1
|
Milacek C, Stefan AN, Bal C, Geist M, Guttmann C, Idzko M, Antoniewicz L. Nebulized alteplase in coronavirus disease 2019 pneumonia: a case series. J Med Case Rep 2024; 18:572. [PMID: 39604994 PMCID: PMC11603999 DOI: 10.1186/s13256-024-04924-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Many patients with severe coronavirus disease 2019 pneumonia exhibit signs of microthrombosis. Previous studies discussed intravenous fibrinolytic agents as potential add-on therapy in these patients. Therefore, we propose the inhalative administration of fibrinolytics as a possible safer alternative. CASE PRESENTATION This case series describes five white male patients, aged 51-78 years, treated with off-label inhalation of alteplase between November and December 2020. All patients suffered from severe severe acute respiratory syndrome coronavirus 2 infection with respiratory failure. Pulmonary embolism was ruled out by pulmonary angiogram in computed tomography scans, and all patients showed signs of coronavirus disease 2019 pneumonia. Four patients improved clinically, while one patient with advanced chronic diseases died due to multiple organ failure. No directly associated adverse effects were observed following inhalation of alteplase. CONCLUSION This case series warrants further attention to investigate inhalative alteplase as an additional treatment in patients with severe coronavirus disease 2019 infection.
Collapse
Affiliation(s)
- Christopher Milacek
- Clinical Division of Pulmonology, Department of Medicine II, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Andreea Nicoleta Stefan
- Clinical Division of Pulmonology, Department of Medicine II, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Christina Bal
- Clinical Division of Pulmonology, Department of Medicine II, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Matthias Geist
- Clinical Division of Pulmonology, Department of Medicine II, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Claudia Guttmann
- Clinical Division of Pulmonology, Department of Medicine II, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Marco Idzko
- Clinical Division of Pulmonology, Department of Medicine II, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Lukasz Antoniewicz
- Clinical Division of Pulmonology, Department of Medicine II, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
2
|
Xiao LX, Zhu DL, Chen J, Lv J, Liu MJ, Dai X, Wang DX, Deng W. Exploring the therapeutic role of early heparin administration in ARDS management: a MIMIC-IV database analysis. J Intensive Care 2024; 12:9. [PMID: 38409068 PMCID: PMC10895755 DOI: 10.1186/s40560-024-00723-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/18/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is a severe respiratory condition characterized by a high mortality rate, the management of which relies on supportive care and a profound understanding of its pathophysiology. Heparin, with its anticoagulant and potential anti-inflammatory properties, offers a new therapeutic opportunity for the treatment of ARDS. METHODS In this retrospective cohort study, we examined the MIMIC-IV database for ARDS patients who received prophylactic heparin within the first 72 h of ICU admission. Employing propensity score matching and inverse probability weighting (IPW) analysis, we evaluated the impact of early heparin use on patient outcomes, focusing on mortality rates. RESULTS Patients who received prophylactic heparin had a significantly lower in-hospital mortality rate compared to those who did not (13.55% vs 17.93%, HR = 0.71, 95% CI: 0.54-0.93, P = 0.012). This result remained significant after propensity score matching (12.75% vs 17.93%, HR = 0.65, 95% CI 0.47-0.90, P = 0.010). Analysis using five different statistical models indicated that early use of heparin significantly reduced the in-hospital mortality rate, with HR = 0.669 (95% CI 0.487-0.919, P = 0.013) in the doubly robust model without balanced covariates; HR = 0.705 (95% CI 0.515-0.965, P = 0.029) with all covariates considered; HR = 0.660 (95% CI 0.491-0.888, P = 0.006) in the propensity score (IPW) model; HR = 0.650 (95% CI 0.470-0.900, P = 0.010) in the propensity score matching model; and HR = 0.706 (95% CI 0.536-0.930, P = 0.013) in the multivariate Cox regression model. Secondary outcomes indicated that heparin use was also associated with reduced mortality rates at 60 days, and 90 days. CONCLUSION This research highlights that early prophylactic administration of heparin may substantially lower mortality in ARDS patients. These findings underscore the potential of heparin as a key component in the management of ARDS, offering a new perspective and novel strategies for clinical treatment.
Collapse
Affiliation(s)
- Ling-Xi Xiao
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - De Liang Zhu
- Department of Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, No. 126, Xiantai Street, Changchun, 130033, China
| | - Juan Chen
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Jing Lv
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Mei-Jun Liu
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Xue Dai
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Dao-Xin Wang
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Wang Deng
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| |
Collapse
|
3
|
Li Q, Xiao C, Gu J, Chen X, Yuan J, Li S, Li W, Gao D, Li L, Liu Y, Shen F. 6-Gingerol ameliorates alveolar hypercoagulation and fibrinolytic inhibition in LPS-provoked ARDS via RUNX1/NF-κB signaling pathway. Int Immunopharmacol 2024; 128:111459. [PMID: 38181675 DOI: 10.1016/j.intimp.2023.111459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/07/2023] [Accepted: 12/25/2023] [Indexed: 01/07/2024]
Abstract
BACKGROUND Alveolar hypercoagulation and fibrinolytic inhibition play a central role in refractory hypoxemia in acute respiratory distress syndrome (ARDS), but it lacks effective drugs for prevention and treatment of this pathophysiology. Our previous experiment confirmed that RUNX1 promoted alveolar hypercoagulation and fibrinolytic inhibition through NF-κB pathway. Other studies demonstrated that 6-gingerol regulated inflammation and metabolism by inhibiting the NF-κB signaling pathway. We assume that 6-gingerol would ameliorate alveolar hypercoagulation and fibrinolytic inhibition via RUNX1/ NF-κB pathway in LPS-induced ARDS. METHODS Rat ARDS model was replicated through LPS inhalation. Before LPS inhalation, the rats were intraperitoneally treated with different doses of 6-gingerol or the same volume of normal saline (NS) for 12 h, and then intratracheal inhalation of LPS for 24 h. In cell experiment, alveolar epithelial cell type II (AECII) was treated with 6-gingerol for 6 h and then with LPS for another 24 h. RUNX1 gene was down-regulated both in pulmonary tissue and in cells. Tissue factor (TF), plasminogen Activator Inhibitor 1(PAI-1) and thrombin were determined by Wester-blot (WB), qPCR or by enzyme-linked immunosorbent (ELISA). Lung injury score, pulmonary edema and pulmonary collagen III in rat were assessed. NF-κB pathway were also observed in vivo and in vitro. The direct binding capability of 6-gingerol to RUNX1 was confirmed by using Drug Affinity Responsive Target Stability test (DARTS). RESULTS 6-gingerol dose-dependently attenuated LPS-induced lung injury and pulmonary edema. LPS administration caused excessive TF and PAI-1 expression both in pulmonary tissue and in AECII cell and a large amount of TF, PAI-1 and thrombin in bronchial alveolar lavage fluid (BALF), which all were effectively decreased by 6-gingerol treatment in a dose-dependent manner. The high collagen Ⅲ level in lung tissue provoked by LPS was significantly abated by 6-gingerol. 6-gingerol was seen to dramatically inhibit the LPS-stimulated activation of NF-κB pathway, indicated by decreases of p-p65/total p65, p-IKKβ/total IKKβ, and also to suppress the RUNX1 expression. RUNX1 gene knock down or RUNX1 inhibitor Ro5-3335 significantly enhanced the efficacies of 6-gingerol in vivo and in vitro, but RUNX1 over expression remarkably impaired the effects of 6-gingerol on TF, PAI-1 and on NF-κB pathway. DARTS result showed that 6-gingerol directly bond to RUNX1 molecules. CONCLUSIONS Our experimental data demonstrated that 6-gingerol ameliorates alveolar hypercoagulation and fibrinolytic inhibition via RUNX1/NF-κB pathway in LPS-induced ARDS. 6-gingerol is expected to be an effective drug in ARDS.
Collapse
Affiliation(s)
- Qing Li
- Department of Intensive Care Unit, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Chuan Xiao
- Department of Intensive Care Unit, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - JiaRun Gu
- Emergency department, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Xianjun Chen
- Department of Intensive Care Unit, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Jia Yuan
- Department of Intensive Care Unit, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Shuwen Li
- Department of Intensive Care Unit, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Wei Li
- Department of Intensive Care Unit, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Daixiu Gao
- Department of Intensive Care Unit, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Lu Li
- Department of Intensive Care Unit, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Ying Liu
- Department of Intensive Care Unit, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Feng Shen
- Department of Intensive Care Unit, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| |
Collapse
|
4
|
Chowdary P, Agarwal B, Peralta MR, Bhagani S, Lee S, Goldring J, Lipman M, Waqif E, Phillips M, Philippou H, Foley JH, Mutch NJ, Ariëns RAS, Stringer KA, Ricciardi F, Watissée M, Hughes D, Nathwani A, Riddell A, Patch D, Buckley J, De Neef M, Dimber R, Diaz-Garcia C, Patel H, Nandani A, Dissanayake U, Chadwick N, Alkhatip AAAMM, Watkinson P, Raith E, Singh S, Wolff T, Jha R, Brill SE, Bakhai A, Evans A, Gilani F, Gomez K. Nebulized Recombinant Tissue Plasminogen Activator (rt-PA) for Acute COVID-19-Induced Respiratory Failure: An Exploratory Proof-of-Concept Trial. J Clin Med 2023; 12:5848. [PMID: 37762789 PMCID: PMC10531875 DOI: 10.3390/jcm12185848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Acute lung injury in COVID-19 results in diffuse alveolar damage with disruption of the alveolar-capillary barrier, coagulation activation, alveolar fibrin deposition and pulmonary capillary thrombi. Nebulized recombinant tissue plasminogen activator (rt-PA) has the potential to facilitate localized thrombolysis in the alveolar compartment and improve oxygenation. In this proof-of-concept safety study, adults with COVID-19-induced respiratory failure and a <300 mmHg PaO2/FiO2 (P/F) ratio requiring invasive mechanical ventilation (IMV) or non-invasive respiratory support (NIRS) received nebulized rt-PA in two cohorts (C1 and C2), alongside standard of care, between 23 April-30 July 2020 and 21 January-19 February 2021, respectively. Matched historical controls (MHC; n = 18) were used in C1 to explore efficacy. Safety co-primary endpoints were treatment-related bleeds and <1.0-1.5 g/L fibrinogen reduction. A variable dosing strategy with clinical efficacy endpoint and minimal safety concerns was determined in C1 for use in C2; patients were stratified by ventilation type to receive 40-60 mg rt-PA daily for ≤14 days. Nine patients in C1 (IMV, 6/9; NIRS, 3/9) and 26 in C2 (IMV, 12/26; NIRS, 14/26) received nebulized rt-PA for a mean (SD) of 6.7 (4.6) and 9.1(4.6) days, respectively. Four bleeds (one severe, three mild) in three patients were considered treatment related. There were no significant fibrinogen reductions. Greater improvements in mean P/F ratio from baseline to study end were observed in C1 compared with MHC (C1; 154 to 299 vs. MHC; 154 to 212). In C2, there was no difference in the baseline P/F ratio of NIRS and IMV patients. However, a larger improvement in the P/F ratio occurred in NIRS patients (NIRS; 126 to 240 vs. IMV; 120 to 188) and fewer treatment days were required (NIRS; 7.86 vs. IMV; 10.5). Nebulized rt-PA appears to be well-tolerated, with a trend towards improved oxygenation, particularly in the NIRS group. Randomized clinical trials are required to demonstrate the clinical effect significance and magnitude.
