.20-489.93], p < 0.001; I2:0%). Meta-analysis showed that higher plasma levels of VWF antigen, tissue-type plasminogen activator (t-PA), plasminogen activator inhibitor-1 antigen (PAI-1) antigen, and soluble thrombomodulin (sTM) were associated with composite poor outcome in COVID-19 patients ([standardized mean difference (SMD) 0.74 [0.33-1.16], p < 0.001; I2:80.4%], [SMD 0.55 [0.19-0.92], p = 0.003; I2:6.4%], [SMD 0.33 [0.04-0.62], p = 0.025; I2:7.9%], and [SMD 0.55 [0.10-0.99], p = 0.015; I2:23.6%], respectively). CONCLUSION: The estimated pooled means show increased levels of VWF antigen in COVID-19 patients. Several biomarkers of endothelial dysfunction, including VFW antigen, t-PA, PAI-1, and sTM, are significantly associated with increased composite poor outcomes in patients with COVID-19. PROSPERO REGISTRATION NUMBER: CRD42021228821.


Sujets)
COVID-19/sang , Endothélium vasculaire/métabolisme , Inhibiteur-1 d'activateur du plasminogène/sang , Thrombomoduline/sang , Activateur tissulaire du plasminogène/sang , Facteur de von Willebrand/analyse , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Marqueurs biologiques/sang , COVID-19/diagnostic , COVID-19/physiopathologie , COVID-19/thérapie , Endothélium vasculaire/physiopathologie , Femelle , Humains , Mâle , Adulte d'âge moyen , Valeur prédictive des tests , Pronostic
11.
Clin Immunol ; 226: 108726, 2021 05.
Article Dans Anglais | MEDLINE | ID: covidwho-1179332

Résumé

Recent studies suggest excessive complement activation in severe coronavirus disease-19 (COVID-19). The latter shares common characteristics with complement-mediated thrombotic microangiopathy (TMA). We hypothesized that genetic susceptibility would be evident in patients with severe COVID-19 (similar to TMA) and associated with disease severity. We analyzed genetic and clinical data from 97 patients hospitalized for COVID-19. Through targeted next-generation-sequencing we found an ADAMTS13 variant in 49 patients, along with two risk factor variants (C3, 21 patients; CFH,34 patients). 31 (32%) patients had a combination of these, which was independently associated with ICU hospitalization (p = 0.022). Analysis of almost infinite variant combinations showed that patients with rs1042580 in thrombomodulin and without rs800292 in complement factor H did not require ICU hospitalization. We also observed gender differences in ADAMTS13 and complement-related variants. In light of encouraging results by complement inhibitors, our study highlights a patient population that might benefit from early initiation of specific treatment.


Sujets)
Protéine ADAMTS13/génétique , COVID-19/génétique , Complément C3/génétique , Prédisposition génétique à une maladie/génétique , Thrombomoduline/génétique , Sujet âgé , Algorithmes , COVID-19/physiopathologie , Activation du complément , Facteur H du complément/génétique , Soins de réanimation , Femelle , Dépistage génétique , Séquençage nucléotidique à haut débit , Hospitalisation/statistiques et données numériques , Humains , Unités de soins intensifs , Mâle , Adulte d'âge moyen , Facteurs de risque , Indice de gravité de la maladie , Microangiopathies thrombotiques/génétique
12.
Elife ; 102021 03 08.
Article Dans Anglais | MEDLINE | ID: covidwho-1122117

Résumé

Extensive fibrin deposition in the lungs and altered levels of circulating blood coagulation proteins in COVID-19 patients imply local derangement of pathways that limit fibrin formation and/or promote its clearance. We examined transcriptional profiles of bronchoalveolar lavage fluid (BALF) samples to identify molecular mechanisms underlying these coagulopathies. mRNA levels for regulators of the kallikrein-kinin (C1-inhibitor), coagulation (thrombomodulin, endothelial protein C receptor), and fibrinolytic (urokinase and urokinase receptor) pathways were significantly reduced in COVID-19 patients. While transcripts for several coagulation proteins were increased, those encoding tissue factor, the protein that initiates coagulation and whose expression is frequently increased in inflammatory disorders, were not increased in BALF from COVID-19 patients. Our analysis implicates enhanced propagation of coagulation and decreased fibrinolysis as drivers of the coagulopathy in the lungs of COVID-19 patients.


