1
|
Pastura P, McDaniel CG, Alharbi S, Fox D, Coleman B, Malik P, Adams DM, Le Cras TD. NRAS Q61R mutation drives elevated angiopoietin-2 expression in human endothelial cells and a genetic mouse model. Pediatr Blood Cancer 2024; 71:e31032. [PMID: 38711167 PMCID: PMC11116044 DOI: 10.1002/pbc.31032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 03/10/2024] [Accepted: 04/10/2024] [Indexed: 05/08/2024]
Abstract
BACKGROUND Angiopoietin-2 (Ang-2) is increased in the blood of patients with kaposiform lymphangiomatosis (KLA) and kaposiform hemangioendothelioma (KHE). While the genetic causes of KHE are not clear, a somatic activating NRASQ61R mutation has been found in the lesions of KLA patients. PROCEDURE Our study tested the hypothesis that the NRASQ61R mutation drives elevated Ang-2 expression in endothelial cells. Ang-2 was measured in human endothelial progenitor cells (EPC) expressing NRASQ61R and a genetic mouse model with endothelial targeted NRASQ61R. To determine the signaling pathways driving Ang-2, NRASQ61R EPC were treated with signaling pathway inhibitors. RESULTS Ang-2 levels were increased in EPC expressing NRASQ61R compared to NRASWT by Western blot analysis of cell lysates and ELISA of the cell culture media. Ang-2 levels were elevated in the blood of NRASQ61R mutant mice. NRASQ61R mutant mice also had reduced platelet counts and splenomegaly with hypervascular lesions, like some KLA patients. mTOR inhibitor rapamycin attenuated Ang-2 expression by NRASQ61R EPC. However, MEK1/2 inhibitor trametinib was more effective blocking increases in Ang-2. CONCLUSIONS Our studies show that the NRASQ61R mutation in endothelial cells induces Ang-2 expression in vitro and in vivo. In cultured human endothelial cells, NRASQ61R drives elevated Ang-2 through MAP kinase and mTOR-dependent signaling pathways.
Collapse
Affiliation(s)
- Patricia Pastura
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - C. Griffin McDaniel
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Sara Alharbi
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Dermot Fox
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Bethany Coleman
- Department of Molecular & Cellular Biosciences, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Punam Malik
- Division of Experimental Hematology and Cancer Biology, Cancer and Blood Disease Institute,
Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Denise M. Adams
- Division of Oncology, Comprehensive Vascular Anomalies Program, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Timothy D. Le Cras
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
2
|
Tsiatsiou P, Kouirouxis K, Tsaireli V, Lanta A, Kassomenaki A, Papaioannou M, Protonotariou E, Skoura L. Angiopoietins as Predictor Indexes in COVID-19 Patients in Delta and Omicron Waves. Curr Issues Mol Biol 2024; 46:3975-3989. [PMID: 38785513 PMCID: PMC11120536 DOI: 10.3390/cimb46050245] [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: 02/18/2024] [Revised: 04/07/2024] [Accepted: 04/19/2024] [Indexed: 05/25/2024] Open
Abstract
This study aimed to explore the correlation between Angiopoietin-1 (Ang-1) and Angiopoietin-2 (Ang-2) concentrations and the Angiopoietin-2/Angiopoietin-1 ratio (Ang-2/Ang-1) with clinical outcomes, potentially serving as disease severity and survival biomarkers. A study at AHEPA University Hospital involved 90 Coronavirus Disease 2019 (COVID-19) adult patients, 30 hospitalized intensive care units (ICU), 30 inward units (non-ICU), and 30 asymptomatic non-hospitalized individuals as controls. Estimated endothelial dysfunction markers related to angiogenesis were measured. There was a statistically significant difference only between outpatient and hospitalized patients (non-ICU-ICU groups) for the Ang-1 and Ang-2 indices. The Ang-2/Ang-1 ratio has differed significantly among the individual patient groups. An ROC analysis was conducted to find an optimal threshold for distinguishing between (outpatients-non-ICU) and (non-ICU-ICU) groups. It was based on Youden's index of 0.1122 and 0.3825, respectively. The Ang-1, Ang-2 levels, and Ang-2/Ang-1 ratio were analyzed as severity indicators in COVID-19 patients. The Ang-2/Ang-1 ratio demonstrated better prognostic and diagnostic utility than individual biomarker levels. Monitoring the Ang-2/Ang-1 ratio can identify COVID-19 patients at risk and assist clinicians in tailoring treatment strategies to improve outcomes.
Collapse
Affiliation(s)
- Panagiota Tsiatsiou
- Department of Microbiology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.K.); (V.T.); (A.L.); (A.K.); (E.P.); (L.S.)
| | - Kyriakos Kouirouxis
- Department of Microbiology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.K.); (V.T.); (A.L.); (A.K.); (E.P.); (L.S.)
| | - Vasiliki Tsaireli
- Department of Microbiology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.K.); (V.T.); (A.L.); (A.K.); (E.P.); (L.S.)
| | - Antonia Lanta
- Department of Microbiology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.K.); (V.T.); (A.L.); (A.K.); (E.P.); (L.S.)
| | - Angeliki Kassomenaki
- Department of Microbiology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.K.); (V.T.); (A.L.); (A.K.); (E.P.); (L.S.)
| | - Maria Papaioannou
- Division of Hematology, First Department of Internal Medicine, AHEPA General Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
| | - Efthymia Protonotariou
- Department of Microbiology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.K.); (V.T.); (A.L.); (A.K.); (E.P.); (L.S.)
| | - Lemonia Skoura
- Department of Microbiology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.K.); (V.T.); (A.L.); (A.K.); (E.P.); (L.S.)
| |
Collapse
|
3
|
Gouzi F, Philippe A, Pastre J, Renaud B, Gendron N, Subileau M, Hua-Huy T, Planquette B, Sanchez O, Smadja DM, Günther S. Recovery of Endothelium-dependent vascular relaxation impairment in convalescent COVID-19 patients: Insight from a pilot study. Respir Med Res 2023; 84:101044. [PMID: 37625374 DOI: 10.1016/j.resmer.2023.101044] [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: 04/12/2023] [Revised: 06/16/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023]
Abstract
BACKGROUND Endothelial dysfunction is a key-feature in acute COVID-19. However, follow-up data regarding endothelial dysfunction and injury after COVID-19 infection are lacking. We aimed to investigate the changes in endothelium-dependent vasorelaxation at baseline and four months after hospital discharge in COVID-19 patients. METHODS Twenty COVID-19 patients were compared to 24 healthy controls. Clinical and morphological data were collected after hospital admission for SARS-CoV-2 infection and reactive hyperaemia index (RHI) measurement was performed with a delay between 24 and 48 h after hospital admission and four months after hospital discharge in the outpatient clinics. Blood tests including inflammatory markers and measurement of post-occlusive vasorelaxation by digital peripheral arterial tonometry were performed at both visits. RESULTS At baseline, COVID-19 patients exhibited reduced RHI compared to controls (p < 0.001), in line with an endothelial dysfunction. At four months follow-up, there was a 51% increase in the RHI (1.69 ± 0.32 to 2.51 ± 0.91; p < 0.01) in favor of endothelium-dependent vascular relaxation recovery. RHI changes were positively correlated with baseline C-reactive protein (r = 0.68; p = 0.02). Compared to COVID-19 patients with a decrease in RHI, COVID-19 patients with an increase in RHI beyond the day-to-day variability (i.e. >11%) had less severe systemic inflammation at baseline. CONCLUSION Convalescent COVID-19 patients showed a recovery of systemic artery endothelial dysfunction, in particular patients with lower inflammation at baseline. Further studies are needed to decipher the interplay between inflammation and endothelial dysfunction in COVID-19 patients.
Collapse
Affiliation(s)
- Fares Gouzi
- PhyMedExp, INSERM - CNRS - Montpellier University, CHRU Montpellier, Montpellier, France; Université Paris Cité, Innovative Therapies in Haemostasis, INSERM UMR_S1140, Paris Cité, F-75006 Paris, France
| | - Aurélien Philippe
- Université Paris Cité, Innovative Therapies in Haemostasis, INSERM UMR_S1140, Paris Cité, F-75006 Paris, France; Hematology Department, AP-HP, Georges Pompidou European Hospital, F-75015 Paris, France
| | - Jean Pastre
- Department of Respiratory Medicine, AP-HP, Georges Pompidou European Hospital, F-75015 Paris, France
| | - Bertrand Renaud
- Unité d'Explorations Fonctionnelles Respiratoires et du Sommeil, AP-HP, Georges Pompidou European Hospital, F-75015 Paris, France; Université Paris Cité, UFR de médecine, F-75006 Paris, France
| | - Nicolas Gendron
- Université Paris Cité, Innovative Therapies in Haemostasis, INSERM UMR_S1140, Paris Cité, F-75006 Paris, France; Hematology Department, AP-HP, Georges Pompidou European Hospital, F-75015 Paris, France
| | - Marielle Subileau
- Unité d'Explorations Fonctionnelles Respiratoires et du Sommeil, AP-HP, Georges Pompidou European Hospital, F-75015 Paris, France
| | - Thông Hua-Huy
- Unité d'Explorations Fonctionnelles Respiratoires et du Sommeil, AP-HP, Georges Pompidou European Hospital, F-75015 Paris, France
| | - Benjamin Planquette
- Université Paris Cité, Innovative Therapies in Haemostasis, INSERM UMR_S1140, Paris Cité, F-75006 Paris, France; Unité d'Explorations Fonctionnelles Respiratoires et du Sommeil, AP-HP, Georges Pompidou European Hospital, F-75015 Paris, France
| | - Olivier Sanchez
- Université Paris Cité, Innovative Therapies in Haemostasis, INSERM UMR_S1140, Paris Cité, F-75006 Paris, France; Unité d'Explorations Fonctionnelles Respiratoires et du Sommeil, AP-HP, Georges Pompidou European Hospital, F-75015 Paris, France
| | - David M Smadja
- Université Paris Cité, Innovative Therapies in Haemostasis, INSERM UMR_S1140, Paris Cité, F-75006 Paris, France; Hematology Department, AP-HP, Georges Pompidou European Hospital, F-75015 Paris, France
| | - Sven Günther
- Université Paris Cité, Innovative Therapies in Haemostasis, INSERM UMR_S1140, Paris Cité, F-75006 Paris, France; Unité d'Explorations Fonctionnelles Respiratoires et du Sommeil, AP-HP, Georges Pompidou European Hospital, F-75015 Paris, France.
| |
Collapse
|
4
|
Karam M, Auclair C. Sphingosine-1-Phosphate as Lung and Cardiac Vasculature Protecting Agent in SARS-CoV-2 Infection. Int J Mol Sci 2023; 24:13088. [PMID: 37685894 PMCID: PMC10488186 DOI: 10.3390/ijms241713088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/16/2023] [Accepted: 08/19/2023] [Indexed: 09/10/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) may cause severe respiratory illness with high mortality. SARS-CoV-2 infection results in a massive inflammatory cell infiltration into the infected lungs accompanied by excessive pro-inflammatory cytokine production. The lung histology of dead patients shows that some areas are severely emphysematous, with enormously dilated blood vessels and micro-thromboses. The inappropriate inflammatory response damaging the pulmonary interstitial arteriolar walls suggests that the respiratory distress may come in a large part from lung vasculature injuries. It has been recently observed that low plasmatic sphingosine-1-phosphate (S1P) is a marker of a worse prognosis of clinical outcome in severe coronavirus disease (COVID) patients. S1P is an angiogenic molecule displaying anti-inflammatory and anti-apoptotic properties, that promote intercellular interactions between endothelial cells and pericytes resulting in the stabilization of arteries and capillaries. In this context, it can be hypothesized that the benefit of a normal S1P level is due to its protective effect on lung vasculature functionality. This paper provides evidence supporting this concept, opening the way for the design of a pharmacological approach involving the use of an S1P lyase inhibitor to increase the S1P level that in turn will rescue the lung vasculature functionality.