Collapse
Affiliation(s)
- Pratima Chowdary
- Katharine Dormandy Haemophilia and Thrombosis Centre, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
- Cancer Institute, University College London, London WC1E 6DD, UK
| | - Banwari Agarwal
- Department of Intensive Care and Anaesthesia, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
| | - Maria Rita Peralta
- Katharine Dormandy Haemophilia and Thrombosis Centre, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
- Cancer Institute, University College London, London WC1E 6DD, UK
| | - Sanjay Bhagani
- Department of Infectious Diseases, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
| | - Simon Lee
- Department of Infectious Diseases, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
| | - James Goldring
- Respiratory Medicine, Royal Free London NHS Foundation Trust, London NW1 2BU, UK
| | - Marc Lipman
- Respiratory Medicine, Royal Free London NHS Foundation Trust, London NW1 2BU, UK
- UCL Respiratory, University College London, London WC1E 6JF, UK;
| | - Emal Waqif
- Katharine Dormandy Haemophilia and Thrombosis Centre, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
| | - Mark Phillips
- Katharine Dormandy Haemophilia and Thrombosis Centre, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
- Cancer Institute, University College London, London WC1E 6DD, UK
| | - Helen Philippou
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9JT, UK
| | | | - Nicola J. Mutch
- Aberdeen Cardiovascular & Diabetes Centre, School of Medicine, Medical Sciences & Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Robert A. S. Ariëns
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9JT, UK
| | - Kathleen A. Stringer
- Department of Clinical Pharmacy, College of Pharmacy University of Michigan, Ann Arbor, MI 48109, USA
- Division of Pulmonary and Critical Care Medicine, School of Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Federico Ricciardi
- Department of Statistical Science, University College London, London WC1E 6BT, UK
| | | | - Derralynn Hughes
- Cancer Institute, University College London, London WC1E 6DD, UK
| | - Amit Nathwani
- Katharine Dormandy Haemophilia and Thrombosis Centre, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
- Cancer Institute, University College London, London WC1E 6DD, UK
| | - Anne Riddell
- Katharine Dormandy Haemophilia and Thrombosis Centre, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
- Haemophilia & Thrombosis Laboratory (Health Services Laboratories), Royal Free Hospital, London WC1H 9AX, UK
| | - David Patch
- Department of Hepatology, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
| | - Jim Buckley
- Department of Intensive Care and Anaesthesia, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
| | - Mark De Neef
- Department of Intensive Care and Anaesthesia, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
| | - Rahul Dimber
- Department of Intensive Care and Anaesthesia, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
| | - Cecilia Diaz-Garcia
- Katharine Dormandy Haemophilia and Thrombosis Centre, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
| | - Honey Patel
- Katharine Dormandy Haemophilia and Thrombosis Centre, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
| | - Aarti Nandani
- Clinical Trials Pharmacy, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
| | - Upuli Dissanayake
- Katharine Dormandy Haemophilia and Thrombosis Centre, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
| | - Nick Chadwick
- Katharine Dormandy Haemophilia and Thrombosis Centre, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
| | - Ahmed A. A. M. M. Alkhatip
- Department of Anaesthesia, Birmingham Children’s Hospital, Birmingham B4 6NH, UK
- Department of Anaesthesia, Faculty of Medicine, Beni-Suef University Hospital, Beni-Suef University, Beni-Suef 2721562, Egypt
| | - Peter Watkinson
- NIHR Biomedical Research Centre Oxford, Oxford University Hospitals NHS Trust, University of Oxford, Oxford OX3 9DU, UK
| | - Eamon Raith
- Bloomsbury Institute for Intensive Care Medicine, Department of Experimental and Translational Medicine, University College London, London WC1E 6JF, UK
- Discipline of Acute Care Medicine, School of Medicine, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Suveer Singh
- Department of Respiratory and Critical Care Medicine, Chelsea & Westminster Hospital, London SW10 9NH, UK
- Department of Adult Intensive Care, Royal Brompton Hospital, London SW3 6NP, UK
- Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, UK
| | - Tony Wolff
- Department of Intensive Care and Anaesthesia, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
| | - Rajeev Jha
- Department of Intensive Care and Anaesthesia, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
| | - Simon E. Brill
- UCL Respiratory, University College London, London WC1E 6JF, UK;
| | - Ameet Bakhai
- Department of Intensive Care and Anaesthesia, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
- Department of Cardiology, Royal Free London NHS Foundation Trust, London NW3 2PS, UK
| | - Alison Evans
- University College London (UCL)/University College London Hospitals NHS Trust (UCLH) Joint Research Office, London WC1E 6BT, UK; (A.E.)
| | - Farhat Gilani
- University College London (UCL)/University College London Hospitals NHS Trust (UCLH) Joint Research Office, London WC1E 6BT, UK; (A.E.)
| | - Keith Gomez
- Katharine Dormandy Haemophilia and Thrombosis Centre, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
- Cancer Institute, University College London, London WC1E 6DD, UK
| |
Collapse
|
5
|
Gao J, Zhang Z, Yan JY, Ge YX, Gao Y. Inflammation and coagulation abnormalities via the activation of the HMGB1‑RAGE/NF‑κB and F2/Rho pathways in lung injury induced by acute hypoxia. Int J Mol Med 2023; 52:67. [PMID: 37350396 PMCID: PMC10555482 DOI: 10.3892/ijmm.2023.5270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 05/15/2023] [Indexed: 06/24/2023] Open
Abstract
High‑altitude acute hypoxia is commonly associated with respiratory cardiovascular diseases. The inability to adapt to acute hypoxia may lead to cardiovascular dysfunction, lung injury and even death. Therefore, understanding the molecular basis of the adaptation to high‑altitude acute hypoxia may reveal novel therapeutic approaches with which to counteract the detrimental consequences of hypoxia. In the present study, a high‑altitude environment was simulated in a rat model in order to investigate the role of the high mobility group protein‑1 (HMGB1)/receptor for advanced glycation end products (RAGE)/NF‑κB and F2/Rho signaling pathways in lung injury induced by acute hypoxia. It was found that acute hypoxia caused inflammation through the HMGB1/RAGE/NF‑κB pathway and coagulation dysfunction through the F2/Rho pathway, both of which may be key processes in acute hypoxia‑induced lung injury. The present study provides new insight into the molecular basis of lung injury induced by acute hypoxia. The simultaneous activation of the HMGB1/RAGE/NF‑κB and F2/Rho signaling pathways plays a critical role in hypoxia‑induced inflammatory responses and coagulation abnormalities, and provides a theoretical basis for the development of potential therapeutic strategies.
Collapse
Affiliation(s)
| | | | - Jia-Yi Yan
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Yun-Xuan Ge
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Yue Gao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| |
Collapse
|
6
|
Hamill GS, Remy KE, Slain KN, Sallee CJ, Khemani R, Smith L, Shein SL. Association of Interventions With Outcomes in Children At-Risk for Pediatric Acute Respiratory Distress Syndrome: A Pediatric Acute Respiratory Distress Syndrome Incidence and Epidemiology Study. Pediatr Crit Care Med 2023; 24:574-583. [PMID: 37409896 DOI: 10.1097/pcc.0000000000003217] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
OBJECTIVES Describe the frequency with which transfusion and medications that modulate lung injury are administered to children meeting at-risk for pediatric acute respiratory distress syndrome (ARF-PARDS) criteria and evaluate for associations of transfusion, fluid balance, nutrition, and medications with unfavorable clinical outcomes. DESIGN Secondary analysis of the Pediatric Acute Respiratory Distress Syndrome Incidence and Epidemiology study, a prospective point prevalence study. All enrolled ARF-PARDS patients were included unless they developed subsequent pediatric acute respiratory distress syndrome (PARDS) within 24 hours of PICU admission or PICU length of stay was less than 24 hours. Univariate and multivariable analyses were used to identify associations between therapies given during the first 2 calendar days after ARF-PARDS diagnosis and subsequent PARDS diagnosis (primary outcome), 28-day PICU-free days (PFDs), and 28-day ventilator-free days (VFDs). SETTING Thirty-seven international PICUs. PATIENTS Two hundred sixty-seven children meeting Pediatric Acute Lung Injury Consensus Conference ARF-PARDS criteria. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS During the first 2 days after meeting ARF-PARDS criteria, 55% of subjects received beta-agonists, 42% received corticosteroids, 28% received diuretics, and 9% were transfused. Subsequent PARDS (15%) was associated with platelet transfusion (n = 11; adjusted odds ratio: 4.75 [95% CI 1.03-21.92]) and diuretics (n = 74; 2.55 [1.19-5.46]) in multivariable analyses that adjusted for comorbidities, PARDS risk factor, initial oxygen saturation by pulse oximetry:Fio2 ratio, and initial type of ventilation. Beta-agonists were associated with lower adjusted odds of subsequent PARDS (0.43 [0.19-0.98]). Platelets and diuretics were also associated with fewer PFDs and fewer VFDs in the multivariable models, and TPN was associated with fewer PFDs. Corticosteroids, net fluid balance, and volume of enteral feeding were not associated with the primary or secondary outcomes. CONCLUSIONS There is an independent association between platelet transfusion, diuretic administration, and unfavorable outcomes in children at risk for PARDS, although this may be related to treatment bias and unmeasured confounders. Nevertheless, prospective evaluation of the role of these management strategies on outcomes in children with ARF-PARDS is needed.
Collapse
Affiliation(s)
- Grant S Hamill
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Rainbow Babies and Children's Hospital, Cleveland, OH
| | - Kenneth E Remy
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Rainbow Babies and Children's Hospital, Cleveland, OH
| | - Katherine N Slain
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Rainbow Babies and Children's Hospital, Cleveland, OH
| | - Colin J Sallee
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, UCLA Mattel Children's Hospital, Los Angeles, CA
| | - Robinder Khemani
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, CA
| | - Lincoln Smith
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Seattle Children's Hospital, Seattle, WA
| | - Steven L Shein
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Rainbow Babies and Children's Hospital, Cleveland, OH
| |
Collapse
|
7
|
Vizzoni L, Migone C, Grassiri B, Zambito Y, Ferro B, Roncucci P, Mori F, Salvatore A, Ascione E, Crea R, Esin S, Batoni G, Piras AM. Biopharmaceutical Assessment of Mesh Aerosolised Plasminogen, a Step towards ARDS Treatment. Pharmaceutics 2023; 15:1618. [PMID: 37376068 PMCID: PMC10300680 DOI: 10.3390/pharmaceutics15061618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/18/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a severe complication of lung injuries, commonly associated with bacterial, fungal and viral infections, including SARS-CoV-2 viral infections. ARDS is strongly correlated with patient mortality and its clinical management is very complex, with no effective treatment presently available. ARDS involves severe respiratory failure, fibrin deposition in both airways and lung parenchyma, with the development of an obstructing hyaline membrane drastically limiting gas exchange. Moreover, hypercoagulation is related to deep lung inflammation, and a pharmacological action toward both aspects is expected to be beneficial. Plasminogen (PLG) is a main component of the fibrinolytic system playing key roles in various inflammation regulatory processes. The inhalation of PLG has been proposed in the form of the off-label administration of an eyedrop solution, namely, a plasminogen-based orphan medicinal product (PLG-OMP), by means of jet nebulisation. Being a protein, PLG is susceptible to partial inactivation under jet nebulisation. The aim of the present work is to demonstrate the efficacy of the mesh nebulisation of PLG-OMP in an in vitro simulation of clinical off-label administration, considering both the enzymatic and immunomodulating activities of PLG. Biopharmaceutical aspects are also investigated to corroborate the feasibility of PLG-OMP administration by inhalation. The nebulisation of the solution was performed using an Aerogen® SoloTM vibrating-mesh nebuliser. Aerosolised PLG showed an optimal in vitro deposition profile, with 90% of the active ingredient impacting the lower portions of a glass impinger. The nebulised PLG remained in its monomeric form, with no alteration of glycoform composition and 94% of enzymatic activity maintenance. Activity loss was observed only when PLG-OMP nebulisation was performed under simulated clinical oxygen administration. In vitro investigations evidenced good penetration of aerosolised PLG through artificial airway mucus, as well as poor permeation across an Air-Liquid Interface model of pulmonary epithelium. The results suggest a good safety profile of inhalable PLG, excluding high systemic absorption but with good mucus diffusion. Most importantly, the aerosolised PLG was capable of reversing the effects of an LPS-activated macrophage RAW 264.7 cell line, demonstrating the immunomodulating activity of PLG in an already induced inflammatory state. All physical, biochemical and biopharmaceutical assessments of mesh aerosolised PLG-OMP provided evidence for its potential off-label administration as a treatment for ARDS patients.
Collapse
Affiliation(s)
- Lucia Vizzoni
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Chiara Migone
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | | | - Ylenia Zambito
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
- Research Centre for Nutraceutical and Healthy Foods “NUTRAFOOD”, University of Pisa, 56124 Pisa, Italy
| | - Baldassare Ferro
- Anestesia e Rianimazione, Azienda USL Toscana Nord Ovest, 57124 Livorno, Italy
| | - Paolo Roncucci
- Anestesia e Rianimazione, Azienda USL Toscana Nord Ovest, 57124 Livorno, Italy
| | - Filippo Mori
- Kedrion S.p.A., Via di Fondovalle, Loc. Bolognana, 55027 Gallicano, Italy
| | - Alfonso Salvatore
- Kedrion S.p.A., Via di Fondovalle, Loc. Bolognana, 55027 Gallicano, Italy
| | - Ester Ascione
- Kedrion S.p.A., Via di Fondovalle, Loc. Bolognana, 55027 Gallicano, Italy
| | - Roberto Crea
- Kedrion S.p.A., Via di Fondovalle, Loc. Bolognana, 55027 Gallicano, Italy
| | - Semih Esin
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
- Centre for Instrument Sharing of University of Pisa (CISUP), 56126 Pisa, Italy
| | - Giovanna Batoni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
- Centre for Instrument Sharing of University of Pisa (CISUP), 56126 Pisa, Italy
| | - Anna Maria Piras
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
- Centre for Instrument Sharing of University of Pisa (CISUP), 56126 Pisa, Italy
| |
Collapse
|
8
|
Pantet O, Combescure C, Ltaief Z, Mazzolai L, Jeanneret S, Manzocchi-Besson S, Stricker H, Cereghetti S, Pugin J, Calmy A, Marti C, Robert-Ebadi H, Fontana P, Righini M, Casini A, Blondon M. Effect of therapeutic anticoagulation on gas exchange in mechanically ventilated COVID-19 patients: A secondary analysis of the COVID-HEP trial. Thromb Res 2023; 224:13-16. [PMID: 36801649 PMCID: PMC9911973 DOI: 10.1016/j.thromres.2023.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/18/2023] [Accepted: 02/03/2023] [Indexed: 02/12/2023]
Affiliation(s)
- Olivier Pantet
- Division of Intensive Care, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | | | - Zied Ltaief
- Division of Intensive Care, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Lucia Mazzolai
- Division of Angiology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | | | | | - Hans Stricker
- Division of Angiology, Locarno Regional Hospital, Locarno, Switzerland
| | - Sara Cereghetti
- Division of Intensive Care, Geneva University Hospitals, Geneva, Switzerland
| | - Jérôme Pugin
- Division of Intensive Care, Geneva University Hospitals, Geneva, Switzerland
| | - Alexandra Calmy
- HIV/AIDS Unit, Division of Infectious Disease, Geneva University Hospitals, Geneva, Switzerland
| | - Christophe Marti
- Division of General Internal Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Helia Robert-Ebadi
- Division of Angiology and Hemostasis, Geneva University Hospitals, Geneva, Switzerland
| | - Pierre Fontana
- Division of Angiology and Hemostasis, Geneva University Hospitals, Geneva, Switzerland
| | - Marc Righini
- Division of Angiology and Hemostasis, Geneva University Hospitals, Geneva, Switzerland
| | - Alessandro Casini
- Division of Angiology and Hemostasis, Geneva University Hospitals, Geneva, Switzerland
| | - Marc Blondon
- Division of Angiology and Hemostasis, Geneva University Hospitals, Geneva, Switzerland.