Sujets)
Coagulation sanguine/génétique , COVID-19/anatomopathologie , Fibrine/génétique , Poumon/anatomopathologie , SARS-CoV-2 , Anticoagulants/métabolisme , Liquide de lavage bronchoalvéolaire , COVID-19/génétique , COVID-19/métabolisme , Récepteur endothélial de la protéine C/génétique , Récepteur endothélial de la protéine C/métabolisme , Fibrine/métabolisme , Expression des gènes , Humains , Système kallicréine-kinine/génétique , Kallicréines/génétique , Kallicréines/métabolisme , Kinines/génétique , Kinines/métabolisme , Poumon/métabolisme , ARN messager/métabolisme , Analyse de séquence d'ARN , Thrombomoduline/génétique , Thrombomoduline/métabolisme , Activateur du plasminogène de type urokinase/génétique , Activateur du plasminogène de type urokinase/métabolisme
13.
Clin Chem Lab Med ; 59(7): 1323-1330, 2021 06 25.
Article Dans Anglais | MEDLINE | ID: covidwho-1067443

Résumé

OBJECTIVES: Thrombin generation (TG) with and without thrombomodulin (TM) was evaluated in COVID-19 patients with different disease severity and thromboprophylaxis regimen, in order to understand the prothrombotic profile. METHODS: We enrolled consecutive patients with confirmed diagnosis of COVID-19 admitted to Medical Departments (MD) or Intensive Care Units (ICU), and 54 healthy controls. RESULTS: Eighty-nine patients were included (mean age 60.4±16.1 years, 68.5% male); 33.7% admitted to ICU. Twenty-four patients (26.9%) were enrolled before thromboprophylaxis administration; 45 patients (50.6%) received standard and 20 (22.5%) intermediate sub-therapeutic dose thromboprophylaxis. Overall, patients with COVID-19 showed a TG profile comparable to that of healthy subjects (i.e. comparable peak height, endogenous thrombin potential [ETP] with and without TM). The only exception was lag time and time to peak, prolonged in COVID-19 patients vs. controls. MD patients showed a similar TG profile to healthy controls, and ICU patients showed significantly decrease ETP (p=0.030) compared to MD. As for thromboprophylaxis, TG profile was significantly increased in COVID-19 patients without thromboprophylaxis vs. controls and vs. those with thromboprophylaxis. In this latter group, ETP inhibition was significantly decreased (p=0.0003) and positively correlated with anti-Xa activity (r=0.49, p=0.0017). However, patients with thromboprophylaxis had similar TG profile vs. controls. Intermediate dose thromboprophylaxis more effectively inhibited TG in severe COVID-19 patients by increasing ETP inhibition via ETP with TM reduction vs. standard dose. CONCLUSIONS: COVID-19 patients showed increased TG at diagnosis. Standard thromboprophylaxis reduced TG to levels of healthy controls. Intermediate sub-therapeutic thromboprophylaxis more effectively inhibited TG by decreasing ETP with TM.