Collapse
Affiliation(s)
| | - Christian Auclair
- AC BioTech, Villejuif Biopark, Cancer Campus, 1 mail du Professeur Georges Mathé, 94800 Villejuif, France;
| |
Collapse
|
5
|
Michels EHA, Appelman B, de Brabander J, van Amstel RBE, Chouchane O, van Linge CCA, Schuurman AR, Reijnders TDY, Sulzer TAL, Klarenbeek AM, Douma RA, Bos LDJ, Wiersinga WJ, Peters-Sengers H, van der Poll T, van Agtmael M, Algera AG, Appelman B, van Baarle F, Beudel M, Bogaard HJ, Bomers M, Bonta P, Bos L, Botta M, de Brabander J, de Bree G, de Bruin S, Bugiani M, Bulle E, Buis DTP, Chouchane O, Cloherty A, Dijkstra M, Dongelmans DA, Dujardin RWG, Elbers P, Fleuren L, Geerlings S, Geijtenbeek T, Girbes A, Goorhuis B, Grobusch MP, Hagens L, Hamann J, Harris V, Hemke R, Hermans SM, Heunks L, Hollmann M, Horn J, Hovius JW, de Jong HK, de Jong MD, Koning R, Lemkes B, Lim EHT, van Mourik N, Nellen J, Nossent EJ, Olie S, Paulus F, Peters E, Pina-Fuentes DAI, van der Poll T, Preckel B, Prins JM, Raasveld J, Reijnders T, de Rotte MCFJ, Schinkel M, Schultz MJ, Schrauwen FAP, Schuurman A, Schuurmans J, Sigaloff K, Slim MA, Smeele P, Smit M, Stijnis CS, Stilma W, Teunissen C, Thoral P, Tsonas AM, Tuinman PR, van der Valk M, Veelo DP, Volleman C, de Vries H, Vught LA, van Vugt M, Wouters D, Zwinderman AHK, Brouwer MC, Wiersinga WJ, Vlaar APJ, van de Beek D. Age-related changes in plasma biomarkers and their association with mortality in COVID-19. Eur Respir J 2023; 62:2300011. [PMID: 37080568 PMCID: PMC10151455 DOI: 10.1183/13993003.00011-2023] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 04/10/2023] [Indexed: 04/22/2023]
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19)-induced mortality occurs predominantly in older patients. Several immunomodulating therapies seem less beneficial in these patients. The biological substrate behind these observations is unknown. The aim of this study was to obtain insight into the association between ageing, the host response and mortality in patients with COVID-19. METHODS We determined 43 biomarkers reflective of alterations in four pathophysiological domains: endothelial cell and coagulation activation, inflammation and organ damage, and cytokine and chemokine release. We used mediation analysis to associate ageing-driven alterations in the host response with 30-day mortality. Biomarkers associated with both ageing and mortality were validated in an intensive care unit and external cohort. RESULTS 464 general ward patients with COVID-19 were stratified according to age decades. Increasing age was an independent risk factor for 30-day mortality. Ageing was associated with alterations in each of the host response domains, characterised by greater activation of the endothelium and coagulation system and stronger elevation of inflammation and organ damage markers, which was independent of an increase in age-related comorbidities. Soluble tumour necrosis factor receptor 1, soluble triggering receptor expressed on myeloid cells 1 and soluble thrombomodulin showed the strongest correlation with ageing and explained part of the ageing-driven increase in 30-day mortality (proportion mediated: 13.0%, 12.9% and 12.6%, respectively). CONCLUSIONS Ageing is associated with a strong and broad modification of the host response to COVID-19, and specific immune changes likely contribute to increased mortality in older patients. These results may provide insight into potential age-specific immunomodulatory targets in COVID-19.
Collapse
Affiliation(s)
- Erik H A Michels
- Amsterdam UMC, location University of Amsterdam, Center for Experimental and Molecular Medicine (CEMM), Amsterdam, The Netherlands
| | - Brent Appelman
- Amsterdam UMC, location University of Amsterdam, Center for Experimental and Molecular Medicine (CEMM), Amsterdam, The Netherlands
| | - Justin de Brabander
- Amsterdam UMC, location University of Amsterdam, Center for Experimental and Molecular Medicine (CEMM), Amsterdam, The Netherlands
| | - Rombout B E van Amstel
- Amsterdam UMC, location University of Amsterdam, Department of Intensive Care Medicine, Amsterdam, The Netherlands
| | - Osoul Chouchane
- Amsterdam UMC, location University of Amsterdam, Center for Experimental and Molecular Medicine (CEMM), Amsterdam, The Netherlands
| | - Christine C A van Linge
- Amsterdam UMC, location University of Amsterdam, Center for Experimental and Molecular Medicine (CEMM), Amsterdam, The Netherlands
| | - Alex R Schuurman
- Amsterdam UMC, location University of Amsterdam, Center for Experimental and Molecular Medicine (CEMM), Amsterdam, The Netherlands
| | - Tom D Y Reijnders
- Amsterdam UMC, location University of Amsterdam, Center for Experimental and Molecular Medicine (CEMM), Amsterdam, The Netherlands
| | - Titia A L Sulzer
- Amsterdam UMC, location University of Amsterdam, Center for Experimental and Molecular Medicine (CEMM), Amsterdam, The Netherlands
| | - Augustijn M Klarenbeek
- Amsterdam UMC, location University of Amsterdam, Center for Experimental and Molecular Medicine (CEMM), Amsterdam, The Netherlands
| | - Renée A Douma
- Flevo Hospital, Department of Internal Medicine, Almere, The Netherlands
| | - Lieuwe D J Bos
- Amsterdam UMC, location University of Amsterdam, Department of Intensive Care Medicine, Amsterdam, The Netherlands
| | - W Joost Wiersinga
- Amsterdam UMC, location University of Amsterdam, Center for Experimental and Molecular Medicine (CEMM), Amsterdam, The Netherlands
- Amsterdam UMC, location University of Amsterdam, Division of Infectious Diseases, Amsterdam, The Netherlands
| | - Hessel Peters-Sengers
- Amsterdam UMC, location University of Amsterdam, Center for Experimental and Molecular Medicine (CEMM), Amsterdam, The Netherlands
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Epidemiology and Data Science, Amsterdam, The Netherlands
| | - Tom van der Poll
- Amsterdam UMC, location University of Amsterdam, Center for Experimental and Molecular Medicine (CEMM), Amsterdam, The Netherlands
- Amsterdam UMC, location University of Amsterdam, Division of Infectious Diseases, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Melegari G, Giuliani E, Di Pietro G, Alberti F, Campitiello M, Bertellini E, Barbieri A. Point-of-care pancreatic stone protein measurement in critically ill COVID-19 patients. BMC Anesthesiol 2023; 23:226. [PMID: 37391718 PMCID: PMC10311849 DOI: 10.1186/s12871-023-02187-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 06/19/2023] [Indexed: 07/02/2023] Open
Abstract
INTRODUCTION Pancreatic stone protein (PSP) is a novel biomarker that is reported to be increased in pneumonia and acute conditions. The primary aim of this study was to prospectively study plasma levels of PSP in a COVID-19 intensive care unit (ICU) population to determine how well PSP performed as a marker of mortality in comparison to other plasma biomarkers, such as C reactive protein (CRP) and procalcitonin (PCT). METHODS We collected clinical data and blood samples from COVID-19 ICU patients at the time of admission (T0), 72 h later (T1), five days later (T2), and finally, seven days later. The PSP plasma level was measured with a point-of-care system; PCT and CRP levels were measured simultaneously with laboratory tests. The inclusion criteria were being a critical COVID-19 ICU patient requiring ventilatory mechanical assistance. RESULTS We enrolled 21 patients and evaluated 80 blood samples; we found an increase in PSP plasma levels according to mixed model analysis over time (p < 0.001), with higher levels found in the nonsurvivor population (p < 0.001). Plasma PSP levels achieved a statistically significant result in terms of the AUROC, with a value higher than 0.7 at T0, T1, T2, and T3. The overall AUROC of PSP was 0.8271 (CI (0.73-0.93), p < 0.001). These results were not observed for CRP and PCT. CONCLUSION These first results suggest the potential advantages of monitoring PSP plasma levels through point-of-care technology, which could be useful in the absence of a specific COVID-19 biomarker. Additional data are needed to confirm these results.
Collapse
Affiliation(s)
- Gabriele Melegari
- Department of Anaesthesia and Intensive Care, Azienda Ospedaliero Universitaria Di Modena, Modena, Italy.
| | | | - Giulia Di Pietro
- School of Anaesthesia and Intensive Care, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesco Alberti
- School of Anaesthesia and Intensive Care, University of Modena and Reggio Emilia, Modena, Italy
| | - Mattia Campitiello
- School of Anaesthesia and Intensive Care, University of Modena and Reggio Emilia, Modena, Italy
| | - Elisabetta Bertellini
- Department of Anaesthesia and Intensive Care, Azienda Ospedaliero Universitaria Di Modena, Modena, Italy
| | - Alberto Barbieri
- School of Anaesthesia and Intensive Care, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
7
|
Cusack R, Bos LD, Povoa P, Martin-Loeches I. Endothelial dysfunction triggers acute respiratory distress syndrome in patients with sepsis: a narrative review. Front Med (Lausanne) 2023; 10:1203827. [PMID: 37332755 PMCID: PMC10272540 DOI: 10.3389/fmed.2023.1203827] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 05/16/2023] [Indexed: 06/20/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a severe organ failure occurring mainly in critically ill patients as a result of different types of insults such as sepsis, trauma or aspiration. Sepsis is the main cause of ARDS, and it contributes to a high mortality and resources consumption both in hospital setting and in the community. ARDS develops mainly an acute respiratory failure with severe and often refractory hypoxemia. ARDS also has long term implications and sequelae. Endothelial damage plays an important role in the pathogenesis of ARDS. Understanding the mechanisms of ARDS presents opportunities for novel diagnostic and therapeutic targets. Biochemical signals can be used in concert to identify and classify patients into ARDS phenotypes allowing earlier effective treatment with personalised therapies. This is a narrative review where we aimed to flesh out the pathogenetic mechanisms and heterogeneity of ARDS. We examine the links between endothelium damage and its contribution to organ failure. We have also investigated future strategies for treatment with a special emphasis in endothelial damage.
Collapse
Affiliation(s)
- Rachael Cusack
- Department of Intensive Care, St. James’s Hospital, Dublin, Ireland
- School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Lieuwe D. Bos
- Intensive Care, Amsterdam UMC Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Pedro Povoa
- NOVA Medical School, CHRC, New University of Lisbon, Lisbon, Portugal
- Center for Clinical Epidemiology and Research Unit of Clinical Epidemiology, OUH Odense University Hospital, Odense, Denmark
- Department of Intensive Care, Hospital de São Francisco Xavier, CHLO, Lisbon, Portugal
| | - Ignacio Martin-Loeches
- Department of Intensive Care, St. James’s Hospital, Dublin, Ireland
- School of Medicine, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
8
|
Wadhwa B, Malhotra V, Kerai S, Husain F, Pandey NB, Saxena KN, Singh V, Quinn TM, Li F, Gaughan E, Shankar-Hari M, Mills B, Antonelli J, Bruce A, Finlayson K, Moore A, Dhaliwal K, Edwards C. Phase 2 randomised placebo-controlled trial of spironolactone and dexamethasone versus dexamethasone in COVID-19 hospitalised patients in Delhi. BMC Infect Dis 2023; 23:326. [PMID: 37189034 PMCID: PMC10184093 DOI: 10.1186/s12879-023-08286-w] [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: 07/06/2022] [Accepted: 04/27/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND In this phase 2 randomised placebo-controlled clinical trial in patients with COVID-19, we hypothesised that blocking mineralocorticoid receptors using a combination of dexamethasone to suppress cortisol secretion and spironolactone is safe and may reduce illness severity. METHODS Hospitalised patients with confirmed COVID-19 were randomly allocated to low dose oral spironolactone (50 mg day 1, then 25 mg once daily for 21 days) or standard of care in a 2:1 ratio. Both groups received dexamethasone 6 mg daily for 10 days. Group allocation was blinded to the patient and research team. Primary outcomes were time to recovery, defined as the number of days until patients achieved WHO Ordinal Scale (OS) category ≤ 3, and the effect of spironolactone on aldosterone, D-dimer, angiotensin II and Von Willebrand Factor (VWF). RESULTS One hundred twenty patients with PCR confirmed COVID were recruited in Delhi from 01 February to 30 April 2021. 74 were randomly assigned to spironolactone and dexamethasone (SpiroDex), and 46 to dexamethasone alone (Dex). There was no significant difference in the time to recovery between SpiroDex and Dex groups (SpiroDex median 4.5 days, Dex median 5.5 days, p = 0.055). SpiroDex patients had significantly lower D-dimer levels on days 4 and 7 (day 7 mean D-dimer: SpiroDex 1.15 µg/mL, Dex 3.15 µg/mL, p = 0.0004) and aldosterone at day 7 (SpiroDex 6.8 ng/dL, Dex 14.52 ng/dL, p = 0.0075). There was no difference in VWF or angiotensin II levels between groups. For secondary outcomes, SpiroDex patients had a significantly greater number of oxygen free days and reached oxygen freedom sooner than the Dex group. Cough scores were no different during the acute illness, however the SpiroDex group had lower scores at day 28. There was no difference in corticosteroid levels between groups. There was no increase in adverse events in patients receiving SpiroDex. CONCLUSION Low dose oral spironolactone in addition to dexamethasone was safe and reduced D-dimer and aldosterone. Time to recovery was not significantly reduced. Phase 3 randomised controlled trials with spironolactone and dexamethasone should be considered. TRIAL REGISTRATION The trial was registered on the Clinical Trials Registry of India TRI: CTRI/2021/03/031721, reference: REF/2021/03/041472. Registered on 04/03/2021.