| |
Collapse
|
9
|
Markin SS, Lapshin RD, Baskina OS, Korotchenko SA, Mukhina IV, Ivanov SV, Semenov MP, Beregovykh VV, Semenov AM. Nebulized Non-Immunogenic Staphylokinase in the Mice Acute Lung Injury Model. Int J Mol Sci 2022; 23:9307. [PMID: 36012579 PMCID: PMC9409086 DOI: 10.3390/ijms23169307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/08/2022] [Accepted: 08/16/2022] [Indexed: 11/26/2022] Open
Abstract
Acute lung injury (ALI) as a model of acute respiratory distress syndrome is characterized by inflammation, complex coagulation, and hematologic abnormalities which result in the formation of fibrin-platelet microthrombi in the pulmonary vessels with the rapid development of progressive respiratory dysfunction. We hypothesize that a nebulized fibrinolytic agent, non-immunogenic staphylokinase (nSta), may be useful for ALI therapy. First, the effect of the nebulized nSta (0.2 mg/kg, 1.0 mg/kg, or 2.0 mg/kg) on the coagulogram parameters was studied in healthy rats. ALI was induced in mice by nebulized administration of lipopolysaccharide (LPS) at a dose of 10 mg/kg. nSta (0.2 mg/kg, 0.4 mg/kg or 0.6 mg/kg) was nebulized 30 min, 24 h, and 48 h after LPS administration. The level of pro-inflammatory cytokines was determined in the blood on the 8th day after LPS and nSta administration. The assessment of lung damage was based on their weighing and microscopic analysis. Fibrin/fibrinogen deposition in the lungs was determined by immunohistochemistry. After nSta nebulization in healthy rats, the fibrinogen blood level as well as activated partial thromboplastin time and prothrombin time did not change. In the nebulized ALI model, the mice showed an increase in lung weight due to their edema and rising fibrin deposition. An imbalance of proinflammatory cytokines was also found. Forty percent of mice with ALI without nSta nebulization had died. Nebulized nSta at a dose of 0.2 mg/kg reduced the severity of ALI: a decrease in interstitial edema and inflammatory infiltration was noted. At a dose of 0.4 mg/kg of nebulized nSta, the animals showed no peribronchial edema and the bronchi had an open clear lumen. At a dose of 0.6 mg/kg of nebulized nSta, the manifestations of ALI were completely eliminated. A significant dose-dependent reduction of the fibrin-positive areas in the lungs of mice with ALI was established. Nebulized nSta had a normalizing effect on the proinflammatory cytokines in blood- interleukin (IL)-1α, IL-17A, IL-6, and granulocyte-macrophage colony-stimulating factor (GM-CSF). These data showed the effectiveness of nebulized nSta and the perspectives of its clinical usage in COVID-19 patients with acute respiratory distress syndrome (ARDS).
Collapse
Affiliation(s)
- Sergey S. Markin
- Experimental Drug Research and Production Zone, Institute of Biomedical Chemistry, 119121 Moscow, Russia
- LLC “SuperGene”, 119270 Moscow, Russia
| | - Roman D. Lapshin
- Central Research Laboratory, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia
| | - Olga S. Baskina
- Central Research Laboratory, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia
| | - Svetlana A. Korotchenko
- Central Research Laboratory, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia
| | - Irina V. Mukhina
- Central Research Laboratory, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia
| | | | | | - Valerii V. Beregovykh
- Experimental Drug Research and Production Zone, Institute of Biomedical Chemistry, 119121 Moscow, Russia
| | | |
Collapse
|
10
|
Amini S, Rezabakhsh A, Hashemi J, Saghafi F, Azizi H, Sureda A, Habtemariam S, Khayat Kashani HR, Hesari Z, Sahebnasagh A. Pharmacotherapy consideration of thrombolytic medications in COVID-19-associated ARDS. J Intensive Care 2022; 10:38. [PMID: 35908022 PMCID: PMC9338522 DOI: 10.1186/s40560-022-00625-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 06/22/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND In late 2019, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) which is responsible for coronavirus disease (COVID-19), was identified as the new pathogen to lead pneumonia in Wuhan, China, which has spread all over the world and developed into a pandemic. Despite the over 1 year of pandemic, due to the lack of an effective treatment plan, the morbidity and mortality of COVID-19 remains high. Efforts are underway to find the optimal management for this viral disease. MAIN BODY SARS-CoV-2 could simultaneously affect multiple organs with variable degrees of severity, from mild to critical disease. Overproduction of pro-inflammatory mediators, exacerbated cellular and humoral immune responses, and coagulopathy such as Pulmonary Intravascular Coagulopathy (PIC) contributes to cell injuries. Considering the pathophysiology of the disease and multiple microthrombi developments in COVID-19, thrombolytic medications seem to play a role in the management of the disease. Beyond the anticoagulation, the exact role of thrombolytic medications in the management of patients with COVID-19-associated acute respiratory distress syndrome (ARDS) is not explicit. This review focuses on current progress in underlying mechanisms of COVID-19-associated pulmonary intravascular coagulopathy, the historical use of thrombolytic drugs in the management of ARDS, and pharmacotherapy considerations of thrombolytic therapy, their possible benefits, and pitfalls in COVID-19-associated ARDS. CONCLUSIONS Inhaled or intravenous administration of thrombolytics appears to be a salvage therapy for severe ARDS associated with COVID-19 by prompt attenuation of lung injury. Considering the pathogenesis of COVID-19-related ARDS and mechanism of action of thrombolytic agents, thrombolytics appear attractive options in stable patients without contraindications.
Collapse
Affiliation(s)
- Shahideh Amini
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Aysa Rezabakhsh
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Hashemi
- Department of Pathobiology and Laboratory Sciences, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Fatemeh Saghafi
- Department of Clinical Pharmacy, Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hossein Azizi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Antoni Sureda
- Research Group On Community Nutrition and Oxidative Stress, University of the Balearic Islands, Palma, Spain
- CIBEROBN (Physiopathology of Obesity and Nutrition CB12/03/30038), Instituto de Salud Carlos III, Madrid, Spain
| | - Solomon Habtemariam
- Pharmacognosy Research Laboratories and Herbal Analysis Services, University of Greenwich, Central Avenue, Chatham-Maritime, Kent, ME4 4TB UK
| | | | - Zahra Hesari
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Adeleh Sahebnasagh
- Clinical Research Center, Department of Internal Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| |
Collapse
|
11
|
Wang Y, Wu Y, Liu B, Yang H, Qian H, Cheng Y, Li X, Yang G, Zheng X, Shen F. Binding domain peptide ameliorates alveolar hypercoagulation and fibrinolytic inhibition in mice with lipopolysaccharide-induced acute respiratory distress syndrome Via NF-κB signaling pathway. Am J Transl Res 2022; 14:3854-3863. [PMID: 35836863 PMCID: PMC9274609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/07/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Alveolar hypercoagulation and fibrinolytic inhibition are shown to be associated with refractory hypoxemia in acute respiratory distress syndrome (ARDS), and the NF-κB pathway is involved in this process. The purpose of this study is to explore the role of NEMO-binding domain peptide (NBDP) in alleviating alveolar hypercoagulation and fibrinolytic inhibition induced by lipopolysaccharide (LPS) in ARDS mice and its related mechanisms. MATERIALS AND METHODS ARDS was induced by inhalation of LPS (mg/L) in adult male BALB/c mice. Mice were treated with intratracheal inhalation of NBDP or saline aerosol at increased concentrations 30 minutes before LPS administration. Six hours after LPS treatment, bronchoalveolar lavage fluids (BALF) were collected and then all mice were euthanized. In addition, coagulation and fibrinolysis associated factors in lung tissues and BALF were detected, and the activation of NF-κB signaling pathway was observed. RESULTS NBDP pretreatment dose-dependently inhibited the expression of tissue factor (TF) and plasminogen activator inhibitor (PAI) 1 in lung tissues, reduced the secretions of TF, PAI-1, thrombin-antithrombin (TAT) complex, and promoted activated protein C (APC) secretion in BALF induced by LPS. LPS-induced high expression of pulmonary procollagen peptide type lll (PIIIP) was also reduced in a dose-dependent manner under NBDP pretreatment. Western blotting showed that NBDP pretreatment significantly attenuated LPS-induced activation of IKKα/β, Iκα and NF-κB p65. NBDP pretreatment also inhibited the DNA binding activity of p65 induced by LPS. We also noticed that NBDP protected mice against LPS-induced lung injury in a dose-dependent manner. CONCLUSIONS The experimental findings demonstrate that through inhibiting the NF-κB signaling pathway, NBDP dose-dependently ameliorates LPS-induced alveolar hypercoagulation and fibrinolytic inhibition, which is expected to be a new therapeutic target to correct the abnormalities of alveolar coagulation and fibrinolytic pathways in ARDS.
Collapse
Affiliation(s)
- Yahui Wang
- Department of Intensive Care Unit, Guizhou Medical University Affiliated HospitalGuiyang 550001, Guizhou, China
- Department of Intensive Care Unit, The People’s Hospital of Weining CountyWeining County 553100, Guizhou, China
| | - Yanqi Wu
- Department of Intensive Care Unit, Guizhou Medical University Affiliated HospitalGuiyang 550001, Guizhou, China
| | - Bo Liu
- Department of Intensive Care Unit, Guizhou Medical University Affiliated HospitalGuiyang 550001, Guizhou, China
| | - Huilin Yang
- Department of Intensive Care Unit, Guizhou Medical University Affiliated HospitalGuiyang 550001, Guizhou, China
| | - Hong Qian
- Department of Intensive Care Unit, Guizhou Medical University Affiliated HospitalGuiyang 550001, Guizhou, China
| | - Yumei Cheng
- Department of Intensive Care Unit, Guizhou Medical University Affiliated HospitalGuiyang 550001, Guizhou, China
| | - Xiang Li
- Department of Intensive Care Unit, Guizhou Medical University Affiliated HospitalGuiyang 550001, Guizhou, China
| | - Guixia Yang
- Department of Intensive Care Unit, Guizhou Medical University Affiliated HospitalGuiyang 550001, Guizhou, China
| | - Xinghao Zheng
- Department of Intensive Care Unit, Guizhou Medical University Affiliated HospitalGuiyang 550001, Guizhou, China
| | - Feng Shen
- Department of Intensive Care Unit, Guizhou Medical University Affiliated HospitalGuiyang 550001, Guizhou, China
| |
Collapse
|
12
|
Ceccato A, Camprubí-Rimblas M, Campaña-Duel E, Areny-Balagueró A, Morales-Quinteros L, Artigas A. Anticoagulant Treatment in Severe ARDS COVID-19 Patients. J Clin Med 2022; 11:2695. [PMID: 35628822 PMCID: PMC9148112 DOI: 10.3390/jcm11102695] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/03/2022] [Accepted: 05/06/2022] [Indexed: 02/08/2023] Open
Abstract
Patients with COVID-19 may complicate their evolution with thromboembolic events. Incidence of thromboembolic complications are high and also, patients with the critically-ill disease showed evidence of microthrombi and microangiopathy in the lung probably due to endothelial damage by directly and indirectly injured endothelial and epithelial cells. Pulmonary embolism, deep venous thrombosis and arterial embolism were reported in patients with COVID-19, and several analytical abnormal coagulation parameters have been described as well. D-dimer, longer coagulation times and lower platelet counts have been associated with poor outcomes. The use of anticoagulation or high doses of prophylactic heparin is controversial. Despite the use of anticoagulation or high prophylactic dose of heparin have been associated with better outcomes in observational studies, only in patients with non-critically ill disease benefits for anticoagulation was observed. In critically-ill patient, anticoagulation was not associated with better outcomes. Other measures such as antiplatelet therapy, fibrinolytic therapy or nebulized anticoagulants are being studied in ongoing clinical trials.
Collapse
Affiliation(s)
- Adrian Ceccato
- Critical Care Research Center, Institut d’Investigació i Innovació Parc Taulí I3PT, ParcTaulí, 08208 Sabadell, Spain; (M.C.-R.); (E.C.-D.); (A.A.-B.); (L.M.-Q.); (A.A.)
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, 41092 Madrid, Spain
| | - Marta Camprubí-Rimblas
- Critical Care Research Center, Institut d’Investigació i Innovació Parc Taulí I3PT, ParcTaulí, 08208 Sabadell, Spain; (M.C.-R.); (E.C.-D.); (A.A.-B.); (L.M.-Q.); (A.A.)