Sujets)
Anticoagulants/usage thérapeutique , COVID-19/sang , Thrombine/analyse , Thrombose/prévention et contrôle , Adulte , Sujet âgé , COVID-19/complications , Femelle , Fondaparinux/usage thérapeutique , Héparine bas poids moléculaire/usage thérapeutique , Humains , Mâle , Adulte d'âge moyen , Études prospectives , SARS-CoV-2 , Thrombomoduline/analyse , Thrombose/sang , Thrombose/étiologie
15.
Trials ; 21(1): 746, 2020 Aug 26.
Article Dans Anglais | MEDLINE | ID: covidwho-731235

Résumé

OBJECTIVES: To investigate the effect of continuous infusion of the potential endothelial cytoprotective agent prostacyclin (Iloprost) 1 ng/kg/min vs. placebo for 72 hours on pulmonary endotheliopathy in mechanically ventilated COVID-19 patients. TRIAL DESIGN: A multicenter, randomized (1:1, active: placebo), blinded, parallel group exploratory trial PARTICIPANTS: Inclusion criteria are: Adult patients (>18 years); Confirmed COVID-19 infection; Need for mechanical interventions; Endothelial biomarker soluble thrombomodulin >4ng/ml. EXCLUSION CRITERIA: Withdrawal from active therapy; Pregnancy (non-pregnancy confirmed by patient being postmenopausal (age 60 or above) or having a negative urine- or plasma-hCG); Known hypersensitivity to iloprost or to any of the other ingredients; Previously included in this trial or a prostacyclin trial within 30 days; Consent cannot be obtained; Life-threatening bleeding defined by the treating physician; Known severe heart failure (NYHA class IV); Suspected acute coronary syndrome The study is conducted at five intensive care units in the Capital Region of Denmark at Rigshospitalet, Herlev Hospital, Hvidovre Hospital, Bispebjerg Hospital, Nordsjællands Hospital. INTERVENTION AND COMPARATOR: The patients are randomized to 72-hours continuous infusion of either prostacyclin (Iloprost/Ilomedin) at a dose of 1 ng/kg/min or Placebo (normal saline). MAIN OUTCOMES: Primary endpoint: Days alive without mechanical ventilation in the intensive care units within 28 days RANDOMISATION: The randomisation sequence is performed in permuted blocks of variable sizes stratified for trial site using centralised, concealed allocation. The randomisation sequence is generated 1:1 (active/placebo) using the online randomisation software 'Sealed Envelope' ( https://www.sealedenvelope.com/ ). Once generated the randomisation sequence is formatted and uploaded into Research Electronic Data Capture system (REDCap) to facilitate centralised, web-based allocation according to local written instruction. BLINDING (MASKING): The following are blinded: all clinicians, patients, investigators, and those assessing the outcomes including the statisticians. NUMBERS TO BE RANDOMISED (SAMPLE SIZE): Forty patients are planned to be randomized to each group, with a total sample size of 80 patients. TRIAL STATUS: Protocol version 1.4 dated May 25, 2020. Recruitment is ongoing. The recruitment was started June 15, 2020 and the anticipated finish of recruitment is February 28, 2021 with 90 days follow up hereafter. TRIAL REGISTRATION: Trial registration at clinicaltrialregisters.eu; EudraCT no. 2020-001296-33 on 3 April 2020 and at ClinicalTrials.gov Identifier: NCT04420741 on 9 June 2020 FULL PROTOCOL: The full protocol is attached as an additional file, accessible from the Trials website (Additional file 1).In the interest in expediting dissemination of this material, the familiar formatting has been eliminated; this Letter serves as a summary of the key elements of the full protocol.


Sujets)
Infections à coronavirus/thérapie , Iloprost/usage thérapeutique , Pneumopathie virale/thérapie , Ventilation artificielle , Vasodilatateurs/usage thérapeutique , Betacoronavirus , COVID-19 , Infections à coronavirus/sang , Infections à coronavirus/traitement médicamenteux , Danemark , Endothélium vasculaire/métabolisme , Endothélium vasculaire/physiopathologie , Humains , Perfusions veineuses , Unités de soins intensifs , Pandémies , Pneumopathie virale/sang , SARS-CoV-2 , Thrombomoduline/métabolisme ,
SÉLECTION CITATIONS
Détails de la recherche