Collapse
Affiliation(s)
- Bharti Wadhwa
- Department of Anaesthesia, Maulana Azad Medical College, New Delhi, India.
| | - Vikas Malhotra
- Department of ENT & Head and Neck Surgery, Maulana Azad Medical College & Associated Hospitals, New Delhi, India
| | - Sukhyanti Kerai
- Department of Anaesthesia, Maulana Azad Medical College, New Delhi, India
| | - Farah Husain
- Department of Anaesthesia, Maulana Azad Medical College, New Delhi, India
| | - Nalini Bala Pandey
- Department of Anaesthesia, Maulana Azad Medical College, New Delhi, India
| | - Kirti N Saxena
- Department of Anaesthesia, Maulana Azad Medical College, New Delhi, India
| | - Vinay Singh
- Department of Anaesthesia, Maulana Azad Medical College, New Delhi, India
| | - Tom M Quinn
- Centre for Inflammation Research, The Queen's Medical Research Institute, BioQuarter, The University of Edinburgh, Edinburgh, EH16 4TJ, UK
- Royal Infirmary of Edinburgh, BioQuarter, Little France, Edinburgh, EH16 4SA, UK
| | - Feng Li
- Centre for Inflammation Research, The Queen's Medical Research Institute, BioQuarter, The University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Erin Gaughan
- Centre for Inflammation Research, The Queen's Medical Research Institute, BioQuarter, The University of Edinburgh, Edinburgh, EH16 4TJ, UK
- Royal Infirmary of Edinburgh, BioQuarter, Little France, Edinburgh, EH16 4SA, UK
| | - Manu Shankar-Hari
- Centre for Inflammation Research, The Queen's Medical Research Institute, BioQuarter, The University of Edinburgh, Edinburgh, EH16 4TJ, UK
- Royal Infirmary of Edinburgh, BioQuarter, Little France, Edinburgh, EH16 4SA, UK
| | - Bethany Mills
- Centre for Inflammation Research, The Queen's Medical Research Institute, BioQuarter, The University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Jean Antonelli
- Centre for Inflammation Research, The Queen's Medical Research Institute, BioQuarter, The University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Annya Bruce
- Centre for Inflammation Research, The Queen's Medical Research Institute, BioQuarter, The University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Keith Finlayson
- Centre for Inflammation Research, The Queen's Medical Research Institute, BioQuarter, The University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Anne Moore
- Centre for Inflammation Research, The Queen's Medical Research Institute, BioQuarter, The University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Kevin Dhaliwal
- Centre for Inflammation Research, The Queen's Medical Research Institute, BioQuarter, The University of Edinburgh, Edinburgh, EH16 4TJ, UK.
- Royal Infirmary of Edinburgh, BioQuarter, Little France, Edinburgh, EH16 4SA, UK.
| | | |
Collapse
|
9
|
Rejeski K, Blumenberg V, Iacoboni G, Lopez-Corral L, Kharboutli S, Hernani R, Petrera A, Müller N, Hildebrand F, Frölich L, Karschnia P, Schmidt C, Cordas dos Santos DM, Piñana JL, Müller F, Martin AA, Dreyling M, von Bergwelt-Baildon M, Barba P, Subklewe M, Bücklein VL. Identifying Early Infections in the Setting of CRS With Routine and Exploratory Serum Proteomics and the HT10 Score Following CD19 CAR-T for Relapsed/Refractory B-NHL. Hemasphere 2023; 7:e858. [PMID: 37038465 PMCID: PMC10082278 DOI: 10.1097/hs9.0000000000000858] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 02/06/2023] [Indexed: 04/08/2023] Open
Abstract
Early fever after chimeric antigen receptor T-cell (CAR-T) therapy can reflect both an infection or cytokine release syndrome (CRS). Identifying early infections in the setting of CRS and neutropenia represents an unresolved clinical challenge. In this retrospective observational analysis, early fever events (day 0-30) were characterized as infection versus CRS in 62 patients treated with standard-of-care CD19.CAR-T for relapsed/refractory B-cell non-Hodgkin lymphoma. Routine serum inflammatory markers (C-reactive protein [CRP], interleukin-6 [IL-6], procalcitonin [PCT]) were recorded daily. Exploratory plasma proteomics were performed longitudinally in 52 patients using a multiplex proximity extension assay (Olink proteomics). Compared with the CRSonly cohort, we noted increased event-day IL-6 (median 2243 versus 64 pg/mL, P = 0.03) and particularly high PCT levels (median 1.6 versus 0.3 µg/L, P < 0.0001) in the patients that developed severe infections. For PCT, an optimal discriminatory threshold of 1.5 µg/L was established (area under the receiver operating characteristic curve [AUCROC] = 0.78). Next, we incorporated day-of-fever PCT levels with the patient-individual CAR-HEMATOTOX score. In a multicenter validation cohort (n = 125), we confirmed the discriminatory capacity of this so-called HT10 score for early infections at first fever (AUCROC = 0.87, P < 0.0001, sens. 86%, spec. 86%). Additionally, Olink proteomics revealed pronounced immune dysregulation and endothelial dysfunction in patients with severe infections as evidenced by an increased ANGPT2/1 ratio and an altered CD40/CD40L-axis. In conclusion, the high discriminatory capacity of the HT10 score for infections highlights the advantage of dynamic risk assessment and supports the incorporation of PCT into routine inflammatory panels. Candidate markers from Olink proteomics may further refine risk-stratification. If validated prospectively, the score will enable risk-adapted decisions on antibiotic use.
Collapse
Affiliation(s)
- Kai Rejeski
- Department of Medicine III – Hematology/Oncology, University Hospital, LMU Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
- German Cancer Consortium (DKTK), Munich Site, and German Cancer Research Center, Heidelberg, Germany
- Bavarian Cancer Research Center (BZKF), Partner Sites Munich and Erlangen, Germany
| | - Viktoria Blumenberg
- Department of Medicine III – Hematology/Oncology, University Hospital, LMU Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
- German Cancer Consortium (DKTK), Munich Site, and German Cancer Research Center, Heidelberg, Germany
- Bavarian Cancer Research Center (BZKF), Partner Sites Munich and Erlangen, Germany
| | - Gloria Iacoboni
- Department of Hematology, Vall d’Hebron Institute of Oncology (VHIO), University Hospital Vall d’Hebron, Barcelona, Spain
- Department of Medicine, Universitat Autònoma of Barcelona (UAB), Bellaterra, Spain
| | - Lucia Lopez-Corral
- Hematology Department, Hospital Clínico Universitario de Salamanca, IBSAL, CIBERONC, Salamanca, Spain
- Centro de Investigación del Cáncer-IBMCC, Salamanca, Spain
| | - Soraya Kharboutli
- Bavarian Cancer Research Center (BZKF), Partner Sites Munich and Erlangen, Germany
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg and University Hospital Erlangen, Germany
| | - Rafael Hernani
- Hematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Agnese Petrera
- Metabolomics and Proteomics Core Facility, Helmholtz Zentrum Munich – German Research Center for Environmental Health, Munich, Germany
| | - Niklas Müller
- Department of Medicine III – Hematology/Oncology, University Hospital, LMU Munich, Germany
| | - Friederike Hildebrand
- Department of Medicine III – Hematology/Oncology, University Hospital, LMU Munich, Germany
| | - Lisa Frölich
- Department of Medicine III – Hematology/Oncology, University Hospital, LMU Munich, Germany
- German Cancer Consortium (DKTK), Munich Site, and German Cancer Research Center, Heidelberg, Germany
| | - Philipp Karschnia
- Department of Neurosurgery, University Hospital, LMU Munich, Germany
| | - Christian Schmidt
- Department of Medicine III – Hematology/Oncology, University Hospital, LMU Munich, Germany
| | - David M. Cordas dos Santos
- Department of Medicine III – Hematology/Oncology, University Hospital, LMU Munich, Germany
- German Cancer Consortium (DKTK), Munich Site, and German Cancer Research Center, Heidelberg, Germany
| | - José Luis Piñana
- Hematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Fabian Müller
- Bavarian Cancer Research Center (BZKF), Partner Sites Munich and Erlangen, Germany
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg and University Hospital Erlangen, Germany
| | - Ana Africa Martin
- Hematology Department, Hospital Clínico Universitario de Salamanca, IBSAL, CIBERONC, Salamanca, Spain
- Centro de Investigación del Cáncer-IBMCC, Salamanca, Spain
| | - Martin Dreyling
- Department of Medicine III – Hematology/Oncology, University Hospital, LMU Munich, Germany
| | - Michael von Bergwelt-Baildon
- Department of Medicine III – Hematology/Oncology, University Hospital, LMU Munich, Germany
- German Cancer Consortium (DKTK), Munich Site, and German Cancer Research Center, Heidelberg, Germany
- Bavarian Cancer Research Center (BZKF), Partner Sites Munich and Erlangen, Germany
| | - Pere Barba
- Department of Hematology, Vall d’Hebron Institute of Oncology (VHIO), University Hospital Vall d’Hebron, Barcelona, Spain
- Department of Medicine, Universitat Autònoma of Barcelona (UAB), Bellaterra, Spain
| | - Marion Subklewe
- Department of Medicine III – Hematology/Oncology, University Hospital, LMU Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
- German Cancer Consortium (DKTK), Munich Site, and German Cancer Research Center, Heidelberg, Germany
- Bavarian Cancer Research Center (BZKF), Partner Sites Munich and Erlangen, Germany
| | - Veit L. Bücklein
- Department of Medicine III – Hematology/Oncology, University Hospital, LMU Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
- German Cancer Consortium (DKTK), Munich Site, and German Cancer Research Center, Heidelberg, Germany
- Bavarian Cancer Research Center (BZKF), Partner Sites Munich and Erlangen, Germany
| |
Collapse
|
10
|
Mohebbi A, Haybar H, Nakhaei Moghaddam F, Rasti Z, Vahid MA, Saki N. Biomarkers of endothelial dysfunction are associated with poor outcome in COVID-19 patients: A systematic review and meta-analysis. Rev Med Virol 2023:e2442. [PMID: 36943015 DOI: 10.1002/rmv.2442] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/08/2023] [Accepted: 03/14/2023] [Indexed: 03/23/2023]
Abstract
Numerous studies have linked coronavirus disease 2019 (COVID-19) with endothelial dysfunction and reported elevated levels of endothelial biomarkers in this disease. We conducted a systematic review and meta-analysis of the published evidence in this respect. A systematic literature search of PubMed and Scopus databases was performed to find studies investigating biomarkers of endothelial dysfunction in COVID-19 patients. Pooled standardized mean differences and their 95% confidence intervals were calculated for each biomarker using random effect model. 74 studies with 7668 patients were included. In comparison to patients with good outcome, those with poor outcome had higher levels of von Willebrand factor (vWF) (SMD: 0.83, 95% CI: 0.59-1.07, p < 0.00001), vWF:ADAMTS13 (1.23, (0.77-1.7), p < 0.00001), angiopoietin-2 (Ang-2) (1.06 (0.6-1.51), p < 0.0001), E-selectin (1.09 (0.55-1.63), p < 0.0001), P-selectin (0.59 (0.24-0.94), p = 0.001), syndecan-1 (0.99 (0.6-1.37), p < 0.00001), mid-regional pro-adrenomedullin (MR-proADM) (1.52 (1.35-1.68), p < 0.00001), vascular endothelial growth factor (0.27 (0.02-0.53), p = 0.03), soluble fms-like tyrosine kinase-1 (sFLT-1) (1.93 (0.65-3.21), p = 0.03) and lower levels of ADAMTS13 antigen (-0.69 (-0.9 to -0.47) p < 0.00001) and activity (-0.84 (-1.06 to -0.61) p < 0.0000). Plasminogen activator inhibitor-1 and tissue plasminogen activator levels were not different between the two groups (p < 0.05). There were elevated levels of endothelial dysfunction biomarkers in COVID-19 patients with poor outcome, indicating their possible role in disease severity and prognosis. In particular, MR-proADM, vWF, syndecan-1 and sFLT-1 showed a significant association with poor outcome in these patients.
Collapse
Affiliation(s)
- Alireza Mohebbi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Habib Haybar
- Department of Cardiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fatemeh Nakhaei Moghaddam
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Zahra Rasti
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Vahid
- Department of Medical Laboratory, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Najmaldin Saki
- Department of Medical Laboratory, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
11
|
Plasminogen activator inhibitor 1 is not a major causative factor for exacerbation in a mouse model of SARS-CoV-2 infection. Sci Rep 2023; 13:3103. [PMID: 36813909 PMCID: PMC9944779 DOI: 10.1038/s41598-023-30305-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 02/21/2023] [Indexed: 02/24/2023] Open
Abstract
Coronavirus disease (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remains a global pandemic. Although several vaccines targeting SARS-CoV-2 spike proteins protect against COVID-19 infection, mutations affecting virus transmissibility and immune evasion potential have reduced their efficacy, leading to the need for a more efficient strategy. Available clinical evidence regarding COVID-19 suggests that endothelial dysfunction with thrombosis is a central pathogenesis of progression to systemic disease, in which overexpression of plasminogen activator inhibitor-1 (PAI-1) may be important. Here we developed a novel peptide vaccine against PAI-1 and evaluated its effect on lipopolysaccharide (LPS)-induced sepsis and SARS-CoV-2 infection in mice. Administration of LPS and mouse-adapted SARS-CoV-2 increased serum PAI-1 levels, although the latter showed smaller levels. In an LPS-induced sepsis model, mice immunized with PAI-1 vaccine showed reduced organ damage and microvascular thrombosis and improved survival compared with vehicle-treated mice. In plasma clot lysis assays, vaccination-induced serum IgG antibodies were fibrinolytic. However, in a SARS-CoV-2 infection model, survival and symptom severity (i.e., body weight reduction) did not differ between vaccine- and vehicle-treated groups. These results indicate that although PAI-1 may promote the severity of sepsis by increasing thrombus formation, it might not be a major contributor to COVID-19 exacerbation.