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, 41092 Madrid, Spain
- Bioscience and Medicine Faculty, Universitat Autònoma de Barcelona, Bellaterra, 08193 Catalunya, Spain
| | - Elena Campaña-Duel
- Critical Care Research Center, Institut d’Investigació i Innovació Parc Taulí I3PT, ParcTaulí, 08208 Sabadell, Spain; (M.C.-R.); (E.C.-D.); (A.A.-B.); (L.M.-Q.); (A.A.)
| | - Aina Areny-Balagueró
- Critical Care Research Center, Institut d’Investigació i Innovació Parc Taulí I3PT, ParcTaulí, 08208 Sabadell, Spain; (M.C.-R.); (E.C.-D.); (A.A.-B.); (L.M.-Q.); (A.A.)
| | - Luis Morales-Quinteros
- Critical Care Research Center, Institut d’Investigació i Innovació Parc Taulí I3PT, ParcTaulí, 08208 Sabadell, Spain; (M.C.-R.); (E.C.-D.); (A.A.-B.); (L.M.-Q.); (A.A.)
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, 41092 Madrid, Spain
- Bioscience and Medicine Faculty, Universitat Autònoma de Barcelona, Bellaterra, 08193 Catalunya, Spain
- Department of Intensive Care Medicine, Hospital Universitari Sant Pau, 08025 Barcelona, Spain
| | - Antonio Artigas
- Critical Care Research Center, Institut d’Investigació i Innovació Parc Taulí I3PT, ParcTaulí, 08208 Sabadell, Spain; (M.C.-R.); (E.C.-D.); (A.A.-B.); (L.M.-Q.); (A.A.)
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, 41092 Madrid, Spain
- Bioscience and Medicine Faculty, Universitat Autònoma de Barcelona, Bellaterra, 08193 Catalunya, Spain
| |
Collapse
|
13
|
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a devastating pandemic. Although most people infected with SARS-CoV-2 develop a mild to moderate disease with virus replication restricted mainly to the upper airways, some progress to having a life-threatening pneumonia. In this Review, we explore recent clinical and experimental advances regarding SARS-CoV-2 pathophysiology and discuss potential mechanisms behind SARS-CoV-2-associated acute respiratory distress syndrome (ARDS), specifically focusing on new insights obtained using novel technologies such as single-cell omics, organoid infection models and CRISPR screens. We describe how SARS-CoV-2 may infect the lower respiratory tract and cause alveolar damage as a result of dysfunctional immune responses. We discuss how this may lead to the induction of a 'leaky state' of both the epithelium and the endothelium, promoting inflammation and coagulation, while an influx of immune cells leads to overexuberant inflammatory responses and immunopathology. Finally, we highlight how these findings may aid the development of new therapeutic interventions against COVID-19.
Collapse
|
14
|
Yakovlev OA, Yudin MA, Chepur SV, Vengerovich NG, Stepanov AV, Babkin AA. Non-Specific Targets for Correction of Pneumonia Caused by Aerosols Containing Damaging Factors of Various Nature. BIOLOGY BULLETIN REVIEWS 2022; 12. [PMCID: PMC9749646 DOI: 10.1134/s207908642206010x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This review article provides data on the current state of the pathogenesis peculiarities of body and lung inflammation (pneumonia) under the influence of damaging factors of various nature: infectious agents, chemical toxicants, as well as incorporated radionuclides, etc. The peculiarities of inflammation itself, as a typical pathological process, are considered. Information on mediators that induce the so-called pro-resolving phase of inflammation manifestations is given. Approaches to the neuroimmune correction of non-specific inflammation are substantiated. Data on the following alternative approaches to the correction of nonspecific inflammation are summarized: factors of the coagulation system, modulators of the integrated stress response, and modulators of sigma-1 receptors. Based on the data presented, general directions for the treatment of nonspecific pneumonia are formulated, including reflexogenic and anti-inflammatory therapy in combination with multimodal drugs, as well as pro-resolving therapy in combination with drugs that prevent fibrosis.
Collapse
Affiliation(s)
- O. A. Yakovlev
- State Research Experimental Institute of Military Medicine, 198515 St. Petersburg, Russia
| | - M. A. Yudin
- State Research Experimental Institute of Military Medicine, 198515 St. Petersburg, Russia ,North-Western State Medical University named after I.I. Mechnikov, 195067 St. Petersburg, Russia
| | - S. V. Chepur
- State Research Experimental Institute of Military Medicine, 198515 St. Petersburg, Russia
| | - N. G. Vengerovich
- State Research Experimental Institute of Military Medicine, 198515 St. Petersburg, Russia ,Saint-Petersburg State Chemical Pharmaceutical University, 197376 St. Petersburg, Russia
| | - A. V. Stepanov
- State Research Experimental Institute of Military Medicine, 198515 St. Petersburg, Russia
| | - A. A. Babkin
- State Research Experimental Institute of Military Medicine, 198515 St. Petersburg, Russia
| |
Collapse
|
15
|
Livingstone SA, Wildi KS, Dalton HJ, Usman A, Ki KK, Passmore MR, Li Bassi G, Suen JY, Fraser JF. Coagulation Dysfunction in Acute Respiratory Distress Syndrome and Its Potential Impact in Inflammatory Subphenotypes. Front Med (Lausanne) 2021; 8:723217. [PMID: 34490308 PMCID: PMC8417599 DOI: 10.3389/fmed.2021.723217] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/29/2021] [Indexed: 12/12/2022] Open
Abstract
The Acute Respiratory Distress Syndrome (ARDS) has caused innumerable deaths worldwide since its initial description over five decades ago. Population-based estimates of ARDS vary from 1 to 86 cases per 100,000, with the highest rates reported in Australia and the United States. This syndrome is characterised by a breakdown of the pulmonary alveolo-epithelial barrier with subsequent severe hypoxaemia and disturbances in pulmonary mechanics. The underlying pathophysiology of this syndrome is a severe inflammatory reaction and associated local and systemic coagulation dysfunction that leads to pulmonary and systemic damage, ultimately causing death in up to 40% of patients. Since inflammation and coagulation are inextricably linked throughout evolution, it is biological folly to assess the two systems in isolation when investigating the underlying molecular mechanisms of coagulation dysfunction in ARDS. Although the body possesses potent endogenous systems to regulate coagulation, these become dysregulated and no longer optimally functional during the acute phase of ARDS, further perpetuating coagulation, inflammation and cell damage. The inflammatory ARDS subphenotypes address inflammatory differences but neglect the equally important coagulation pathway. A holistic understanding of this syndrome and its subphenotypes will improve our understanding of underlying mechanisms that then drive translation into diagnostic testing, treatments, and improve patient outcomes.
Collapse
Affiliation(s)
- Samantha A Livingstone
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Karin S Wildi
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.,Cardiovascular Research Institute Basel (CRIB), Basel, Switzerland
| | | | - Asad Usman
- Department of Anesthesiology and Critical Care, The University of Pennsylvania, Philadelphia, PA, United States
| | - Katrina K Ki
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Margaret R Passmore
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Gianluigi Li Bassi
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.,Department of Pulmonology and Critical Care, Hospital Clínic de Barcelona, Universitad de Barcelona and IDIBAPS, CIBERES, Barcelona, Spain
| | - Jacky Y Suen
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - John F Fraser
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
16
|
Yang H, Qian H, Liu B, Wu Y, Cheng Y, Zheng X, Li X, Yang G, He T, Li S, Shen F. Triptolide dose-dependently improves LPS-induced alveolar hypercoagulation and fibrinolysis inhibition through NF-κB inactivation in ARDS mice. Biomed Pharmacother 2021; 139:111569. [PMID: 34243622 DOI: 10.1016/j.biopha.2021.111569] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/20/2021] [Accepted: 03/31/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Alveolar hypercoagulation and fibrinolysis inhibition were associated with the refractory hypoxemia and the high mortality in patient with acute respiratory distress syndrome (ARDS), and NF-κB pathway was confirmed to contribute to the process. Triptolide (TP) significantly inhibited NF-κB pathway and thus depressed accessive inflammatory response in ARDS. We speculate that TP could improve alveolar hypercoagulation and fibrinolytic inhibition in LPS-induced ARDS via NF-κB inactivation. PURPOSE The aim of this experiment was to explore the efficacy and potential mechanism of TP on alveolar hypercoagulation and fibrinolysis inhibition in LPS-induced ARDS in mice. METHODS 50 μl of LPS (5 mg/ml) was inhalationally given to C57BL/6 mice to set up ARDS model. Male mice were randomly accepted with LPS, LPS + TP (1 μg/kg, 10 μg/kg, 50 μg/kg respectively), or with NEMO Binding domain peptide (NBD), an inhibitor of NF-κB. TP (1 μg/kg, 10 μg/kg, 50 μg/kg) were intraperitoneally injected or 10 μg/50 μl of NBD solution were inhaled 30 min before LPS inhalation. A same volume of normal saline (NS) substituted for TP in mice in control. The endpoint of experiment was at 8 hours after LPS stimulation. Pulmonary tissues were taken for hematoxylin-eosin (HE) staining, wet / dry ratio and for lung injury scores (LIS). Tissue factor (TF) and plasminogen activator inhibitor (PAI)-1 in lung tissue were detected by Western-blotting and by quantitative Real-time PCR(qPCR) respectively. Concentrations of TF, PAI-1, thrombin-antithrombin complex (TAT), procollagen peptide type Ⅲ (PⅢP) and activated protein C (APC) in bronchoalveolar lavage fluid (BALF) were measured by ELISA. NF-κB activation and p65-DNA binding activity in pulmonary tissue were simultaneously determined. RESULTS LPS stimulation resulted in pulmonary edema, neutrophils infiltration, obvious alveolar collapse, interstitial congestion, with high LIS, which were all dose-dependently ameliorated by Triptolide. LPS also dramatically promoted the expressions of TF and PAI-1 either in mRNA or in protein in lung tissue, and significantly stimulated the secretions of TF, PAI-1, TAT, PⅢP but inhibited APC production in BALF, which were all reversed by triptolide treatment in dose-dependent manner. TP dose-dependently inhibited the activation of NF-κB pathway induced by LPS, indicated by the changes of phosphorylations of p65 (p-p65), p-IKKα/β and p-IκBα, and weakened p65-DNA binding activity. TP and NBD had same efficacies either on alveolar hypercoagulation and fibrinolysis inhibition or on NF-κB signalling pathway in ARDS mice. CONCLUSIONS TP dose-dependently improves alveolar hypercoagulation and fibrinolysis inhibition in ARDS mice through inhibiting NF-κB signaling pathway. Our data demonstrate that TP is expected to be an effective selection in ARDS.
Collapse
Affiliation(s)
- Huilin Yang
- Department of Intensive Care Unit, Guizhou Medical University Affiliated Hospital, Guiyang 550001, China.
| | - Hong Qian
- Department of Intensive Care Unit, Guizhou Medical University Affiliated Hospital, Guiyang 550001, China.
| | - Bo Liu
- Department of Intensive Care Unit, Guizhou Medical University Affiliated Hospital, Guiyang 550001, China.
| | - Yanqi Wu
- Department of Intensive Care Unit, Guizhou Medical University Affiliated Hospital, Guiyang 550001, China.
| | - Yumei Cheng
- Department of Intensive Care Unit, Guizhou Medical University Affiliated Hospital, Guiyang 550001, China.
| | - Xinghao Zheng
- Department of Intensive Care Unit, Guizhou Medical University Affiliated Hospital, Guiyang 550001, China.
| | - Xiang Li
- Department of Intensive Care Unit, Guizhou Medical University Affiliated Hospital, Guiyang 550001, China.
| | - Guixia Yang
- Department of Intensive Care Unit, Guizhou Medical University Affiliated Hospital, Guiyang 550001, China.
| | - Tianhui He
- Department of Intensive Care Unit, Guizhou Medical University Affiliated Hospital, Guiyang 550001, China.
| | - Shuwen Li
- Department of Intensive Care Unit, Guizhou Medical University Affiliated Hospital, Guiyang 550001, China.
| | - Feng Shen
- Department of Intensive Care Unit, Guizhou Medical University Affiliated Hospital, Guiyang 550001, China.
| |
Collapse
|
17
|
Mazilu L, Katsiki N, Nikolouzakis TK, Aslanidis MI, Lazopoulos G, Kouretas D, Tsatsakis A, Suceveanu AI, Stoian AP, Parepa IR, Voinea F, Suceveanu AP, Arsene AL, Velescu BȘ, Vesa C, Nitipir C. Thrombosis and Haemostasis challenges in COVID-19 - Therapeutic perspectives of heparin and tissue-type plasminogen activator and potential toxicological reactions-a mini review. Food Chem Toxicol 2021; 148:111974. [PMID: 33421462 PMCID: PMC7837001 DOI: 10.1016/j.fct.2021.111974] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/19/2020] [Accepted: 01/02/2021] [Indexed: 12/20/2022]
Abstract
The coronavirus disease (COVID)-19 pandemic is a major challenge for the health systems worldwide. Acute respiratory distress syndrome (ARDS), is one of the most common complications of the COVID-19 infection. The activation of the coagulation system plays an important role in the pathogenesis of ARDS. The development of lung coagulopathy involves thrombin generation and fibrinolysis inhibition. Unfractionated heparin and its recently introduced counterpart low molecular weight heparin (LMWH), are widely used anticoagulants with a variety of clinical indications allowing for limited and manageable physio-toxicologic side effects while the use of protamine sulfate, heparin's effective antidote, has made their use even safer. Tissue-type plasminogen activator (tPA) is approved as intravenous thrombolytic treatment. The present narrative review discusses the use of heparin and tPA in the treatment of COVID-19-induced ARDS and their related potential physio-toxicologic side effects. The article is a quick review of articles on anticoagulation in COVID infection and the potential toxicologic reactions associated with these drugs.