Collapse
|
12
|
Karabulut Uzunçakmak S. SARS-CoV-2 Infection and Candidate Biomarkers. Eurasian J Med 2022; 54:16-22. [PMID: 36655440 PMCID: PMC11163343 DOI: 10.5152/eurasianjmed.2022.22305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 11/27/2022] [Indexed: 01/19/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 is a virus that can still infect individuals and whose deadly effects continue despite the current vaccines and drugs. Since 2019, many studies on the pathogenesis of the disease have been completed and continue to be done. In addition to the diagnosis and treatment of the disease, many molecules that can be markers of the disease have been investigated. In the early stages of the pandemic, many nonspecific and infection-related laboratory findings and chest computed tomography were used to obtain information about the diagnosis of the disease. The more individual molecules became associated with the disease yet. The purpose of this review is to summarize the impact and role of many molecules associated with coronavirus disease-2019 infection that have been previously used and newly revealed. Numerous studies are summarized in this review. The obtained data show that previously used laboratory findings and new potential biomarkers are not specific to the disease. New potential biomarkers have been associated with the severity of the disease itself, as can be seen with lung imaging and even with routine laboratory findings. One of the important points that are seen frequently in studies is that the effectiveness of these molecules has been shown not only in coronavirus disease-2019 infection but also in many other diseases. This removes the pathogenesis of the disease from being a unique mechanism created by the Severe acute respiratory syndrome coronavirus 2 and provides a general perspective formed by viral or bacterial infections. However, there are still many molecular changes that need to be investigated. Future studies will continue to update themselves with the mutations of the virus.
Collapse
|
13
|
Fogarty H, Ward SE, Townsend L, Karampini E, Elliott S, Conlon N, Dunne J, Kiersey R, Naughton A, Gardiner M, Byrne M, Bergin C, O'Sullivan JM, Martin‐Loeches I, Nadarajan P, Bannan C, Mallon PW, Curley GF, Preston RJS, Rehill AM, Baker RI, Cheallaigh CN, O'Donnell JS. Sustained VWF-ADAMTS-13 axis imbalance and endotheliopathy in long COVID syndrome is related to immune dysfunction. J Thromb Haemost 2022; 20:2429-2438. [PMID: 35875995 PMCID: PMC9349977 DOI: 10.1111/jth.15830] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/28/2022] [Accepted: 07/19/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Prolonged recovery is common after acute SARS-CoV-2 infection; however, the pathophysiological mechanisms underpinning Long COVID syndrome remain unknown. VWF/ADAMTS-13 imbalance, dysregulated angiogenesis, and immunothrombosis are hallmarks of acute COVID-19. We hypothesized that VWF/ADAMTS-13 imbalance persists in convalescence together with endothelial cell (EC) activation and angiogenic disturbance. Additionally, we postulate that ongoing immune cell dysfunction may be linked to sustained EC and coagulation activation. PATIENTS AND METHODS Fifty patients were reviewed at a minimum of 6 weeks following acute COVID-19. ADAMTS-13, Weibel Palade Body (WPB) proteins, and angiogenesis-related proteins were assessed and clinical evaluation and immunophenotyping performed. Comparisons were made with healthy controls (n = 20) and acute COVID-19 patients (n = 36). RESULTS ADAMTS-13 levels were reduced (p = 0.009) and the VWF-ADAMTS-13 ratio was increased in convalescence (p = 0.0004). Levels of platelet factor 4 (PF4), a putative protector of VWF, were also elevated (p = 0.0001). A non-significant increase in WPB proteins Angiopoietin-2 (Ang-2) and Osteoprotegerin (OPG) was observed in convalescent patients and WPB markers correlated with EC parameters. Enhanced expression of 21 angiogenesis-related proteins was observed in convalescent COVID-19. Finally, immunophenotyping revealed significantly elevated intermediate monocytes and activated CD4+ and CD8+ T cells in convalescence, which correlated with thrombin generation and endotheliopathy markers, respectively. CONCLUSION Our data provide insights into sustained EC activation, dysregulated angiogenesis, and VWF/ADAMTS-13 axis imbalance in convalescent COVID-19. In keeping with the pivotal role of immunothrombosis in acute COVID-19, our findings support the hypothesis that abnormal T cell and monocyte populations may be important in the context of persistent EC activation and hemostatic dysfunction during convalescence.
Collapse
Affiliation(s)
- Helen Fogarty
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular SciencesRoyal College of Surgeons in IrelandDublinIreland
| | - Soracha E. Ward
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular SciencesRoyal College of Surgeons in IrelandDublinIreland
| | - Liam Townsend
- Department of Infectious DiseasesSt James's HospitalDublinIreland
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine InstituteTrinity College DublinDublinIreland
| | - Ellie Karampini
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular SciencesRoyal College of Surgeons in IrelandDublinIreland
| | - Stephanie Elliott
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular SciencesRoyal College of Surgeons in IrelandDublinIreland
| | - Niall Conlon
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine InstituteTrinity College DublinDublinIreland
- Department of ImmunologySt James's HospitalDublinIreland
| | - Jean Dunne
- Department of ImmunologySt James's HospitalDublinIreland
| | - Rachel Kiersey
- Department of ImmunologySt James's HospitalDublinIreland
| | | | - Mary Gardiner
- Department of ImmunologySt James's HospitalDublinIreland
| | - Mary Byrne
- National Coagulation CentreSt James's HospitalDublinIreland
| | - Colm Bergin
- Department of Infectious DiseasesSt James's HospitalDublinIreland
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine InstituteTrinity College DublinDublinIreland
| | - Jamie M. O'Sullivan
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular SciencesRoyal College of Surgeons in IrelandDublinIreland
| | | | | | - Ciaran Bannan
- Department of Infectious DiseasesSt James's HospitalDublinIreland
| | - Patrick W. Mallon
- Centre for Experimental Pathogen Host ResearchUniversity College DublinDublinIreland
- St Vincent's University HospitalDublinIreland
| | - Gerard F. Curley
- Department of Anaesthesia and Critical CareRoyal College of Surgeons in IrelandDublinIreland
| | - Roger J. S. Preston
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular SciencesRoyal College of Surgeons in IrelandDublinIreland
- National Children's Research CentreOur Lady's Children's Hospital CrumlinDublinIreland
| | - Aisling M. Rehill
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular SciencesRoyal College of Surgeons in IrelandDublinIreland
| | - Ross I. Baker
- Western Australia Centre for Thrombosis and Haemostasis, Perth Blood InstituteMurdoch UniversityPerthWestern AustraliaAustralia
- Irish‐Australian Blood Collaborative (IABC) NetworkDublinIreland
| | - Cliona Ni Cheallaigh
- Department of Infectious DiseasesSt James's HospitalDublinIreland
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine InstituteTrinity College DublinDublinIreland
| | - James S. O'Donnell
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular SciencesRoyal College of Surgeons in IrelandDublinIreland
- National Coagulation CentreSt James's HospitalDublinIreland
- National Children's Research CentreOur Lady's Children's Hospital CrumlinDublinIreland
- Irish‐Australian Blood Collaborative (IABC) NetworkDublinIreland
| |
Collapse
|
14
|
Sojka M, Drelich-Zbroja A, Kuczyńska M, Cheda M, Dąbrowska I, Kopyto E, Halczuk I, Zbroja M, Cyranka W, Jargiełło T. Ischemic and Hemorrhagic Cerebrovascular Events Related to COVID-19 Coagulopathy and Hypoxemia. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:11823. [PMID: 36142094 PMCID: PMC9517511 DOI: 10.3390/ijerph191811823] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/12/2022] [Accepted: 09/15/2022] [Indexed: 06/16/2023]
Abstract
Since the very beginning of the COVID-19 pandemic, numerous researchers have made an effort to determine the molecular composition of the SARS-CoV-2 virus, and the exact pathomechanism through which the virus exerts such a devastating effect on the host/infected organism. Recent scientific evidence highlights the affinity of the virus towards ACE2 receptors, which are widespread in multiple human systems, including the central nervous system (CNS) and cerebral vessels. Such an affinity may explain endothelial dysfunction and damage that is observed in COVID-positive patients in histopathological studies, with subsequent dysregulation of the cerebral circulation leading to transient or acute cerebrovascular accidents. In this paper, we aimed to evaluate the effects of COVID-related hypoxemia and direct viral invasion on the cerebral circulation, with special respect to the postulated pathomechanism, vulnerable groups of patients, clinical course and outcomes, as well as diagnostic imaging findings.
Collapse
Affiliation(s)
- Michał Sojka
- Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-090 Lublin, Poland
| | - Anna Drelich-Zbroja
- Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-090 Lublin, Poland
| | - Maryla Kuczyńska
- Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-090 Lublin, Poland
| | - Mateusz Cheda
- Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-090 Lublin, Poland
| | - Izabela Dąbrowska
- Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-090 Lublin, Poland
| | - Ewa Kopyto
- Students’ Scientific Society at the Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-090 Lublin, Poland
| | - Izabela Halczuk
- Students’ Scientific Society at the Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-090 Lublin, Poland
| | - Monika Zbroja
- Students’ Scientific Society at the Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-090 Lublin, Poland
| | - Weronika Cyranka
- Students’ Scientific Society at the Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-090 Lublin, Poland
| | - Tomasz Jargiełło
- Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-090 Lublin, Poland
| |
Collapse
|
15
|
Melegari G, Critelli RM, Lasagni S, Romagnoli D, Bertellini E, Villa E. Dynamic Angiopoietin-2 Serum Level as Endothelial Damage Marker and Potential Therapeutic Target. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1336-1337. [PMID: 36064255 PMCID: PMC9439706 DOI: 10.1016/j.ajpath.2022.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 11/17/2022]
Affiliation(s)
- Gabriele Melegari
- Department of Anaesthesia and Intensive Care, Azienda Ospedaliero Universitaria di Modena, Modena, Italy
| | - Rosina M Critelli
- Surgical, Medical and Dental Department of Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Simone Lasagni
- Surgical, Medical and Dental Department of Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Dante Romagnoli
- Surgical, Medical and Dental Department of Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Elisabetta Bertellini
- Department of Anaesthesia and Intensive Care, Azienda Ospedaliero Universitaria di Modena, Modena, Italy
| | - Erica Villa
- Surgical, Medical and Dental Department of Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
16
|
Price DR, Benedetti E, Krumsiek J. Authors' Reply. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1337-1338. [PMID: 36064256 PMCID: PMC9439708 DOI: 10.1016/j.ajpath.2022.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 11/28/2022]
Affiliation(s)
- David R Price
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York-Presbyterian Hospital-Weill Cornell Medical Center, Weill Cornell Medicine, New York, New York; Department of Medicine, New York-Presbyterian Hospital-Weill Cornell Medical Center, New York, New York.
| | - Elisa Benedetti
- Institute of Computational Biomedicine, Weill Cornell Medicine, New York, New York; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York
| | - Jan Krumsiek
- Institute of Computational Biomedicine, Weill Cornell Medicine, New York, New York; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York
| |
Collapse
|
17
|
Abstract
COVID-19 is a primary respiratory illness that is frequently complicated by systemic involvement of the vasculature. Vascular involvement leads to an array of complications ranging from thrombosis to pulmonary edema secondary to loss of barrier function. This review will address the vasculopathy of COVID-19 with a focus on the role of the endothelium in orchestrating the systemic response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. The endothelial receptor systems and molecular pathways activated in the setting of COVID-19 and the consequences of these inflammatory and prothrombotic changes on endothelial cell function will be discussed. The sequelae of COVID-19 vascular involvement at the level of organ systems will also be addressed, with an emphasis on the pulmonary vasculature but with consideration of effects on other vascular beds. The dramatic changes in endothelial phenotypes associated with COVID-19 has enabled the identification of biomarkers that could help guide therapy and predict outcomes. Knowledge of vascular pathogenesis in COVID-19 has also informed therapeutic approaches that may control its systemic sequelae. Because our understanding of vascular response in COVID-19 continues to evolve, we will consider areas of controversy, such as the extent to which SARS-CoV-2 directly infects endothelium and the degree to which vascular responses to SARS-CoV-2 are unique or common to those of other viruses capable of causing severe respiratory disease. This conceptual framework describing how SARS-CoV-2 infection affects endothelial inflammation, prothrombotic transformation, and barrier dysfunction will provide a context for interpreting new information as it arises addressing the vascular complications of COVID-19.
Collapse
Affiliation(s)
| | | | - Alec A Schmaier
- Division of Hemostasis and Thrombosis and
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| |
Collapse
|
18
|
Schuurman AR, Reijnders TDY, van Engelen TSR, Léopold V, de Brabander J, van Linge C, Schinkel M, Pereverzeva L, Haak BW, Brands X, Kanglie MMNP, van den Berk IAH, Douma RA, Faber DR, Nanayakkara PWB, Stoker J, Prins JM, Scicluna BP, Wiersinga WJ, van der Poll T. The host response in different aetiologies of community-acquired pneumonia. EBioMedicine 2022; 81:104082. [PMID: 35660785 PMCID: PMC9155985 DOI: 10.1016/j.ebiom.2022.104082] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/06/2022] [Accepted: 05/13/2022] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Community-acquired pneumonia (CAP) can be caused by a variety of pathogens, of which Streptococcus pneumoniae, Influenza and currently SARS-CoV-2 are the most common. We sought to identify shared and pathogen-specific host response features by directly comparing different aetiologies of CAP. METHODS We measured 72 plasma biomarkers in a cohort of 265 patients hospitalized for CAP, all sampled within 48 hours of admission, and 28 age-and sex matched non-infectious controls. We stratified the biomarkers into several pathophysiological domains- antiviral response, vascular response and function, coagulation, systemic inflammation, and immune checkpoint markers. We directly compared CAP caused by SARS-CoV-2 (COVID-19, n=39), Streptococcus pneumoniae (CAP-strep, n=27), Influenza (CAP-flu, n=22) and other or unknown pathogens (CAP-other, n=177). We adjusted the comparisons for age, sex and disease severity scores. FINDINGS Biomarkers reflective of a stronger cell-mediated antiviral response clearly separated COVID-19 from other CAPs (most notably granzyme B). Biomarkers reflecting activation and function of the vasculature showed endothelial barrier integrity was least affected in COVID-19, while glycocalyx degradation and angiogenesis were enhanced relative to other CAPs. Notably, markers of coagulation activation, including D-dimer, were not different between the CAP groups. Ferritin was most increased in COVID-19, while other systemic inflammation biomarkers such as IL-6 and procalcitonin were highest in CAP-strep. Immune checkpoint markers showed distinctive patterns in viral and non-viral CAP, with highly elevated levels of Galectin-9 in COVID-19. INTERPRETATION Our investigation provides insight into shared and distinct pathophysiological mechanisms in different aetiologies of CAP, which may help guide new pathogen-specific therapeutic strategies. FUNDING This study was financially supported by the Dutch Research Council, the European Commission and the Netherlands Organization for Health Research and Development.
Collapse
Affiliation(s)
- Alex R Schuurman
- Centre for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centres - Location AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Tom D Y Reijnders
- Centre for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centres - Location AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Tjitske S R van Engelen
- Centre for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centres - Location AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Valentine Léopold
- Centre for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centres - Location AMC, University of Amsterdam, Amsterdam, the Netherlands; Department of Anaesthesiology and Intensive Care, GH St Louis-Lariboisière, Inserm UMR-S 942 (MASCOT), Université de Paris, 75010 Paris, France
| | - Justin de Brabander
- Centre for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centres - Location AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Christine van Linge
- Centre for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centres - Location AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Michiel Schinkel
- Centre for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centres - Location AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Liza Pereverzeva
- Centre for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centres - Location AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Bastiaan W Haak
- Centre for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centres - Location AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Xanthe Brands
- Centre for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centres - Location AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Maadrika M N P Kanglie
- Department of Radiology, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, The Netherlands
| | - Inge A H van den Berk
- Department of Radiology, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, The Netherlands
| | - Renée A Douma
- Department of Internal Medicine, Flevo Hospital, Almere, the Netherlands
| | - Daniël R Faber
- Department of Internal Medicine, BovenIJ Hospital, Amsterdam, the Netherlands
| | - Prabath W B Nanayakkara
- Department of Internal Medicine, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, the Netherlands
| | - Jaap Stoker
- Department of Radiology, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, The Netherlands
| | - Jan M Prins
- Department of Internal Medicine, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, the Netherlands
| | - Brendon P Scicluna
- Centre for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centres - Location AMC, University of Amsterdam, Amsterdam, the Netherlands; Centre for Molecular Medicine and Biobanking, University of Malta, Malta; Department of Applied Biomedical Science, Faculty of Health Sciences, Mater Dei hospital, University of Malta, Malta
| | - W Joost Wiersinga
- Centre for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centres - Location AMC, University of Amsterdam, Amsterdam, the Netherlands; Department of Internal Medicine, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, the Netherlands
| | - Tom van der Poll
- Centre for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centres - Location AMC, University of Amsterdam, Amsterdam, the Netherlands; Department of Internal Medicine, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
19
|
Gene Networks of Hyperglycemia, Diabetic Complications, and Human Proteins Targeted by SARS-CoV-2: What Is the Molecular Basis for Comorbidity? Int J Mol Sci 2022; 23:ijms23137247. [PMID: 35806251 PMCID: PMC9266766 DOI: 10.3390/ijms23137247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 12/10/2022] Open
Abstract
People with diabetes are more likely to have severe COVID-19 compared to the general population. Moreover, diabetes and COVID-19 demonstrate a certain parallelism in the mechanisms and organ damage. In this work, we applied bioinformatics analysis of associative molecular networks to identify key molecules and pathophysiological processes that determine SARS-CoV-2-induced disorders in patients with diabetes. Using text-mining-based approaches and ANDSystem as a bioinformatics tool, we reconstructed and matched networks related to hyperglycemia, diabetic complications, insulin resistance, and beta cell dysfunction with networks of SARS-CoV-2-targeted proteins. The latter included SARS-CoV-2 entry receptors (ACE2 and DPP4), SARS-CoV-2 entry associated proteases (TMPRSS2, CTSB, and CTSL), and 332 human intracellular proteins interacting with SARS-CoV-2. A number of genes/proteins targeted by SARS-CoV-2 (ACE2, BRD2, COMT, CTSB, CTSL, DNMT1, DPP4, ERP44, F2RL1, GDF15, GPX1, HDAC2, HMOX1, HYOU1, IDE, LOX, NUTF2, PCNT, PLAT, RAB10, RHOA, SCARB1, and SELENOS) were found in the networks of vascular diabetic complications and insulin resistance. According to the Gene Ontology enrichment analysis, the defined molecules are involved in the response to hypoxia, reactive oxygen species metabolism, immune and inflammatory response, regulation of angiogenesis, platelet degranulation, and other processes. The results expand the understanding of the molecular basis of diabetes and COVID-19 comorbidity.
Collapse
|
20
|
Wang R, Yang M, Jiang L, Huang M. Role of Angiopoietin-Tie axis in vascular and lymphatic systems and therapeutic interventions. Pharmacol Res 2022; 182:106331. [PMID: 35772646 DOI: 10.1016/j.phrs.2022.106331] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/11/2022] [Accepted: 06/24/2022] [Indexed: 12/29/2022]
Abstract
The Angiopoietin (Ang)-Tyrosine kinase with immunoglobulin-like and EGF-like domains (Tie) axis is an endothelial cell-specific ligand-receptor signaling pathway necessary for vascular and lymphatic development. The Ang-Tie axis is involved in regulating angiogenesis, vascular remodeling, vascular permeability, and inflammation to maintain vascular quiescence. Disruptions in the Ang-Tie axis are involved in many vascular and lymphatic system diseases and play an important role in physiological and pathological vascular conditions. Given recent advances in the Ang-Tie axis in the vascular and lymphatic systems, this review focuses on the multiple functions of the Ang-Tie axis in inflammation-induced vascular permeability, vascular remodeling, atherosclerosis, ocular angiogenesis, tumor angiogenesis, and metastasis. A summary of relevant therapeutic approaches to the Ang-Tie axis, including therapeutic antibodies, recombinant proteins and small molecule drugs are also discussed. The purpose of this review is to provide new hypotheses and identify potential therapeutic strategies based on the Ang-Tie signaling axis for the treatment of vascular and lymphatic-related diseases.
Collapse
Affiliation(s)
- Rui Wang
- College of Chemistry, Fuzhou University, Fuzhou 350116, Fujian, China
| | - Moua Yang
- Division of Hemostasis & Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA02215, United States
| | - Longguang Jiang
- College of Chemistry, Fuzhou University, Fuzhou 350116, Fujian, China.
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fuzhou 350116, Fujian, China.
| |
Collapse
|
21
|
Hultström M, Fromell K, Larsson A, Persson B, Nilsson B, Quaggin SE, Betsholtz C, Frithiof R, Lipcsey M, Jeansson M. Angiopoietin-2 Inhibition of Thrombomodulin-Mediated Anticoagulation-A Novel Mechanism That May Contribute to Hypercoagulation in Critically Ill COVID-19 Patients. Biomedicines 2022; 10:1333. [PMID: 35740360 PMCID: PMC9220312 DOI: 10.3390/biomedicines10061333] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/23/2022] [Accepted: 06/02/2022] [Indexed: 01/08/2023] Open
Abstract
Hypercoagulation and endothelial dysfunction play central roles in severe forms of COVID-19 infections, but the molecular mechanisms involved are unclear. Increased plasma levels of the inflammatory cytokine and TIE2 receptor antagonist Angiopoietin-2 were reported in severely ill COVID-19 patients. In vitro experiments suggest that Angiopoietin-2 bind and inhibits thrombomodulin. Thrombomodulin is expressed on the luminal surface of endothelial cells where it is an important member of the intrinsic anticoagulant pathway through activation of protein C. Using clinical data, mouse models, and in vitro assays, we tested if Angiopoietin-2 plays a causal role in COVID-19-associated hypercoagulation through direct inhibition of thrombin/thrombomodulin-mediated physiological anticoagulation. Angiopoietin-2 was measured in 61 patients at admission, and after 10 days in the 40 patients remaining in the ICU. We found that Angiopoietin-2 levels were increased in COVID-19 patients in correlation with disease severity, hypercoagulation, and mortality. In support of a direct effect of Angiopoietin-2 on coagulation, we found that injected Angiopoietin-2 in mice associated to thrombomodulin and resulted in a shortened tail bleeding time, decreased circulating levels of activated protein C, and increased plasma thrombin/antithrombin complexes. Conversely, bleeding time was increased in endothelial-specific Angiopoietin-2 knockout mice, while knockout of Tie2 had no effect on tail bleeding. Using in vitro assays, we found that Angiopoietin-2 inhibited thrombomodulin-mediated anticoagulation and protein C activation in human donor plasma. Our data suggest a novel in vivo mechanism for Angiopoietin-2 in COVID-19-associated hypercoagulation, implicating that Angiopoietin-2 inhibitors may be effective in the treatment of hypercoagulation in severe COVID-19 infection.
Collapse
Affiliation(s)
- Michael Hultström
- Anaesthesiology and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University, 751 85 Uppsala, Sweden; (M.H.); (R.F.); (M.L.)
- Integrative Physiology, Department of Medical Cell Biology, Uppsala University, 751 23 Uppsala, Sweden
| | - Karin Fromell
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (K.F.); (B.P.); (B.N.); (C.B.)
| | - Anders Larsson
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, 751 85 Uppsala, Sweden;
| | - Barbro Persson
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (K.F.); (B.P.); (B.N.); (C.B.)
| | - Bo Nilsson
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (K.F.); (B.P.); (B.N.); (C.B.)
| | - Susan E. Quaggin
- Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
- Division of Nephrology and Hypertension, Northwestern University, Chicago, IL 60611, USA
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (K.F.); (B.P.); (B.N.); (C.B.)
- Department of Medicine Huddinge, Karolinska Institutet, 141 52 Huddinge, Sweden
| | - Robert Frithiof
- Anaesthesiology and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University, 751 85 Uppsala, Sweden; (M.H.); (R.F.); (M.L.)
| | - Miklos Lipcsey
- Anaesthesiology and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University, 751 85 Uppsala, Sweden; (M.H.); (R.F.); (M.L.)
- Hedenstierna Laboratory, CIRRUS, Anaesthesiology and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University, 751 23 Uppsala, Sweden
| | - Marie Jeansson
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (K.F.); (B.P.); (B.N.); (C.B.)
- Department of Medicine Huddinge, Karolinska Institutet, 141 52 Huddinge, Sweden
| |
Collapse
|
22
|
Dynamics of circulating calprotectin accurately predict the outcome of moderate COVID-19 patients. EBioMedicine 2022; 80:104077. [PMID: 35644124 PMCID: PMC9132728 DOI: 10.1016/j.ebiom.2022.104077] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/26/2022] [Accepted: 05/10/2022] [Indexed: 12/11/2022] Open
|
23
|
Pischel L, Goshua G. Cost-effectiveness of Tocilizumab in Severe Coronavirus Disease 2019: to See or Not to See. Clin Infect Dis 2021; 73:2119-2120. [PMID: 33999992 PMCID: PMC8194546 DOI: 10.1093/cid/ciab459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Indexed: 11/25/2022] Open
Affiliation(s)
- Lauren Pischel
- Section of Infectious Diseases, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - George Goshua
- Section of Hematology, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Health Policy and Management, Harvard T.H. Chan School of Public Health Boston, Massachusetts, USA
| |
Collapse
|
24
|
Brunet-Ratnasingham E, Anand SP, Gantner P, Dyachenko A, Moquin-Beaudry G, Brassard N, Beaudoin-Bussières G, Pagliuzza A, Gasser R, Benlarbi M, Point F, Prévost J, Laumaea A, Niessl J, Nayrac M, Sannier G, Orban C, Messier-Peet M, Butler-Laporte G, Morrison DR, Zhou S, Nakanishi T, Boutin M, Descôteaux-Dinelle J, Gendron-Lepage G, Goyette G, Bourassa C, Medjahed H, Laurent L, Rébillard RM, Richard J, Dubé M, Fromentin R, Arbour N, Prat A, Larochelle C, Durand M, Richards JB, Chassé M, Tétreault M, Chomont N, Finzi A, Kaufmann DE. Integrated immunovirological profiling validates plasma SARS-CoV-2 RNA as an early predictor of COVID-19 mortality. SCIENCE ADVANCES 2021; 7:eabj5629. [PMID: 34826237 PMCID: PMC8626074 DOI: 10.1126/sciadv.abj5629] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Despite advances in COVID-19 management, identifying patients evolving toward death remains challenging. To identify early predictors of mortality within 60 days of symptom onset (DSO), we performed immunovirological assessments on plasma from 279 individuals. On samples collected at DSO11 in a discovery cohort, high severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral RNA (vRNA), low receptor binding domain–specific immunoglobulin G and antibody-dependent cellular cytotoxicity, and elevated cytokines and tissue injury markers were strongly associated with mortality, including in patients on mechanical ventilation. A three-variable model of vRNA, with predefined adjustment by age and sex, robustly identified patients with fatal outcome (adjusted hazard ratio for log-transformed vRNA = 3.5). This model remained robust in independent validation and confirmation cohorts. Since plasma vRNA’s predictive accuracy was maintained at earlier time points, its quantitation can help us understand disease heterogeneity and identify patients who may benefit from new therapies.