Collapse
Affiliation(s)
- Laura Mazilu
- Oncology Department, Clinical Emergency Hospital, Faculty of Medicine, "Ovidius" University, Constanța, Romania
| | - Niki Katsiki
- First Department of Internal Medicine, Diabetes Center, Division of Endocrinology and Metabolism, AHEPA University Hospital, Thessaloniki, Greece
| | | | | | - George Lazopoulos
- Department of Cardiothoracic Surgery, University General Hospital of Heraklion, Medical School, University of Crete, Greece
| | - Dimitrios Kouretas
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, Greece
| | - Aristidis Tsatsakis
- Department of Biochemistry and Biotechnology, University of Thessaly, Larisa, 41500, Greece
| | - Andra-Iulia Suceveanu
- Gastroenterology Department, Clinical Emergency Hospital, Faculty of Medicine, "Ovidius" University, Constanța, Romania
| | - Anca-Pantea Stoian
- Department of Diabetes, Nutrition and Metabolic Diseases, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania.
| | - Irinel-Raluca Parepa
- Cardiology Department, Clinical Emergency Hospital, Faculty of Medicine, "Ovidius" University, Constanța, Romania
| | - Felix Voinea
- Urology Department, Clinical Emergency Hospital, Faculty of Medicine, "Ovidius" University, Constanța, Romania
| | - Adrian Paul Suceveanu
- Internal Medicine Department, Clinical Emergency Hospital, Faculty of Medicine, "Ovidius" University, Constanța, Romania
| | - Andreea Letiția Arsene
- Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| | - Bruno Ștefan Velescu
- Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| | - Cosmin Vesa
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| | - Cornelia Nitipir
- Oncology Department, Elias Emergency Hospital, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
18
|
Chen W, Pan JY. Anatomical and Pathological Observation and Analysis of SARS and COVID-19: Microthrombosis Is the Main Cause of Death. Biol Proced Online 2021; 23:4. [PMID: 33472576 PMCID: PMC7816139 DOI: 10.1186/s12575-021-00142-y] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 01/11/2021] [Indexed: 12/22/2022] Open
Abstract
The spread of the coronavirus (SARS-CoV-2, COVID-19 for short) has caused a large number of deaths around the world. We summarized the data reported in the past few months and emphasized that the main causes of death of COVID-19 patients are DAD (Diffuse Alveolar Damage) and DIC (Disseminated intravascular coagulation). Microthrombosis is a prominent clinical feature of COVID-19, and 91.3% of dead patients had microthrombosis.Endothelial damage caused by SARS-CoV-2 cell invasion and subsequent host response disorders involving inflammation and coagulation pathways play a key role in the progression of severe COVID-19. Microvascular thrombosis may lead to microcirculation disorders and multiple organ failure lead to death.The characteristic pathological changes of DAD include alveolar epithelial and vascular endothelial injury, increased alveolar membrane permeability, large numbers of neutrophil infiltration, alveolar hyaline membrane formation, and hypoxemia and respiratory distress as the main clinical manifestations. DAD leads to ARDS in COVID-19 patients. DIC is a syndrome characterized by the activation of systemic intravascular coagulation, which leads to extensive fibrin deposition in the blood. Its occurrence and development begin with the expression of tissue factor and interact with physiological anticoagulation pathways. The down-regulation of fibrin and the impaired fibrinolysis together lead to extensive fibrin deposition.DIC is described as a decrease in the number of platelets and an increase in fibrin degradation products, such as D-dimer and low fibrinogen. The formation of microthrombus leads to the disturbance of microcirculation, which in turn leads to the death of the patient. However, the best prevention and treatment of COVID-19 microthrombosis is still uncertain.This review discusses the latest findings of basic and clinical research on COVID-19-related microthrombosis, and then we proposed the theory of microcirculation perfusion bundle therapy to explore effective methods for preventing and treating COVID-19-related microthrombosis. Further research is urgently needed to clarify how SARS-CoV-2 infection causes thrombotic complications, and how it affects the course and severity of the disease. To cultivate a more comprehensive understanding of the underlying mechanism of this disease. Raise awareness of the importance of preventing and treating microthrombosis in patients with COVID-19.
Collapse
Affiliation(s)
- Wenjing Chen
- School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jing Ye Pan
- School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Critical Care and Artificial Intelligence, Wenzhou, China
| |
Collapse
|
19
|
The Association of Low Molecular Weight Heparin Use and In-hospital Mortality Among Patients Hospitalized with COVID-19. Cardiovasc Drugs Ther 2021; 36:113-120. [PMID: 33394360 PMCID: PMC7779648 DOI: 10.1007/s10557-020-07133-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/17/2020] [Indexed: 01/08/2023]
Abstract
Purpose To determine the association between low molecular weight heparin (LMWH) use and mortality in hospitalized COVID-19 patients. Methods We conducted a retrospective study of patients consecutively enrolled from two major academic hospitals exclusively for COVID-19 in Wuhan, China, from January 26, 2020, to March 26, 2020. The primary outcome was adjusted in-hospital mortality in the LMWH group compared with the non-LMWH group using the propensity score. Results Overall, 525 patients with COVID-19 enrolled with a median age of 64 years (IQR 19), and 49.33% men. Among these, 120 (22.86%) were treated with LMWH. Compared with the non-LMWH group, the LMWH group was more likely to be older and male; had a history of hypertension, diabetes, coronary heart disease (CHD), or stroke; and had more severe COVID-19 parameters such as higher inflammatory cytokines or D-dimer. Compared with non-LMWH group, LMWH group had a higher unadjusted in-hospital mortality rate (21.70% vs. 11.10%; p = 0.004), but a lower adjusted mortality risk (adjusted odds ratio [OR], 0.20; 95% CI, 0.09–0.46). A propensity score-weighting analysis demonstrated similar findings (adjusted OR, 0.18; 95% CI, 0.10–0.30). Subgroup analysis showed a significant survival benefit among those who were severely (adjusted OR, 0.07; 95% CI, 0.02–0.23) and critically ill (adjusted OR, 0.32; 95% CI, 0.15–0.65), as well as among the elderly patients’ age > 65, IL-6 > 10 times upper limit level, and D-dimer > 5 times upper limit level. Conclusions Among hospitalized COVID-19 patients, LMWH use was associated with lower all-cause in-hospital mortality than non-LMWH users. The survival benefit was particularly significant among more severely ill patients.
Collapse
|
20
|
Cheng Y, Liu B, Qian H, Yang H, Wang Y, Wu Y, Shen F. BAY11-7082 inhibits the expression of tissue factor and plasminogen activator inhibitor-1 in type-II alveolar epithelial cells following TNF-α stimulation via the NF-κB pathway. Exp Ther Med 2020; 21:177. [PMID: 33552241 DOI: 10.3892/etm.2020.9608] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 11/17/2020] [Indexed: 12/26/2022] Open
Abstract
Pulmonary inflammation strongly promotes alveolar hypercoagulation and fibrinolytic inhibition. NF-κB signaling regulates the expression of molecules associated with coagulation and fibrinolytic inhibition in type-II alveolar epithelial cells (AECII) stimulated by lipopolysaccharide. However, whether TNF-α-induced alveolar hypercoagulation and fibrinolysis inhibition is also associated with the NF-κB pathway remains to be determined. The aim of the present study was to determine whether BAY11-7082, an inhibitor of the NF-κB pathway, inhibits the expressions of tissue factor (TF) and plasminogen activator inhibitor-1 (PAI-1) in AECⅡ in response to TNF-α. Rat AECII were treated with BAY11-7082 for 24 h and stimulated with TNF-α for 1 h. The expression of TF and PAI-1 were determined using western blotting and reverse transcription-quantitative PCR. The concentrations of TF and PAI-1 in culture supernatant were also measured by ELISA. Moreover, levels of NF-κB p65 (p65), phosphorylated (p)-p65 (p-p65), inhibitor of NF-κB α (IκBα) and p-IκBα were also evaluated. Immunofluorescence was used to detect p65 levels in cell nuclei. TNF-α significantly promoted TF and PAI-1 expression either at the mRNA or protein level in AECII cells. Concentrations of TF and PAI-1 in supernatant also significantly increased upon TNF-α stimulation. Furthermore, TNF-α upregulated the levels of p-IκBα, p65, and p-p65 in the cytoplasm. Immunofluorescence analysis indicated that TNF-α increased p65 translocation from the cytoplasm to the nucleus. However, AECII pre-treated with BAY11-7082 expressed lower levels of TF and PAI-1 following TNF-α treatment. Levels of p-IκBα, p65 and p-p65 in the cytoplasm also decreased, and translocation of p65 from cytoplasm into the nucleus was inhibited by BAY11-7082 pretreatment. These findings suggest that BAY11-7082 improves the hypercoagulation and fibrinolytic inhibition induced by TNF-α in alveolar epithelial cells via the NF-κB signaling pathway. BAY11-7082 might represent a therapeutic option for alveolar hypercoagulation and fibrinolytic inhibition in acute respiratory distress syndrome.
Collapse
Affiliation(s)
- Yumei Cheng
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Bo Liu
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Hong Qian
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Huilin Yang
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Yahui Wang
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Yanqi Wu
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Feng Shen
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| |
Collapse
|
21
|
Transesophageal echocardiography-associated tracheal microaspiration and ventilator-associated pneumonia in intubated critically ill patients: a multicenter prospective observational study. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2020; 24:679. [PMID: 33287866 PMCID: PMC7719845 DOI: 10.1186/s13054-020-03380-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 11/09/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Microaspiration of gastric and oropharyngeal secretions is the main causative mechanism of ventilator-associated pneumonia (VAP). Transesophageal echocardiography (TEE) is a routine investigation tool in intensive care unit and could enhance microaspiration. This study aimed at evaluating the impact of TEE on microaspiration and VAP in intubated critically ill adult patients. METHODS It is a four-center prospective observational study. Microaspiration biomarkers (pepsin and salivary amylase) concentrations were quantitatively measured on tracheal aspirates drawn before and after TEE. The primary endpoint was the percentage of patients with TEE-associated microaspiration, defined as: (1) ≥ 50% increase in biomarker concentration between pre-TEE and post-TEE samples, and (2) a significant post-TEE biomarker concentration (> 200 μg/L for pepsin and/or > 1685 IU/L for salivary amylase). Secondary endpoints included the development of VAP within three days after TEE and the evolution of tracheal cuff pressure throughout TEE. RESULTS We enrolled 100 patients (35 females), with a median age of 64 (53-72) years. Of the 74 patients analyzed for biomarkers, 17 (23%) got TEE-associated microaspiration. However, overall, pepsin and salivary amylase levels were not significantly different between before and after TEE, with wide interindividual variability. VAP occurred in 19 patients (19%) within 3 days following TEE. VAP patients had a larger tracheal tube size and endured more attempts of TEE probe introduction than their counterparts but showed similar aspiration biomarker concentrations. TEE induced an increase in tracheal cuff pressure, especially during insertion and removal of the probe. CONCLUSIONS We could not find any association between TEE-associated microaspiration and the development of VAP during the three days following TEE in intubated critically ill patients. However, our study cannot formally rule out a role for TEE because of the high rate of VAP observed after TEE and the limitations of our methods.
Collapse
|
22
|
Lemos ACB, do Espírito Santo DA, Salvetti MC, Gilio RN, Agra LB, Pazin-Filho A, Miranda CH. Therapeutic versus prophylactic anticoagulation for severe COVID-19: A randomized phase II clinical trial (HESACOVID). Thromb Res 2020; 196:359-366. [PMID: 32977137 PMCID: PMC7503069 DOI: 10.1016/j.thromres.2020.09.026] [Citation(s) in RCA: 193] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/15/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Coronavirus disease 2019 (COVID-19) causes a hypercoagulable state. Several autopsy studies have found microthrombi in pulmonary circulation. METHODS In this randomized, open-label, phase II study, we randomized COVID-19 patients requiring mechanical ventilation to receive either therapeutic enoxaparin or the standard anticoagulant thromboprophylaxis. We evaluated the gas exchange over time through the ratio of partial pressure of arterial oxygen (PaO2) to the fraction of inspired oxygen (FiO2) at baseline, 7, and 14 days after randomization, the time until successful liberation from mechanical ventilation, and the ventilator-free days. RESULTS Ten patients were assigned to the therapeutic enoxaparin and ten patients to prophylactic anticoagulation. There was a statistically significant increase in the PaO2/FiO2 ratio over time in the therapeutic group (163 [95% confidence interval - CI 133-193] at baseline, 209 [95% CI 171-247] after 7 days, and 261 [95% CI 230-293] after 14 days), p = 0.0004. In contrast, we did not observe this improvement over time in the prophylactic group (184 [95% CI 146-222] at baseline, 168 [95% CI 142-195] after 7 days, and 195 [95% CI 128-262] after 14 days), p = 0.487. Patients of the therapeutic group had a higher ratio of successful liberation from mechanical ventilation (hazard ratio: 4.0 [95% CI 1.035-15.053]), p = 0.031 and more ventilator-free days (15 days [interquartile range IQR 6-16] versus 0 days [IQR 0-11]), p = 0.028 when compared to the prophylactic group. CONCLUSION Therapeutic enoxaparin improves gas exchange and decreases the need for mechanical ventilation in severe COVID-19. TRIAL REGISTRATION REBEC RBR-949z6v.
Collapse
Affiliation(s)
- Anna Cristina Bertoldi Lemos
- Division of Emergency Medicine, Department of Internal Medicine, Ribeirão Preto School of Medicine, São Paulo University, Ribeirão Preto, SP, Brazil
| | - Douglas Alexandre do Espírito Santo
- Division of Emergency Medicine, Department of Internal Medicine, Ribeirão Preto School of Medicine, São Paulo University, Ribeirão Preto, SP, Brazil
| | - Maísa Cabetti Salvetti
- Hospital Estadual de Américo Brasiliense, Ribeirão Preto School of Medicine, São Paulo University, Ribeirão Preto, SP, Brazil
| | - Renato Noffs Gilio
- Hospital Estadual de Américo Brasiliense, Ribeirão Preto School of Medicine, São Paulo University, Ribeirão Preto, SP, Brazil
| | - Lucas Barbosa Agra
- Division of Emergency Medicine, Department of Internal Medicine, Ribeirão Preto School of Medicine, São Paulo University, Ribeirão Preto, SP, Brazil
| | - Antonio Pazin-Filho
- Division of Emergency Medicine, Department of Internal Medicine, Ribeirão Preto School of Medicine, São Paulo University, Ribeirão Preto, SP, Brazil
| | - Carlos Henrique Miranda
- Division of Emergency Medicine, Department of Internal Medicine, Ribeirão Preto School of Medicine, São Paulo University, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
23
|
Repurposing of Plasminogen: An Orphan Medicinal Product Suitable for SARS-CoV-2 Inhalable Therapeutics. Pharmaceuticals (Basel) 2020; 13:ph13120425. [PMID: 33260813 PMCID: PMC7761183 DOI: 10.3390/ph13120425] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/21/2020] [Accepted: 11/25/2020] [Indexed: 12/13/2022] Open
Abstract
The SARS-CoV-2 infection is associated with pulmonary coagulopathy, which determines the deposition of fibrin in the air spaces and lung parenchyma. The resulting lung lesions compromise patient pulmonary function and increase mortality, or end in permanent lung damage for those who have recovered from the COVID-19 disease. Therefore, local pulmonary fibrinolysis can be efficacious in degrading pre-existing fibrin clots and reducing the conversion of lung lesions into lasting scars. Plasminogen is considered a key player in fibrinolysis processes, and in view of a bench-to-bedside translation, we focused on the aerosolization of an orphan medicinal product (OMP) for ligneous conjunctivitis: human plasminogen (PLG-OMP) eye drops. As such, the sterile and preservative-free solution guarantees the pharmaceutical quality of GMP production and meets the Ph. Eur. requirements of liquid preparations for nebulization. PLG-OMP aerosolization was evaluated both from technological and stability viewpoints, after being submitted to either jet or ultrasonic nebulization. Jet nebulization resulted in a more efficient delivery of an aerosol suitable for pulmonary deposition. The biochemical investigation highlighted substantial protein integrity maintenance with the percentage of native plasminogen band > 90%, in accordance with the quality specifications of PLG-OMP. In a coherent way, the specific activity of plasminogen is maintained within the range 4.8–5.6 IU/mg (PLG-OMP pre-nebulization: 5.0 IU/mg). This is the first study that focuses on the technological and biochemical aspects of aerosolized plasminogen, which could affect both treatment efficacy and clinical dosage delivery. Increasing evidence for the need of local fibrinolytic therapy could merge with the availability of PLG-OMP as an easy handling solution, readily aerosolizable for a fast translation into an extended clinical efficacy assessment in COVID-19 patients.
Collapse
|
24
|
Liu Y, Xiang D, Zhang H, Yao H, Wang Y. Hypoxia-Inducible Factor-1: A Potential Target to Treat Acute Lung Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8871476. [PMID: 33282113 PMCID: PMC7685819 DOI: 10.1155/2020/8871476] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/29/2020] [Accepted: 11/04/2020] [Indexed: 02/07/2023]
Abstract
Acute lung injury (ALI) is an acute hypoxic respiratory insufficiency caused by various intra- and extrapulmonary injury factors. Presently, excessive inflammation in the lung and the apoptosis of alveolar epithelial cells are considered to be the key factors in the pathogenesis of ALI. Hypoxia-inducible factor-1 (HIF-1) is an oxygen-dependent conversion activator that is closely related to the activity of reactive oxygen species (ROS). HIF-1 has been shown to play an important role in ALI and can be used as a potential therapeutic target for ALI. This manuscript will introduce the progress of HIF-1 in ALI and explore the feasibility of applying inhibitors of HIF-1 to ALI, which brings hope for the treatment of ALI.
Collapse
Affiliation(s)
- Yang Liu
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan 430071, China
| | - Du Xiang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan 430071, China
| | - Hengcheng Zhang
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, 02115 MA, USA
| | - Hanlin Yao
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan 430071, China
| | - Yanfeng Wang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan 430071, China
| |
Collapse
|
25
|
Frazer JS, Tyrynis Everden AJ. Emerging patterns of hypercoagulability associated with critical COVID-19: A review. TRENDS IN ANAESTHESIA AND CRITICAL CARE 2020; 34:4-13. [PMID: 38620391 PMCID: PMC7346831 DOI: 10.1016/j.tacc.2020.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 07/02/2020] [Accepted: 07/07/2020] [Indexed: 12/21/2022]
Abstract
While the COVID-19 pandemic sweeps the world, much evidence is being gathered regarding its novel pathological mechanisms. It is the authors' clinical experience that patients in the intensive care unit suffering from COVID-19 are extremely pro-coagulable, with venous and arterial thromboembolism frequently observed, and losses of vascular access lines and filtration circuits to thrombosis now commonplace. Here, we explore the evidence for hypercoagulability in this group, presenting evidence of both a localised pulmonary hypercoagulability, and a systemic hypercoagulability resulting in thrombosis distant to the pulmonary vasculature. Furthermore, we discuss the possible risk factors exacerbated by, or selected for in COVID-19. We review the available evidence for use of plasma D-dimer as a prognostic marker, exploring the possibility that it acts as a marker of a COVID-19-associated hypercoagulability. We review the evidence for a pro-coagulant subtype of disseminated intravascular coagulation, discussing its clinical significance. Finally, we discuss the current evidence surrounding treatment of COVID-19 hypercoagulability, including prophylactic and treatment-dose heparin, thrombolytic agents, antiplatelet agents, and direct thrombin inhibitors, among others. We suggest areas in which further investigation is urgently needed to reduce the startling incidence of thrombosis in this group, a complication no doubt contributing to morbidity and mortality.
Collapse
Affiliation(s)
- John Scott Frazer
- Somerville College, University of Oxford, Woodstock Road, Oxford, OX2 6HD, UK
- Buckinghamshire Healthcare NHS Trust, Aylesbury, UK
| | | |
Collapse
|
26
|
Sriram K, Insel PA. Proteinase-activated receptor 1: A target for repurposing in the treatment of COVID-19? Br J Pharmacol 2020; 177:4971-4974. [PMID: 32639031 PMCID: PMC7361899 DOI: 10.1111/bph.15194] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 06/18/2020] [Accepted: 06/22/2020] [Indexed: 12/13/2022] Open
Abstract
In the search to rapidly identify effective therapies that will mitigate the morbidity and mortality of COVID‐19, attention has been directed towards the repurposing of existing drugs. Candidates for repurposing include drugs that target COVID‐19 pathobiology, including agents that alter angiotensin signalling. Recent data indicate that key findings in COVID‐19 patients include thrombosis and endotheliitis. Activation of proteinase‐activated receptor 1 (PAR1), in particular by the serine protease thrombin, is a critical element in platelet aggregation and coagulation. PAR1 activation also impacts on the actions of other cell types involved in COVID‐19 pathobiology, including endothelial cells, fibroblasts and pulmonary alveolar epithelial cells. Vorapaxar is an approved inhibitor of PAR1, used for treatment of patients with myocardial infarction or peripheral arterial disease. We discuss evidence for a possible beneficial role for vorapaxar in the treatment of COVID‐19 patients and other as‐yet non‐approved antagonists of PAR1 and proteinase‐activated receptor 4 (PAR4). Linked Articles This article is part of a themed issue on The Pharmacology of COVID‐19. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.21/issuetoc
Collapse
Affiliation(s)
- Krishna Sriram
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Paul A Insel
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA.,Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
27
|
van Haren FMP, Page C, Laffey JG, Artigas A, Camprubi-Rimblas M, Nunes Q, Smith R, Shute J, Carroll M, Tree J, Carroll M, Singh D, Wilkinson T, Dixon B. Nebulised heparin as a treatment for COVID-19: scientific rationale and a call for randomised evidence. Crit Care 2020; 24:454. [PMID: 32698853 PMCID: PMC7374660 DOI: 10.1186/s13054-020-03148-2] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 07/03/2020] [Indexed: 12/16/2022] Open
Abstract
Nebulised unfractionated heparin (UFH) has a strong scientific and biological rationale and warrants urgent investigation of its therapeutic potential, for COVID-19-induced acute respiratory distress syndrome (ARDS). COVID-19 ARDS displays the typical features of diffuse alveolar damage with extensive pulmonary coagulation activation resulting in fibrin deposition in the microvasculature and formation of hyaline membranes in the air sacs. Patients infected with SARS-CoV-2 who manifest severe disease have high levels of inflammatory cytokines in plasma and bronchoalveolar lavage fluid and significant coagulopathy. There is a strong association between the extent of the coagulopathy and poor clinical outcomes.The anti-coagulant actions of nebulised UFH limit fibrin deposition and microvascular thrombosis. Trials in patients with acute lung injury and related conditions found inhaled UFH reduced pulmonary dead space, coagulation activation, microvascular thrombosis and clinical deterioration, resulting in increased time free of ventilatory support. In addition, UFH has anti-inflammatory, mucolytic and anti-viral properties and, specifically, has been shown to inactivate the SARS-CoV-2 virus and prevent its entry into mammalian cells, thereby inhibiting pulmonary infection by SARS-CoV-2. Furthermore, clinical studies have shown that inhaled UFH safely improves outcomes in other inflammatory respiratory diseases and also acts as an effective mucolytic in sputum-producing respiratory patients. UFH is widely available and inexpensive, which may make this treatment also accessible for low- and middle-income countries.These potentially important therapeutic properties of nebulised UFH underline the need for expedited large-scale clinical trials to test its potential to reduce mortality in COVID-19 patients.
Collapse
Affiliation(s)
- Frank M P van Haren
- Australian National University, Medical School, Canberra, Australia.
- Intensive Care Unit, the Canberra Hospital, Canberra, Australia.
| | - Clive Page
- Sackler Institute of Pulmonary Pharmacology, King's College London, London, UK
| | - John G Laffey
- Anaesthesia and Intensive Care Medicine, School of Medicine, and Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland
- Department of Anaesthesia, University Hospital Galway, Saolta Hospital Group, Galway, Ireland
| | - Antonio Artigas
- Critical Center, Corporació Sanitaria Parc Tauli , CIBER Enfermedades Respiratorias, Autonomous University of Barcelona, Sabadell, Spain
| | - Marta Camprubi-Rimblas
- Institut d'Investigació I Innovació Parc Tauli (I3PT), CIBER de Enfermedades Respiratorias, Sabadell, Spain
| | - Quentin Nunes
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Roger Smith
- Department of Critical Care Medicine, St Vincent's Hospital, Melbourne, Australia
| | - Janis Shute
- School of Pharmacy and Biomedical Science, University of Portsmouth, Portsmouth, UK
| | - Mary Carroll
- Department of Respiratory Medicine, University of Southampton, Southampton, UK
| | - Julia Tree
- National Infection Service, Public Health England, Porton Down, UK
| | - Miles Carroll
- National Infection Service, Public Health England, Porton Down, UK
| | - Dave Singh
- Medicines Evaluation Unit, University of Manchester, Manchester, UK
| | - Tom Wilkinson
- Department of Respiratory Medicine, University of Southampton, Southampton, UK
| | - Barry Dixon
- Department of Critical Care Medicine, St Vincent's Hospital, Melbourne, Australia
| |
Collapse
|
28
|
Degani G, Altomare A, Digiovanni S, Arosio B, Fritz G, Raucci A, Aldini G, Popolo L. Prothrombin is a binding partner of the human receptor of advanced glycation end products. J Biol Chem 2020; 295:12498-12511. [PMID: 32665403 DOI: 10.1074/jbc.ra120.013692] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/11/2020] [Indexed: 01/02/2023] Open
Abstract
The receptor for advanced glycation end products (RAGE) plays a key role in mammal physiology and in the etiology and progression of inflammatory and oxidative stress-based diseases. In adults, RAGE expression is normally high only in the lung where the protein concentrates in the basal membrane of alveolar Type I epithelial cells. In diseases, RAGE levels increase in the affected tissues and sustain chronic inflammation. RAGE exists as a membrane glycoprotein with an ectodomain, a transmembrane helix, and a short carboxyl-terminal tail, or as a soluble ectodomain that acts as a decoy receptor (sRAGE). VC1 domain is responsible for binding to the majority of RAGE ligands including advanced glycation end products (AGEs), S100 proteins, and HMGB1. To ascertain whether other ligands exist, we analyzed by MS the material pulled down by VC1 from human plasma. Twenty of 295 identified proteins were selected and associated to coagulation and complement processes and to extracellular matrix. Four of them contained a γ-carboxyl glutamic acid (Gla) domain, a calcium-binding module, and prothrombin (PT) was the most abundant. Using MicroScale thermophoresis, we quantified the interaction of PT with VC1 and sRAGE in the absence or presence of calcium that acted as a competitor. PT devoid of the Gla domain (PT des-Gla) did not bind to sRAGE, providing further evidence that the Gla domain is critical for the interaction. Finally, the presence of VC1 delayed plasma clotting in a dose-dependent manner. We propose that RAGE is involved in modulating blood coagulation presumably in conditions of lung injury.