Collapse
Affiliation(s)
- Elsa Brunet-Ratnasingham
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
| | - Sai Priya Anand
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
| | - Pierre Gantner
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
| | - Alina Dyachenko
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Gaël Moquin-Beaudry
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Department of Neuroscience, Université de Montréal, Montréal, QC, Canada
| | - Nathalie Brassard
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Guillaume Beaudoin-Bussières
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
| | - Amélie Pagliuzza
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Romain Gasser
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Mehdi Benlarbi
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Floriane Point
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Jérémie Prévost
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
| | - Annemarie Laumaea
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Julia Niessl
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
| | - Manon Nayrac
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
| | - Gérémy Sannier
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
| | - Catherine Orban
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
- Centre hospitalier de l’Université de Montréal (CHUM), Montréal, QC, Canada
| | - Marc Messier-Peet
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Centre hospitalier de l’Université de Montréal (CHUM), Montréal, QC, Canada
| | - Guillaume Butler-Laporte
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, QC, Canada
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada
| | - David R. Morrison
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, QC, Canada
| | - Sirui Zhou
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, QC, Canada
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada
| | - Tomoko Nakanishi
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Kyoto-McGill International Collaborative School in Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Japan Society for the Promotion of Science, 5-3-1 Kojimachi, Chiyoda-ku, 102-0083 Tokyo, Japan
| | - Marianne Boutin
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
| | - Jade Descôteaux-Dinelle
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
| | - Gabrielle Gendron-Lepage
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Guillaume Goyette
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Catherine Bourassa
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Halima Medjahed
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Laetitia Laurent
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, QC, Canada
| | - Rose-Marie Rébillard
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Department of Neuroscience, Université de Montréal, Montréal, QC, Canada
| | - Jonathan Richard
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
| | - Mathieu Dubé
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Rémi Fromentin
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Nathalie Arbour
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Department of Neuroscience, Université de Montréal, Montréal, QC, Canada
| | - Alexandre Prat
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Department of Neuroscience, Université de Montréal, Montréal, QC, Canada
| | - Catherine Larochelle
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Department of Neuroscience, Université de Montréal, Montréal, QC, Canada
| | - Madeleine Durand
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Centre hospitalier de l’Université de Montréal (CHUM), Montréal, QC, Canada
| | - J. Brent Richards
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, QC, Canada
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Department of Twin Research, King’s College London, London, UK
| | - Michaël Chassé
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Centre hospitalier de l’Université de Montréal (CHUM), Montréal, QC, Canada
| | - Martine Tétreault
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Department of Neuroscience, Université de Montréal, Montréal, QC, Canada
| | - Nicolas Chomont
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
- Corresponding author. (N.C.); (A.F.); (D.E.K.)
| | - Andrés Finzi
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Corresponding author. (N.C.); (A.F.); (D.E.K.)
| | - Daniel E. Kaufmann
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
- Centre hospitalier de l’Université de Montréal (CHUM), Montréal, QC, Canada
- Département de Médecine, Université de Montréal, Montréal, QC, Canada
- Corresponding author. (N.C.); (A.F.); (D.E.K.)
| |
Collapse
|
25
|
Šikić J, Planinić Z, Matišić V, Friščić T, Molnar V, Jagačić D, Vujičić L, Tudorić N, Postružin Gršić L, Ljubičić Đ, Primorac D. COVID-19: The Impact on Cardiovascular System. Biomedicines 2021; 9:1691. [PMID: 34829920 PMCID: PMC8615470 DOI: 10.3390/biomedicines9111691] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 01/08/2023] Open
Abstract
SARS-CoV-2 has been circulating in population worldwide for the past year and a half, and thus a vast amount of scientific literature has been produced in order to study the biology of the virus and the pathophysiology of COVID-19, as well as to determine the best way to prevent infection, treat the patients and eliminate the virus. SARS-CoV-2 binding to the ACE2 receptor is the key initiator of COVID-19. The ability of SARS-CoV-2 to infect various types of cells requires special attention to be given to the cardiovascular system, as it is commonly affected. Thorough diagnostics and patient monitoring are beneficial in reducing the risk of cardiovascular morbidity and to ensure the most favorable outcomes for the infected patients, even after they are cured of the acute disease. The multidisciplinary nature of the fight against the COVID-19 pandemic requires careful consideration from the attending clinicians, in order to provide fast and reliable treatment to their patients in accordance with evidence-based medicine principles. In this narrative review, we reviewed the available literature on cardiovascular implications of COVID-19; both the acute and the chronic.
Collapse
Affiliation(s)
- Jozica Šikić
- Department of Cardiology, Clinical Hospital Sveti Duh, 10000 Zagreb, Croatia; (Z.P.); (T.F.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (V.M.); (D.J.); (L.V.); (N.T.); (Đ.L.); (D.P.)
| | - Zrinka Planinić
- Department of Cardiology, Clinical Hospital Sveti Duh, 10000 Zagreb, Croatia; (Z.P.); (T.F.)
| | - Vid Matišić
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (V.M.); (D.J.); (L.V.); (N.T.); (Đ.L.); (D.P.)
| | - Tea Friščić
- Department of Cardiology, Clinical Hospital Sveti Duh, 10000 Zagreb, Croatia; (Z.P.); (T.F.)
| | - Vilim Molnar
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (V.M.); (D.J.); (L.V.); (N.T.); (Đ.L.); (D.P.)
| | - Dorijan Jagačić
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (V.M.); (D.J.); (L.V.); (N.T.); (Đ.L.); (D.P.)
| | - Lovro Vujičić
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (V.M.); (D.J.); (L.V.); (N.T.); (Đ.L.); (D.P.)
| | - Neven Tudorić
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (V.M.); (D.J.); (L.V.); (N.T.); (Đ.L.); (D.P.)
| | | | - Đivo Ljubičić
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (V.M.); (D.J.); (L.V.); (N.T.); (Đ.L.); (D.P.)
- Clinical Hospital Dubrava, 10000 Zagreb, Croatia
| | - Dragan Primorac
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (V.M.); (D.J.); (L.V.); (N.T.); (Đ.L.); (D.P.)
- Eberly College of Science, The Pennsylvania State University, University Park, State College, PA 16802, USA
- The Henry C. Lee College of Criminal Justice and Forensic Sciences, University of New Haven, West Haven, CT 06516, USA
- Medical School, University of Split, 21000 Split, Croatia
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Medical School, University of Rijeka, 51000 Rijeka, Croatia
- Medical School REGIOMED, 96 450 Coburg, Germany
- Medical School, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW Coronavirus disease 2019 (COVID-19) is an infectious disease caused by severe acute respiratory syndrome coronavirus-2. Over the past year, COVID-19 has posed a significant threat to global health. Although the infection is associated with mild symptoms in many patients, a significant proportion of patients develop a prothrombotic state due to a combination of alterations in coagulation and immune cell function. The purpose of this review is to discuss the pathophysiological characteristics of COVID-19 that contribute to the immunothrombosis. RECENT FINDINGS Endotheliopathy during COVID-19 results in increased multimeric von Willebrand factor release and the potential for increased platelet adhesion to the endothelium. In addition, decreased anticoagulant proteins on the surface of endothelial cells further alters the hemostatic balance. Soluble coagulation markers are also markedly dysregulated, including plasminogen activator inhibitor-1 and tissue factor, leading to COVID-19 induced coagulopathy. Platelet hyperreactivity results in increased platelet-neutrophil and -monocyte aggregates further exacerbating the coagulopathy observed during COVID-19. Finally, the COVID-19-induced cytokine storm primes neutrophils to release neutrophil extracellular traps, which trap platelets and prothrombotic proteins contributing to pulmonary thrombotic complications. SUMMARY Immunothrombosis significantly contributes to the pathophysiology of COVID-19. Understanding the mechanisms behind COVID-19-induced coagulopathy will lead to future therapies for patients.
Collapse
Affiliation(s)
- Irina Portier
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, 84112
| | - Robert A. Campbell
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, 84112
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, 84132
| | - Frederik Denorme
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, 84112
| |
Collapse
|
27
|
Schmaier AA, Pajares Hurtado GM, Manickas-Hill ZJ, Sack KD, Chen SM, Bhambhani V, Quadir J, Nath AK, Collier ARY, Ngo D, Barouch DH, Shapiro NI, Gerszten RE, Yu XG, Peters KG, Flaumenhaft R, Parikh SM. Tie2 activation protects against prothrombotic endothelial dysfunction in COVID-19. JCI Insight 2021; 6:e151527. [PMID: 34506304 PMCID: PMC8564889 DOI: 10.1172/jci.insight.151527] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 09/09/2021] [Indexed: 12/27/2022] Open
Abstract
Endothelial dysfunction accompanies the microvascular thrombosis commonly observed in severe COVID-19. Constitutively, the endothelial surface is anticoagulant, a property maintained at least in part via signaling through the Tie2 receptor. During inflammation, the Tie2 antagonist angiopoietin-2 (Angpt-2) is released from endothelial cells and inhibits Tie2, promoting a prothrombotic phenotypic shift. We sought to assess whether severe COVID-19 is associated with procoagulant endothelial dysfunction and alterations in the Tie2/angiopoietin axis. Primary HUVECs treated with plasma from patients with severe COVID-19 upregulated the expression of thromboinflammatory genes, inhibited the expression of antithrombotic genes, and promoted coagulation on the endothelial surface. Pharmacologic activation of Tie2 with the small molecule AKB-9778 reversed the prothrombotic state induced by COVID-19 plasma in primary endothelial cells. Lung autopsies from patients with COVID-19 demonstrated a prothrombotic endothelial signature. Assessment of circulating endothelial markers in a cohort of 98 patients with mild, moderate, or severe COVID-19 revealed endothelial dysfunction indicative of a prothrombotic state. Angpt-2 concentrations rose with increasing disease severity, and the highest levels were associated with worse survival. These data highlight the disruption of Tie2/angiopoietin signaling and procoagulant changes in endothelial cells in severe COVID-19. Our findings provide rationale for current trials of Tie2-activating therapy with AKB-9778 in COVID-19.
Collapse
Affiliation(s)
- Alec A. Schmaier
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | | | | | - Kelsey D. Sack
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Siyu M. Chen
- Cardiovascular Research Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Victoria Bhambhani
- Cardiovascular Research Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Juweria Quadir
- Cardiovascular Research Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Anjali K. Nath
- Cardiovascular Research Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | | | - Debby Ngo
- Division of Pulmonary, Critical Care and Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Dan H. Barouch
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Center for Virology and Vaccine Research, and
| | - Nathan I. Shapiro
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Robert E. Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Xu G. Yu
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
- Infectious Diseases Division, Brigham and Women’s Hospital and Harvard Medical School, Massachusetts, Boston USA
| | - MGH COVID-19 Collection and Processing Team
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- The MGH COVID-19 Collection and Processing Team is detailed in Supplemental Acknowledgments
| | | | | | - Samir M. Parikh
- Division of Nephrology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
- Division of Nephrology, University of Texas Southwestern, Dallas, Texas, USA
| |
Collapse
|
28
|
Akwii RG, Mikelis CM. Targeting the Angiopoietin/Tie Pathway: Prospects for Treatment of Retinal and Respiratory Disorders. Drugs 2021; 81:1731-1749. [PMID: 34586603 PMCID: PMC8479497 DOI: 10.1007/s40265-021-01605-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2021] [Indexed: 12/21/2022]
Abstract
Anti-angiogenic approaches have significantly advanced the treatment of vascular-related pathologies. The ephemeral outcome and known side effects of the current vascular endothelial growth factor (VEGF)-based anti-angiogenic treatments have intensified research on other growth factors. The angiopoietin/Tie (Ang/Tie) family has an established role in vascular physiology and regulates angiogenesis, vascular permeability, and inflammatory responses. The Ang/Tie family consists of angiopoietins 1-4, their receptors, tie1 and 2 and the vascular endothelial-protein tyrosine phosphatase (VE-PTP). Modulation of Tie2 activation has provided a promising outcome in preclinical models and has led to clinical trials of Ang/Tie-targeting drug candidates for retinal disorders. Although less is known about the role of Ang/Tie in pulmonary disorders, several studies have revealed great potential of the Ang/Tie family members as drug targets for pulmonary vascular disorders as well. In this review, we summarize the functions of the Ang/Tie pathway in retinal and pulmonary vascular physiology and relevant disorders and highlight promising drug candidates targeting this pathway currently being or expected to be under clinical evaluation for retinal and pulmonary vascular disorders.