Collapse
Affiliation(s)
- Genny Degani
- Department of Biosciences, University of Milan, Milan, Italy
| | | | | | - Beatrice Arosio
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.,Geriatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico and University of Milan, Via Pace 9, Milan, Italy
| | - Guenter Fritz
- Institute of Microbiology, University of Hohenheim, Stuttgart, Germany
| | - Angela Raucci
- Experimental Cardio-oncology and Cardiovascular Aging Unit, Centro Cardiologico Monzino-IRCCS, Via Carlo Parea, 4, Milan, Italy
| | - Giancarlo Aldini
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Laura Popolo
- Department of Biosciences, University of Milan, Milan, Italy
| |
Collapse
|
29
|
Stoneham SM, Milne KM, Nuttall E, Frew GH, Sturrock BR, Sivaloganathan H, Ladikou EE, Drage S, Phillips B, Chevassut TJ, Eziefula AC. Thrombotic risk in COVID-19: a case series and case-control study. Clin Med (Lond) 2020; 20:e76-e81. [PMID: 32423903 PMCID: PMC7385762 DOI: 10.7861/clinmed.2020-0228] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND A possible association between COVID-19 infection and thrombosis, either as a direct consequence of the virus or as a complication of inflammation, is emerging in the literature. Data on the incidence of venous thromboembolism (VTE) are extremely limited. METHODS We describe three cases of thromboembolism refractory to heparin treatment, the incidence of VTE in an inpatient cohort, and a case-control study to identify risk factors associated with VTE. RESULTS We identified 274 confirmed (208) or probable (66) COVID-19 patients. 21 (7.7%) were diagnosed with VTE. D-dimer was elevated in both cases (confirmed VTE) and controls (no confirmed VTE) but higher levels were seen in confirmed VTE cases (4.1 vs 1.2 μg/mL, p<0.001). CONCLUSION Incidence of VTE is high in patients hospitalised with COVID-19. Urgent clinical trials are needed to evaluate the role of anticoagulation in COVID-19. Monitoring of D-dimer and anti-factor Xa levels may be beneficial in guiding management.
Collapse
Affiliation(s)
- Simon M Stoneham
- Brighton and Sussex University Hospitals NHS Trust, Brighton, UK and Brighton and Sussex Medical School, Falmer, UK
| | - Kate M Milne
- Brighton and Sussex Medical School, Brighton, UK and Royal Sussex County Hospital, Brighton, UK
| | | | - Georgina H Frew
- Brighton and Sussex University Hospitals NHS Trust, Brighton, UK
| | - Beattie Rh Sturrock
- Brighton and Sussex Medical School, Brighton UK and Royal Sussex County Hospital, Brighton, UK
| | - Helena Sivaloganathan
- Brighton and Sussex University Hospitals NHS Trust, Brighton, UK and Brighton and Sussex Medical School, Falmer, UK
| | - Eleni E Ladikou
- Brighton and Sussex University Hospitals NHS Trust, Brighton, UK and Brighton and Sussex Medical School, Falmer, UK
| | - Stephen Drage
- Brighton and Sussex University Hospitals NHS Trust, Brighton, UK
| | - Barbara Phillips
- Brighton and Sussex University Hospitals NHS Trust, Brighton, UK and Brighton and Sussex Medical School, Falmer, UK
| | - Timothy Jt Chevassut
- Brighton and Sussex Medical School, Brighton UK and consultant haematologist, Royal Sussex County Hospital, Brighton, UK
| | - Alice C Eziefula
- Brighton and Sussex University Hospitals NHS Trust, Brighton, UK and Brighton and Sussex Medical School, Falmer, UK
| |
Collapse
|
30
|
Negrin LL, Dedeyan M, Plesser S, Hajdu S. Impact of Polytrauma and Acute Respiratory Distress Syndrome on Markers of Fibrinolysis: A Prospective Pilot Study. Front Med (Lausanne) 2020; 7:194. [PMID: 32582720 PMCID: PMC7280477 DOI: 10.3389/fmed.2020.00194] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 04/23/2020] [Indexed: 11/13/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS), which is associated with major morbidity and high mortality, is commonly developed by polytraumatized patients. Its pathogenesis is complex, and its development is difficult to anticipate, as candidate biomarkers for the prediction of ARDS were found not to be reliable for clinical use. In this prospective study, we assessed the serum antigen levels of tissue plasminogen activator (tPA) and plasminogen activator inhibitor type-1 (PAI-1) of 28 survivors of blunt polytrauma (age ≥18 years; injury severity score ≥16) at admission and on days 1, 3, 5, 7, 10, 14, and 21 of hospitalization. Our results show that these patients presented high mean tPA and PAI-1 antigen levels at admission; despite their decline, these parameters remained elevated for 3 weeks. Over this period, the mean tPA antigen level was higher in polytrauma victims suffering from ARDS than in those without ARDS, whereas the mean PAI-1 level was higher in polytrauma victims sustaining pneumonia than in those without pneumonia. Moreover, in each individual developing ARDS, the polytrauma-related elevated tPA antigen level either continued to rise after admission or suffered a second increase up to the onset of ARDS, declining immediately thereafter. Therefore, our findings support the assessment of serum tPA antigen levels after the initial treatment of polytraumatized patients, as this parameter shows potential as a biomarker for the development of ARDS and for the consequent identification of high-risk individuals.
Collapse
Affiliation(s)
- Lukas L Negrin
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Michel Dedeyan
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Stefan Plesser
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Stefan Hajdu
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
31
|
Guglielmetti G, Quaglia M, Sainaghi PP, Castello LM, Vaschetto R, Pirisi M, Corte FD, Avanzi GC, Stratta P, Cantaluppi V. "War to the knife" against thromboinflammation to protect endothelial function of COVID-19 patients. Crit Care 2020; 24:365. [PMID: 32560665 PMCID: PMC7303575 DOI: 10.1186/s13054-020-03060-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 06/04/2020] [Indexed: 12/22/2022] Open
Abstract
In this viewpoint, we summarize the relevance of thromboinflammation in COVID-19 and discuss potential mechanisms of endothelial injury as a key point for the development of lung and distant organ dysfunction, with a focus on direct viral infection and cytokine-mediated injury. Entanglement between inflammation and coagulation and resistance to heparin provide a rationale to consider other therapeutic approaches in order to preserve endothelial function and limit microthrombosis, especially in severe forms. These strategies include nebulized heparin, N-acetylcysteine, plasma exchange and/or fresh frozen plasma, plasma derivatives to increase the level of endogenous anticoagulants (tissue factor pathway inhibitor, activated protein C, thrombomodulin, antithrombin), dipyridamole, complement blockers, different types of stem cells, and extracellular vesicles. An integrated therapy including these drugs has the potential to improve outcomes in COVID-19.
Collapse
Affiliation(s)
- Gabriele Guglielmetti
- Department of Translational Medicine, School of Medicine of the University of Piemonte Orientale (UPO), Novara, Italy
| | - Marco Quaglia
- Department of Translational Medicine, School of Medicine of the University of Piemonte Orientale (UPO), Novara, Italy
| | - Pier Paolo Sainaghi
- Department of Translational Medicine, School of Medicine of the University of Piemonte Orientale (UPO), Novara, Italy
| | - Luigi Mario Castello
- Department of Translational Medicine, School of Medicine of the University of Piemonte Orientale (UPO), Novara, Italy
| | - Rosanna Vaschetto
- Department of Translational Medicine, School of Medicine of the University of Piemonte Orientale (UPO), Novara, Italy
| | - Mario Pirisi
- Department of Translational Medicine, School of Medicine of the University of Piemonte Orientale (UPO), Novara, Italy
| | - Francesco Della Corte
- Department of Translational Medicine, School of Medicine of the University of Piemonte Orientale (UPO), Novara, Italy
| | - Gian Carlo Avanzi
- Department of Translational Medicine, School of Medicine of the University of Piemonte Orientale (UPO), Novara, Italy
| | - Piero Stratta
- Department of Translational Medicine, School of Medicine of the University of Piemonte Orientale (UPO), Novara, Italy
| | - Vincenzo Cantaluppi
- Department of Translational Medicine, School of Medicine of the University of Piemonte Orientale (UPO), Novara, Italy
| |
Collapse
|
32
|
Carsana L, Sonzogni A, Nasr A, Rossi RS, Pellegrinelli A, Zerbi P, Rech R, Colombo R, Antinori S, Corbellino M, Galli M, Catena E, Tosoni A, Gianatti A, Nebuloni M. Pulmonary post-mortem findings in a series of COVID-19 cases from northern Italy: a two-centre descriptive study. THE LANCET. INFECTIOUS DISEASES 2020; 20:1135-1140. [PMID: 32526193 PMCID: PMC7279758 DOI: 10.1016/s1473-3099(20)30434-5] [Citation(s) in RCA: 943] [Impact Index Per Article: 188.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/07/2020] [Accepted: 05/15/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND COVID-19 is characterised by respiratory symptoms, which deteriorate into respiratory failure in a substantial proportion of cases, requiring intensive care in up to a third of patients admitted to hospital. Analysis of the pathological features in the lung tissues of patients who have died with COVID-19 could help us to understand the disease pathogenesis and clinical outcomes. METHODS We systematically analysed lung tissue samples from 38 patients who died from COVID-19 in two hospitals in northern Italy between Feb 29 and March 24, 2020. The most representative areas identified at macroscopic examination were selected, and tissue blocks (median seven, range five to nine) were taken from each lung and fixed in 10% buffered formalin for at least 48 h. Tissues were assessed with use of haematoxylin and eosin staining, immunohistochemical staining for inflammatory infiltrate and cellular components (including staining with antibodies against CD68, CD3, CD45, CD61, TTF1, p40, and Ki-67), and electron microscopy to identify virion localisation. FINDINGS All cases showed features of the exudative and proliferative phases of diffuse alveolar damage, which included capillary congestion (in all cases), necrosis of pneumocytes (in all cases), hyaline membranes (in 33 cases), interstitial and intra-alveolar oedema (in 37 cases), type 2 pneumocyte hyperplasia (in all cases), squamous metaplasia with atypia (in 21 cases), and platelet-fibrin thrombi (in 33 cases). The inflammatory infiltrate, observed in all cases, was largely composed of macrophages in the alveolar lumina (in 24 cases) and lymphocytes in the interstitium (in 31 cases). Electron microscopy revealed that viral particles were predominantly located in the pneumocytes. INTERPRETATION The predominant pattern of lung lesions in patients with COVID-19 patients is diffuse alveolar damage, as described in patients infected with severe acute respiratory syndrome and Middle East respiratory syndrome coronaviruses. Hyaline membrane formation and pneumocyte atypical hyperplasia are frequent. Importantly, the presence of platelet-fibrin thrombi in small arterial vessels is consistent with coagulopathy, which appears to be common in patients with COVID-19 and should be one of the main targets of therapy. FUNDING None.
Collapse
Affiliation(s)
- Luca Carsana
- Department of Pathology, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Aurelio Sonzogni
- Department of Pathology, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Ahmed Nasr
- Department of Pathology, Papa Giovanni XXIII Hospital, Bergamo, Italy; Department of Pathology, University of Milano-Bicocca, Milan, Italy
| | | | | | - Pietro Zerbi
- Department of Pathology, Papa Giovanni XXIII Hospital, Bergamo, Italy; Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Roberto Rech
- Department of Anaesthesiology and Intensive Care Unit, Luigi Sacco Hospital, Milan, Italy
| | - Riccardo Colombo
- Department of Anaesthesiology and Intensive Care Unit, Luigi Sacco Hospital, Milan, Italy
| | - Spinello Antinori
- Department of Infectious Diseases, Luigi Sacco Hospital, Milan, Italy; Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Mario Corbellino
- Department of Infectious Diseases, Luigi Sacco Hospital, Milan, Italy
| | - Massimo Galli
- Department of Infectious Diseases, Luigi Sacco Hospital, Milan, Italy; Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Emanuele Catena
- Department of Anaesthesiology and Intensive Care Unit, Luigi Sacco Hospital, Milan, Italy
| | - Antonella Tosoni
- Department of Pathology, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Andrea Gianatti
- Department of Pathology, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Manuela Nebuloni
- Department of Pathology, Papa Giovanni XXIII Hospital, Bergamo, Italy; Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy.
| |
Collapse
|
33
|
McCarthy SD, González HE, Higgins BD. Future Trends in Nebulized Therapies for Pulmonary Disease. J Pers Med 2020; 10:E37. [PMID: 32397615 PMCID: PMC7354528 DOI: 10.3390/jpm10020037] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/05/2020] [Accepted: 05/07/2020] [Indexed: 12/15/2022] Open
Abstract
Aerosol therapy is a key modality for drug delivery to the lungs of respiratory disease patients. Aerosol therapy improves therapeutic effects by directly targeting diseased lung regions for rapid onset of action, requiring smaller doses than oral or intravenous delivery and minimizing systemic side effects. In order to optimize treatment of critically ill patients, the efficacy of aerosol therapy depends on lung morphology, breathing patterns, aerosol droplet characteristics, disease, mechanical ventilation, pharmacokinetics, and the pharmacodynamics of cell-drug interactions. While aerosol characteristics are influenced by drug formulations and device mechanisms, most other factors are reliant on individual patient variables. This has led to increased efforts towards more personalized therapeutic approaches to optimize pulmonary drug delivery and improve selection of effective drug types for individual patients. Vibrating mesh nebulizers (VMN) are the dominant device in clinical trials involving mechanical ventilation and emerging drugs. In this review, we consider the use of VMN during mechanical ventilation in intensive care units. We aim to link VMN fundamentals to applications in mechanically ventilated patients and look to the future use of VMN in emerging personalized therapeutic drugs.