Collapse
Affiliation(s)
- Racheal Grace Akwii
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, 1406 S. Coulter St., Amarillo, TX, 79106, USA
| | - Constantinos M Mikelis
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, 1406 S. Coulter St., Amarillo, TX, 79106, USA.
| |
Collapse
|
29
|
Francischetti IM, Toomer K, Zhang Y, Jani J, Siddiqui Z, Brotman DJ, Hooper JE, Kickler TS. Upregulation of pulmonary tissue factor, loss of thrombomodulin and immunothrombosis in SARS-CoV-2 infection. EClinicalMedicine 2021; 39:101069. [PMID: 34377969 PMCID: PMC8342934 DOI: 10.1016/j.eclinm.2021.101069] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND SARS-CoV-2 infection is associated with thrombotic and microvascular complications. The cause of coagulopathy in the disease is incompletely understood. METHODS A single-center cross-sectional study including 66 adult COVID-19 patients (40 moderate, 26 severe disease), and 9 controls, performed between 04/2020 and 10/2020. Markers of coagulation, endothelial cell function [angiopoietin-1,-2, P-selectin, von Willebrand Factor Antigen (WF:Ag), von Willebrand Factor Ristocetin Cofactor, ADAMTS13, thrombomodulin, soluble Endothelial cell Protein C Receptor (sEPCR), Tissue Factor Pathway Inhibitor], neutrophil activation (elastase, citrullinated histones) and fibrinolysis (tissue-type plasminogen activator, plasminogen activator inhibitor-1) were evaluated using ELISA. Tissue Factor (TF) was estimated by antithrombin-FVIIa complex (AT/FVIIa) and microparticles-TF (MP-TF). We correlated each marker and determined its association with severity. Expression of pulmonary TF, thrombomodulin and EPCR was determined by immunohistochemistry in 9 autopsies. FINDINGS Comorbidities were frequent in both groups, with older age associated with severe disease. All patients were on prophylactic anticoagulants. Three patients (4.5%) developed pulmonary embolism. Mortality was 7.5%. Patients presented with mild alterations in the coagulogram (compensated state). Biomarkers of endothelial cell, neutrophil activation and fibrinolysis were elevated in severe vs moderate disease; AT/FVIIa and MP-TF levels were higher in severe patients. Logistic regression revealed an association of D-dimers, angiopoietin-1, vWF:Ag, thrombomodulin, white blood cells, absolute neutrophil count (ANC) and hemoglobin levels with severity, with ANC and vWF:Ag identified as independent factors. Notably, postmortem specimens demonstrated epithelial expression of TF in the lung of fatal COVID-19 cases with loss of thrombomodulin staining, implying in a shift towards a procoagulant state. INTERPRETATION Coagulation dysregulation has multifactorial etiology in SARS-Cov-2 infection. Upregulation of pulmonary TF with loss of thrombomodulin emerge as a potential link to immunothrombosis, and therapeutic targets in the disease. FUNDING John Hopkins University School of Medicine.
Collapse
Key Words
- ADAMTS13, a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13
- ALC, absolute lymphocyte count
- ALI, Acute Lung Injury
- AMC, absolute monocyte count
- ANC, absolute neutrophil count
- AT/VIIa, antithrombin-FVIIa complex
- Coagulation
- ELISA, enzyme-linked immunosorbent assay
- Hb, hemoglobin
- Hemostasis
- ICU, intensive care unit
- Ixolaris
- LMWH, low molecular weight heparin
- MP-TF, Microparticles-Tissue Factor
- PAI-1, plasminogen activator inhibitor-1
- PAR, protease-activated receptor
- TF, Tissue Factor
- TFPI, Tissue Factor Pathway Inhibitor
- Thrombosis
- WBC, white blood cells
- sEPCR, soluble Endothelial cell Protein C Receptor
- t-PA, tissue-type plasminogen activator
- vWF, von Willebrand Factor
- vWF:Ag, von Willebrand Factor Antigen
- vWF:RCo, von Willebrand Factor Ristocetin Cofactor
Collapse
Affiliation(s)
- Ivo M.B. Francischetti
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Corresponding author.
| | - Kevin Toomer
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yifan Zhang
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Jayesh Jani
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Zishan Siddiqui
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Daniel J. Brotman
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jody E. Hooper
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Thomas S. Kickler
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
30
|
Mobayen G, Dhutia A, Clarke C, Prendecki M, McAdoo S, Keniyopoullos R, Malik T, Laffan M, Willicombe M, McKinnon T. Severe COVID-19 is associated with endothelial activation and abnormal glycosylation of von Willebrand factor in patients undergoing hemodialysis. Res Pract Thromb Haemost 2021; 5:e12582. [PMID: 34532629 PMCID: PMC8435526 DOI: 10.1002/rth2.12582] [Citation(s) in RCA: 10] [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: 05/08/2021] [Revised: 07/19/2021] [Accepted: 07/27/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND A major clinical feature of severe coronavirus diease 2019 (COVID-19) is microvascular thrombosis linked to endothelial cell activation. Consistent with this, a number of studies have shown that patients with severe COVID-19 have highly elevated plasma levels of von Willebrand Factor (VWF) that may contribute to the prothrombotic phenotype. In the current study, we investigated the extent of endothelial activation in patients receiving hemodialysis who had either mild or severe COVID-19. METHODS Plasma VWF, ADAMTS-13, angiopoietin-2 (Ang2), and syndecan-1 levels were determined by ELISA. The sialic acid content of VWF was investigated using a modified ELISA to measure elderberry bark lectin, specific for sialic acid residues, binding to VWF. RESULTS Patients receiving hemodialysis with severe COVID-19 had significantly higher plasma levels of VWF and lower ADAMTS-13. VWF levels peaked and were sustained during the first 10 days after positive confirmation of infection. While Ang2 trended toward being higher in severely ill patients, this did not reach significance; however, severely ill patients had significantly higher soluble syndecan-1 levels, with high levels related to risk of death. Finally, higher VWF levels in severely ill patients were correlated with lower VWF sialic acid content. CONCLUSIONS Severe COVID-19 in patients undergoing hemodialysis is associated with both acute and sustained activation of the endothelium, leading to alteration of the VWF/ADAMTS-13 axis. Lower VWF sialic acid content represents altered VWF processing and further confirms the disturbance caused to the endothelium in COVID-19.
Collapse
Affiliation(s)
- Golzar Mobayen
- Department of Immunology and InflammationCentre for HaematologyCommonwealth BuildingHammersmith CampusImperial College of Science Technology and MedicineLondonUK
| | - Amrita Dhutia
- Department of Immunology and InflammationCentre for Inflammatory DiseaseCommonwealth BuildingHammersmith CampusImperial College of Science Technology and MedicineLondonUK
| | - Candice Clarke
- Department of Immunology and InflammationCentre for Inflammatory DiseaseCommonwealth BuildingHammersmith CampusImperial College of Science Technology and MedicineLondonUK
| | - Maria Prendecki
- Department of Immunology and InflammationCentre for Inflammatory DiseaseCommonwealth BuildingHammersmith CampusImperial College of Science Technology and MedicineLondonUK
| | - Stephen McAdoo
- Department of Immunology and InflammationCentre for Inflammatory DiseaseCommonwealth BuildingHammersmith CampusImperial College of Science Technology and MedicineLondonUK
| | - Renos Keniyopoullos
- Department of Immunology and InflammationCentre for HaematologyCommonwealth BuildingHammersmith CampusImperial College of Science Technology and MedicineLondonUK
| | - Talat Malik
- Department of Immunology and InflammationCentre for Inflammatory DiseaseCommonwealth BuildingHammersmith CampusImperial College of Science Technology and MedicineLondonUK
| | - Michael Laffan
- Department of Immunology and InflammationCentre for HaematologyCommonwealth BuildingHammersmith CampusImperial College of Science Technology and MedicineLondonUK
| | - Michelle Willicombe
- Department of Immunology and InflammationCentre for Inflammatory DiseaseCommonwealth BuildingHammersmith CampusImperial College of Science Technology and MedicineLondonUK
| | - Thomas McKinnon
- Department of Immunology and InflammationCentre for HaematologyCommonwealth BuildingHammersmith CampusImperial College of Science Technology and MedicineLondonUK
| |
Collapse
|
31
|
Diagnostic Significance of Serum Galectin-3 in Hospitalized Patients with COVID-19-A Preliminary Study. Biomolecules 2021; 11:biom11081136. [PMID: 34439802 PMCID: PMC8393726 DOI: 10.3390/biom11081136] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/15/2021] [Accepted: 07/25/2021] [Indexed: 12/26/2022] Open
Abstract
Severe coronavirus disease 2019 (COVID-19) is associated with hyperinflammation leading to organ injury, including respiratory failure. Galectin-3 was implicated in innate immunological response to infections and in chronic fibrosis. The aim of our preliminary study was the assessment of the diagnostic utility of serum galectin-3 in patients with COVID-19. The prospective observational study included adult patients admitted with active COVID-19 and treated in tertiary hospital between June and July 2020. The diagnosis was confirmed by the quantitative detection of nucleic acid of severe acute respiratory syndrome coronavirus 2 in nasopharyngeal swabs. Galectin-3 was measured by enzyme immunoassay in serum samples obtained during the first five days of hospital stay. We included 70 patients aged 25 to 73 years; 90% had at least one comorbidity. During the hospital stay, 32.9% were diagnosed with COVID-19 pneumonia and 12.9% required treatment in the intensive care unit (ICU). Serum galectin-3 was significantly increased in patients who developed pneumonia, particularly those who required ICU admission. Positive correlations were found between galectin-3 and inflammatory markers (interleukin-6, C-reactive protein, ferritin, pentraxin-3), a marker of endothelial injury (soluble fms-like tyrosine kinase-1), and a range of tissue injury markers. Serum galectin-3 enabled the diagnosis of pneumonia with moderate diagnostic accuracy and the need for ICU treatment with high diagnostic accuracy. Our findings strengthen the hypothesis that galectin-3 may be involved in severe COVID-19. Further studies are planned to confirm the preliminary results and to verify possible associations of galectin-3 with long-term consequences of COVID-19, including pulmonary fibrosis.