Collapse
Affiliation(s)
- Sean D. McCarthy
- Anaesthesia, School of Medicine, National University of Ireland Galway, H91 TK33 Galway, Ireland; (S.D.M.); (H.E.G.)
- Lung Biology Group, Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, H91 TK33 Galway, Ireland
| | - Héctor E. González
- Anaesthesia, School of Medicine, National University of Ireland Galway, H91 TK33 Galway, Ireland; (S.D.M.); (H.E.G.)
- Lung Biology Group, Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, H91 TK33 Galway, Ireland
| | - Brendan D. Higgins
- Physiology, School of Medicine, National University of Ireland Galway, H91 TK33 Galway, Ireland
| |
Collapse
|
34
|
Low-Molecular-Weight Heparin Reduces Ventilation-Induced Lung Injury through Hypoxia Inducible Factor-1α in a Murine Endotoxemia Model. Int J Mol Sci 2020; 21:ijms21093097. [PMID: 32353952 PMCID: PMC7247708 DOI: 10.3390/ijms21093097] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/02/2020] [Accepted: 04/27/2020] [Indexed: 12/18/2022] Open
Abstract
Patients with sepsis frequently require mechanical ventilation (MV) to survive. However, MV has been shown to induce the production of proinflammatory cytokines, causing ventilator-induced lung injury (VILI). It has been demonstrated that hypoxia-inducible factor (HIF)-1α plays a crucial role in inducing both apoptotic and inflammatory processes. Low-molecular-weight heparin (LMWH) has been shown to have anti-inflammatory activities. However, the effects of HIF-1α and LMWH on sepsis-related acute lung injury (ALI) have not been fully delineated. We hypothesized that LMWH would reduce lung injury, production of free radicals and epithelial apoptosis through the HIF-1α pathway. Male C57BL/6 mice were exposed to 6-mL/kg or 30-mL/kg MV for 5 h. Enoxaparin, 4 mg/kg, was administered subcutaneously 30 min before MV. We observed that MV with endotoxemia induced microvascular permeability; interleukin-6, tumor necrosis factor-α, macrophage inflammatory protein-2 and vascular endothelial growth factor protein production; neutrophil infiltration; oxidative loads; HIF-1α mRNA activation; HIF-1α expression; bronchial epithelial apoptosis; and decreased respiratory function in mice (p < 0.05). Endotoxin-induced augmentation of VILI and epithelial apoptosis were reduced in the HIF-1α-deficient mice and in the wild-type mice following enoxaparin administration (p < 0.05). Our data suggest that enoxaparin reduces endotoxin-augmented MV-induced ALI, partially by inhibiting the HIF-1α pathway.
Collapse
|
35
|
Carla A, Pereira B, Boukail H, Audard J, Pinol-Domenech N, De Carvalho M, Blondonnet R, Zhai R, Morand D, Lambert C, Sapin V, Ware LB, Calfee CS, Bastarache JA, Laffey JG, Juffermans NP, Bos LD, Artigas A, Rocco PRM, Matthay MA, McAuley DF, Constantin JM, Jabaudon M. Acute respiratory distress syndrome subphenotypes and therapy responsive traits among preclinical models: protocol for a systematic review and meta-analysis. Respir Res 2020; 21:81. [PMID: 32264897 PMCID: PMC7137453 DOI: 10.1186/s12931-020-01337-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 03/17/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Subphenotypes were recently reported within clinical acute respiratory distress syndrome (ARDS), with distinct outcomes and therapeutic responses. Experimental models have long been used to mimic features of ARDS pathophysiology, but the presence of distinct subphenotypes among preclinical ARDS remains unknown. This review will investigate whether: 1) subphenotypes can be identified among preclinical ARDS models; 2) such subphenotypes can identify some responsive traits. METHODS We will include comparative preclinical (in vivo and ex vivo) ARDS studies published between 2009 and 2019 in which pre-specified therapies were assessed (interleukin (IL)-10, IL-2, stem cells, beta-agonists, corticosteroids, fibroblast growth factors, modulators of the receptor for advanced glycation end-products pathway, anticoagulants, and halogenated agents) and outcomes compared to a control condition. The primary outcome will be a composite of the four key features of preclinical ARDS as per the American Thoracic Society consensus conference (histologic evidence of lung injury, altered alveolar-capillary barrier, lung inflammatory response, and physiological dysfunction). Secondary outcomes will include the single components of the primary composite outcome, net alveolar fluid clearance, and death. MEDLINE, Embase, and Cochrane databases will be searched electronically and data from eligible studies will be extracted, pooled, and analyzed using random-effects models. Individual study reporting will be assessed according to the Animal Research: Reporting of In Vivo Experiments guidelines. Meta-regressions will be performed to identify subphenotypes prior to comparing outcomes across subphenotypes and treatment effects. DISCUSSION This study will inform on the presence and underlying pathophysiological features of subphenotypes among preclinical models of ARDS and should help to determine whether sufficient evidence exists to perform preclinical trials of subphenotype-targeted therapies, prior to potential clinical translation. SYSTEMATIC REVIEW REGISTRATION PROSPERO (ID: CRD42019157236).
Collapse
Affiliation(s)
- Adrien Carla
- Department of Perioperative Medicine, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Bruno Pereira
- Biostatistics Unit, Department of Clinical Research and Innovation (DRCI), CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Hanifa Boukail
- Department of Perioperative Medicine, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Jules Audard
- Department of Perioperative Medicine, CHU Clermont-Ferrand, Clermont-Ferrand, France
- GReD, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, Clermont-Ferrand, France
| | | | | | - Raiko Blondonnet
- Department of Perioperative Medicine, CHU Clermont-Ferrand, Clermont-Ferrand, France
- GReD, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Ruoyang Zhai
- GReD, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Dominique Morand
- Department of Perioperative Medicine, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Céline Lambert
- Biostatistics Unit, Department of Clinical Research and Innovation (DRCI), CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Vincent Sapin
- GReD, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, Clermont-Ferrand, France
- Department of Medical Biochemistry and Molecular Biology, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Lorraine B. Ware
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN USA
| | - Carolyn S. Calfee
- Division of Pulmonary and Critical Care Medicine, Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA USA
| | - Julie A. Bastarache
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN USA
| | - John G. Laffey
- Keenan Research Centre for Biomedical Science, Hospital for Sick Children, Departments of Anesthesia and Critical Care Medicine, St. Michael’s Hospital, Departments of Anesthesia, Physiology and Interdepartmental Division of Critical Care, University of Toronto, Toronto, Canada
- Regenerative Medicine Institute at CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Nicole P. Juffermans
- Department of Intensive Care Medicine, Department of Respiratory Medicine, and Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Lieuwe D. Bos
- Department of Intensive Care Medicine, Department of Respiratory Medicine, and Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Antonio Artigas
- Corporació Sanitaria Parc Tauli, CIBER de Enfermedades Respiratorias, Autonomous University of Barcelona, Barcelona, Spain
| | - Patricia R. M. Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Michael A. Matthay
- Division of Pulmonary and Critical Care Medicine, Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA USA
| | - Daniel F. McAuley
- Wellcome-Wolfson Institute for Experimental Medicine, Queens University Belfast and Regional Intensive Care Unit, Belfast Health and Social Care Trust, Belfast, UK
| | - Jean-Michel Constantin
- Department of Anesthesiology and Critical Care, Sorbonne University, GRC 29, AP-HP, DMU DREAM, Pitié-Salpêtrière Hospital, Paris, France
| | - Matthieu Jabaudon
- Department of Perioperative Medicine, CHU Clermont-Ferrand, Clermont-Ferrand, France
- GReD, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, Clermont-Ferrand, France
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN USA
| | - for the ESICM Translational Biology Group of the Acute Respiratory Failure section
- Department of Perioperative Medicine, CHU Clermont-Ferrand, Clermont-Ferrand, France
- Biostatistics Unit, Department of Clinical Research and Innovation (DRCI), CHU Clermont-Ferrand, Clermont-Ferrand, France
- GReD, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, Clermont-Ferrand, France
- Université Clermont Auvergne, Health Library, Clermont-Ferrand, France
- Department of Medical Biochemistry and Molecular Biology, CHU Clermont-Ferrand, Clermont-Ferrand, France
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN USA
- Division of Pulmonary and Critical Care Medicine, Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN USA
- Keenan Research Centre for Biomedical Science, Hospital for Sick Children, Departments of Anesthesia and Critical Care Medicine, St. Michael’s Hospital, Departments of Anesthesia, Physiology and Interdepartmental Division of Critical Care, University of Toronto, Toronto, Canada
- Regenerative Medicine Institute at CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
- Department of Intensive Care Medicine, Department of Respiratory Medicine, and Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Corporació Sanitaria Parc Tauli, CIBER de Enfermedades Respiratorias, Autonomous University of Barcelona, Barcelona, Spain
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Wellcome-Wolfson Institute for Experimental Medicine, Queens University Belfast and Regional Intensive Care Unit, Belfast Health and Social Care Trust, Belfast, UK
- Department of Anesthesiology and Critical Care, Sorbonne University, GRC 29, AP-HP, DMU DREAM, Pitié-Salpêtrière Hospital, Paris, France
| |
Collapse
|
36
|
Lu X, Wang C, Wu D, Zhang C, Xiao C, Xu F. Quantitative proteomics reveals the mechanisms of hydrogen-conferred protection against hyperoxia-induced injury in type II alveolar epithelial cells. Exp Lung Res 2019; 44:464-475. [PMID: 30973277 DOI: 10.1080/01902148.2019.1601296] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Purpose/Aim: Exposure to hyperoxia leads to lung injury both in vivo and in vitro, molecular hydrogen has been reported to protect against hyperoxia-induced lung injury; however, the underlying molecular mechanisms remain largely unknown. The objective of this study was to characterize differentially regulated proteins and biological processes in hydrogen-treated hyperoxic primary type II alveolar epithelial cells (AECIIs) to elucidate the protective mechanism of hydrogen using quantitative proteomics. Materials and Methods: AECIIs were divided into three groups that were cultured for 24 h in three different conditions: control (21% oxygen), hyperoxia (95% oxygen), and hyperoxia + hydrogen. Morphologic examination, flow cytometric analysis, cell viability assessment and analysis of the expression of apoptosis-associated proteins Bax and Bcl-2 as well as AECI markers (AQP5, T1α) and an AECII marker (SP-C) were performed for each group. The TMT labeling quantitative proteome technique was used to detect changes in the protein expression profile, and bioinformatics analysis was performed. Results: Hydrogen plays a protective role in hyperoxia-induced damage in AECIIs, as evidenced by reduced apoptosis, increased viability and survival, improved morphology, and enhanced transdifferentiation of AECIIs into AECIs. A total of 5782 proteins were identified in our study, of which 162 were significantly altered in abundance after hyperoxia exposure, and 97 were significantly altered in abundance in response to hydrogen treatment. The Gene Ontology and KEGG enrichment analyses identified a large number of proteins and biological processes that may responsible for the protective effect of hydrogen, including VEGFA, PDGFB, IGFBP3, EDN1, NADPH oxidase, the coagulation cascade, etc. Conclusions: Molecular hydrogen protects AECIIs from hyperoxic injury by complex mechanisms involving a variety of proteins and biological processes, such as VEGFA, PDGFB, IGFBP3, EDN1, NADPH oxidase and the coagulation cascade. These findings suggest novel pathways that need to be investigated as possible therapeutic targets for hyperoxia-induced lung injury.
Collapse
Affiliation(s)
- Xue Lu
- a Department of Pediatric Intensive Care Unit , Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders , Chongqing , China.,b China International Science and Technology Cooperation Base of Child Development and Critical Disorders , Chongqing , China.,c Chongqing Key Laboratory of Pediatrics , Chongqing , China
| | - Chao Wang
- a Department of Pediatric Intensive Care Unit , Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders , Chongqing , China.,b China International Science and Technology Cooperation Base of Child Development and Critical Disorders , Chongqing , China.,c Chongqing Key Laboratory of Pediatrics , Chongqing , China
| | - Dan Wu
- a Department of Pediatric Intensive Care Unit , Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders , Chongqing , China.,b China International Science and Technology Cooperation Base of Child Development and Critical Disorders , Chongqing , China.,c Chongqing Key Laboratory of Pediatrics , Chongqing , China
| | - Chao Zhang
- a Department of Pediatric Intensive Care Unit , Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders , Chongqing , China.,b China International Science and Technology Cooperation Base of Child Development and Critical Disorders , Chongqing , China.,c Chongqing Key Laboratory of Pediatrics , Chongqing , China
| | - Changxue Xiao
- a Department of Pediatric Intensive Care Unit , Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders , Chongqing , China.,b China International Science and Technology Cooperation Base of Child Development and Critical Disorders , Chongqing , China.,c Chongqing Key Laboratory of Pediatrics , Chongqing , China
| | - Feng Xu
- a Department of Pediatric Intensive Care Unit , Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders , Chongqing , China.,b China International Science and Technology Cooperation Base of Child Development and Critical Disorders , Chongqing , China.,c Chongqing Key Laboratory of Pediatrics , Chongqing , China
| |
Collapse
|