Collapse
|
32
|
Aman J, Duijvelaar E, Botros L, Kianzad A, Schippers JR, Smeele PJ, Azhang S, Bartelink IH, Bayoumy AA, Bet PM, Boersma W, Bonta PI, Boomars KAT, Bos LDJ, van Bragt JJMH, Braunstahl GJ, Celant LR, Eger KAB, Geelhoed JJM, van Glabbeek YLE, Grotjohan HP, Hagens LA, Happe CM, Hazes BD, Heunks LMA, van den Heuvel M, Hoefsloot W, Hoek RJA, Hoekstra R, Hofstee HMA, Juffermans NP, Kemper EM, Kos R, Kunst PWA, Lammers A, van der Lee I, van der Lee EL, Maitland-van der Zee AH, Mau Asam PFM, Mieras A, Muller M, Neefjes L, Nossent EJ, Oswald LMA, Overbeek MJ, Pamplona C, Paternotte N, Pronk N, de Raaf MA, van Raaij BFM, Reijrink M, Schultz MJ, Serpa Neto A, Slob EM, Smeenk FWJM, Smit MR, Smits AJ, Stalenhoef JE, Tuinman PR, Vanhove ALEM, Wessels JN, van Wezenbeek JCC, Vonk Noordegraaf A, de Man FS, Bogaard HJ. Imatinib in patients with severe COVID-19: a randomised, double-blind, placebo-controlled, clinical trial. THE LANCET RESPIRATORY MEDICINE 2021; 9:957-968. [PMID: 34147142 PMCID: PMC8232929 DOI: 10.1016/s2213-2600(21)00237-x] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/12/2021] [Accepted: 05/14/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND The major complication of COVID-19 is hypoxaemic respiratory failure from capillary leak and alveolar oedema. Experimental and early clinical data suggest that the tyrosine-kinase inhibitor imatinib reverses pulmonary capillary leak. METHODS This randomised, double-blind, placebo-controlled, clinical trial was done at 13 academic and non-academic teaching hospitals in the Netherlands. Hospitalised patients (aged ≥18 years) with COVID-19, as confirmed by an RT-PCR test for SARS-CoV-2, requiring supplemental oxygen to maintain a peripheral oxygen saturation of greater than 94% were eligible. Patients were excluded if they had severe pre-existing pulmonary disease, had pre-existing heart failure, had undergone active treatment of a haematological or non-haematological malignancy in the previous 12 months, had cytopenia, or were receiving concomitant treatment with medication known to strongly interact with imatinib. Patients were randomly assigned (1:1) to receive either oral imatinib, given as a loading dose of 800 mg on day 0 followed by 400 mg daily on days 1-9, or placebo. Randomisation was done with a computer-based clinical data management platform with variable block sizes (containing two, four, or six patients), stratified by study site. The primary outcome was time to discontinuation of mechanical ventilation and supplemental oxygen for more than 48 consecutive hours, while being alive during a 28-day period. Secondary outcomes included safety, mortality at 28 days, and the need for invasive mechanical ventilation. All efficacy and safety analyses were done in all randomised patients who had received at least one dose of study medication (modified intention-to-treat population). This study is registered with the EU Clinical Trials Register (EudraCT 2020-001236-10). FINDINGS Between March 31, 2020, and Jan 4, 2021, 805 patients were screened, of whom 400 were eligible and randomly assigned to the imatinib group (n=204) or the placebo group (n=196). A total of 385 (96%) patients (median age 64 years [IQR 56-73]) received at least one dose of study medication and were included in the modified intention-to-treat population. Time to discontinuation of ventilation and supplemental oxygen for more than 48 h was not significantly different between the two groups (unadjusted hazard ratio [HR] 0·95 [95% CI 0·76-1·20]). At day 28, 15 (8%) of 197 patients had died in the imatinib group compared with 27 (14%) of 188 patients in the placebo group (unadjusted HR 0·51 [0·27-0·95]). After adjusting for baseline imbalances between the two groups (sex, obesity, diabetes, and cardiovascular disease) the HR for mortality was 0·52 (95% CI 0·26-1·05). The HR for mechanical ventilation in the imatinib group compared with the placebo group was 1·07 (0·63-1·80; p=0·81). The median duration of invasive mechanical ventilation was 7 days (IQR 3-13) in the imatinib group compared with 12 days (6-20) in the placebo group (p=0·0080). 91 (46%) of 197 patients in the imatinib group and 82 (44%) of 188 patients in the placebo group had at least one grade 3 or higher adverse event. The safety evaluation revealed no imatinib-associated adverse events. INTERPRETATION The study failed to meet its primary outcome, as imatinib did not reduce the time to discontinuation of ventilation and supplemental oxygen for more than 48 consecutive hours in patients with COVID-19 requiring supplemental oxygen. The observed effects on survival (although attenuated after adjustment for baseline imbalances) and duration of mechanical ventilation suggest that imatinib might confer clinical benefit in hospitalised patients with COVID-19, but further studies are required to validate these findings. FUNDING Amsterdam Medical Center Foundation, Nederlandse Organisatie voor Wetenschappelijk Onderzoek/ZonMW, and the European Union Innovative Medicines Initiative 2.
Collapse
Affiliation(s)
- Jurjan Aman
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Erik Duijvelaar
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Liza Botros
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Azar Kianzad
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Job R Schippers
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Patrick J Smeele
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Sara Azhang
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands; Department of Pulmonology, Haaglanden Medisch Centrum, The Hague, Netherlands
| | - Imke H Bartelink
- Department of Pharmacy, Amsterdam UMC, VUMC, Amsterdam, Netherlands
| | - Ahmed A Bayoumy
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands; Department of Pulmonology, Chest Unit, Suez Canal University, Suez, Egypt
| | - Pierre M Bet
- Department of Pharmacy, Amsterdam UMC, VUMC, Amsterdam, Netherlands
| | - Wim Boersma
- Department of Pulmonology, Noordwest Ziekenhuisgroep, Alkmaar, Netherlands
| | - Peter I Bonta
- Department of Respiratory Medicine, Amsterdam UMC, AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Karin A T Boomars
- Department of Pulmonology, Erasmus Medisch Centrum, Rotterdam, Netherlands
| | - Lieuwe D J Bos
- Department of Respiratory Medicine, Amsterdam UMC, AMC, University of Amsterdam, Amsterdam, Netherlands; Department of Intensive Care, Amsterdam UMC, AMC, Amsterdam, Netherlands
| | - Job J M H van Bragt
- Department of Respiratory Medicine, Amsterdam UMC, AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Gert-Jan Braunstahl
- Department of Pulmonology, Sint Franciscus Ziekenhuis, Rotterdam, Netherlands
| | - Lucas R Celant
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Katrien A B Eger
- Department of Respiratory Medicine, Amsterdam UMC, AMC, University of Amsterdam, Amsterdam, Netherlands
| | | | - Yurika L E van Glabbeek
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Hans P Grotjohan
- Department of Pulmonology, Isala Ziekenhuizen, Zwolle, Netherlands
| | - Laura A Hagens
- Department of Intensive Care, Amsterdam UMC, AMC, Amsterdam, Netherlands
| | - Chris M Happe
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Boaz D Hazes
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Leo M A Heunks
- Department of Intensive Care, Amsterdam UMC, VUMC, Amsterdam, Netherlands
| | | | | | - Rianne J A Hoek
- Department of Pharmacy, Amsterdam UMC, VUMC, Amsterdam, Netherlands
| | - Romke Hoekstra
- Department of Pulmonology, Antonius Ziekenhuis, Sneek, Netherlands
| | - Herman M A Hofstee
- Department of Internal Medicine, Haaglanden Medisch Centrum, The Hague, Netherlands
| | - Nicole P Juffermans
- Department of Intensive Care, Amsterdam UMC, AMC, Amsterdam, Netherlands; Department of Intensive Care, Onze Lieve Vrouwe Gasthuis, Amsterdam, Netherlands
| | | | - Renate Kos
- Department of Respiratory Medicine, Amsterdam UMC, AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Peter W A Kunst
- Department of Pulmonology, Onze Lieve Vrouwe Gasthuis, Amsterdam, Netherlands
| | - Ariana Lammers
- Department of Respiratory Medicine, Amsterdam UMC, AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Ivo van der Lee
- Department of Pulmonology, Spaarne Gasthuis, Haarlem, Netherlands
| | - E Laurien van der Lee
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | | | - Pearl F M Mau Asam
- Department of Respiratory Medicine, Amsterdam UMC, AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Adinda Mieras
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Mirte Muller
- Department of Pulmonology, Catharina Ziekenhuis, Eindhoven, Netherlands
| | - Liesbeth Neefjes
- Department of Pulmonology, Catharina Ziekenhuis, Eindhoven, Netherlands
| | - Esther J Nossent
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Laurien M A Oswald
- Department of Pulmonology, Sint Franciscus Ziekenhuis, Rotterdam, Netherlands
| | - Maria J Overbeek
- Department of Pulmonology, Haaglanden Medisch Centrum, The Hague, Netherlands
| | - Carolina Pamplona
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Nienke Paternotte
- Department of Pulmonology, Noordwest Ziekenhuisgroep, Alkmaar, Netherlands
| | - Niels Pronk
- Department of Pulmonology, Gelre Ziekenhuis, Apeldoorn, Netherlands
| | - Michiel A de Raaf
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Bas F M van Raaij
- Department of Pulmonology Leiden University Medical Center, Leiden, Netherlands
| | - Merlijn Reijrink
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Marcus J Schultz
- Department of Intensive Care, Amsterdam UMC, AMC, Amsterdam, Netherlands
| | - Ary Serpa Neto
- Department of Critical Care Medicine and Institute of Education and Research, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Elise M Slob
- Department of Respiratory Medicine, Amsterdam UMC, AMC, University of Amsterdam, Amsterdam, Netherlands
| | | | - Marry R Smit
- Department of Intensive Care, Amsterdam UMC, AMC, Amsterdam, Netherlands
| | - A Josien Smits
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Janneke E Stalenhoef
- Department of Internal Medicine, Onze Lieve Vrouwe Gasthuis, Amsterdam, Netherlands
| | - Pieter R Tuinman
- Department of Intensive Care, Amsterdam UMC, VUMC, Amsterdam, Netherlands
| | - Arthur L E M Vanhove
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Jessie N Wessels
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Jessie C C van Wezenbeek
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Anton Vonk Noordegraaf
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Frances S de Man
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Harm J Bogaard
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands.
| |
Collapse
|
33
|
Schmaier AA, Hurtado GP, Manickas-Hill ZJ, Sack KD, Chen SM, Bhambhani V, Quadir J, Nath AK, Collier ARY, Ngo D, Barouch DH, Gerszten RE, Yu XG, Peters K, Flaumenhaft R, Parikh SM. Tie2 activation protects against prothrombotic endothelial dysfunction in COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021. [PMID: 34031665 DOI: 10.1101/2021.05.13.21257070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Profound endothelial dysfunction accompanies the microvascular thrombosis commonly observed in severe COVID-19. In the quiescent state, the endothelial surface is anticoagulant, a property maintained at least in part via constitutive signaling through the Tie2 receptor. During inflammation, the Tie2 antagonist angiopoietin-2 (Angpt-2) is released from activated endothelial cells and inhibits Tie2, promoting a prothrombotic phenotypic shift. We sought to assess whether severe COVID-19 is associated with procoagulant dysfunction of the endothelium and alterations in the Tie2-angiopoietin axis. Primary human endothelial cells treated with plasma from patients with severe COVID-19 upregulated the expression of thromboinflammatory genes, inhibited expression of antithrombotic genes, and promoted coagulation on the endothelial surface. Pharmacologic activation of Tie2 with the small molecule AKB-9778 reversed the prothrombotic state induced by COVID-19 plasma in primary endothelial cells. On lung autopsy specimens from COVID-19 patients, we found a prothrombotic endothelial signature as evidenced by increased von Willebrand Factor and loss of anticoagulant proteins. Assessment of circulating endothelial markers in a cohort of 98 patients with mild, moderate, or severe COVID-19 revealed profound endothelial dysfunction indicative of a prothrombotic state. Angpt-2 concentrations rose with increasing disease severity and highest levels were associated with worse survival. These data highlight the disruption of Tie2-angiopoietin signaling and procoagulant changes in endothelial cells in severe COVID-19. Moreover, our findings provide novel rationale for current trials of Tie2 activating therapy with AKB-9778 in severe COVID-19 disease.
Collapse
|
34
|
Melegari G, Rivi V, Zelent G, Nasillo V, De Santis E, Melegari A, Bevilacqua C, Zoli M, Meletti S, Barbieri A. Mild to Severe Neurological Manifestations of COVID-19: Cases Reports. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18073673. [PMID: 33915937 PMCID: PMC8036948 DOI: 10.3390/ijerph18073673] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/28/2021] [Accepted: 03/29/2021] [Indexed: 02/07/2023]
Abstract
The main focus of Coronavirus disease 2019 (COVID-19) infection is pulmonary complications through virus-related neurological manifestations, ranging from mild to severe, such as encephalitis, cerebral thrombosis, neurocognitive (dementia-like) syndrome, and delirium. The hospital screening procedures for quickly recognizing neurological manifestations of COVID-19 are often complicated by other coexisting symptoms and can be obscured by the deep sedation procedures required for critically ill patients. Here, we present two different case-reports of COVID-19 patients, describing neurological complications, diagnostic imaging such as olfactory bulb damage (a mild and unclear underestimated complication) and a severe and sudden thrombotic stroke complicated with hemorrhage with a low-level cytokine storm and respiratory symptom resolution. We discuss the possible mechanisms of virus entrance, together with the causes of COVID-19-related encephalitis, olfactory bulb damage, ischemic stroke, and intracranial hemorrhage.
Collapse
Affiliation(s)
- Gabriele Melegari
- Anaesthesia and Intensive Care, Azienda Ospedaliero-Universitaria di Modena, 41125 Modena, Italy
- Correspondence: ; Tel.: +39-0593961536
| | - Veronica Rivi
- Department of Biomedical, Metabolic and Neural Sciences, Neuroscience Post Graduate School, University of Modena and Reggio Emilia, 41125 Modena, Italy; (V.R.); (M.Z.)
| | - Gabriele Zelent
- Neuroradiology, Azienda Ospedaliero-Universitaria di Modena, 41125 Modena, Italy;
| | - Vincenzo Nasillo
- Department of Laboratory Medicine, Azienda Unità Sanitaria Locale, 41125 Modena, Italy; (V.N.); (E.D.S.); (A.M.)
| | - Elena De Santis
- Department of Laboratory Medicine, Azienda Unità Sanitaria Locale, 41125 Modena, Italy; (V.N.); (E.D.S.); (A.M.)
| | - Alessandra Melegari
- Department of Laboratory Medicine, Azienda Unità Sanitaria Locale, 41125 Modena, Italy; (V.N.); (E.D.S.); (A.M.)
| | - Claudia Bevilacqua
- School of Anaesthesia and Intensive Care, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.B.); (A.B.)
| | - Michele Zoli
- Department of Biomedical, Metabolic and Neural Sciences, Neuroscience Post Graduate School, University of Modena and Reggio Emilia, 41125 Modena, Italy; (V.R.); (M.Z.)
| | - Stefano Meletti
- Neurology, Azienda Ospedaliera Universitaria di Modena, 41125 Modena, Italy;
| | - Alberto Barbieri
- School of Anaesthesia and Intensive Care, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.B.); (A.B.)
| |
Collapse
|