1
|
Semprasert N, Maneethorn P, Kooptiwut S. The protective effect of imatinib against pancreatic β-cell apoptosis induced by dexamethasone via increased GSTP1 expression and reduced oxidative stress. Sci Rep 2024; 14:17691. [PMID: 39085384 PMCID: PMC11291718 DOI: 10.1038/s41598-024-68429-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024] Open
Abstract
Glucocorticoids (GCs) are known to stimulate pancreatic beta (β)-cell apoptosis via several mechanisms, including oxidative stress. Our previous study suggested an increase in dexamethasone-induced pancreatic β-cell apoptosis via a reduction of glutathione S-transferase P1 (GSTP1), which is an antioxidant enzyme. Imatinib, which is a tyrosine kinase inhibitor, also exerts antioxidant effect. This study aims to test our hypothesis that imatinib would prevent pancreatic β-cell apoptosis induced by dexamethasone via increased GSTP1 expression and reduced oxidative stress. Our results revealed that dexamethasone significantly increased apoptosis in INS-1 cells when compared to the control, and that imatinib significantly decreased INS-1 cell apoptosis induced by dexamethasone. Moreover, dexamethasone significantly increased superoxide production in INS-1 cells when compared to the control; however, imatinib, when combined with dexamethasone, significantly reduced superoxide production in INS-1 cells. Dexamethasone significantly decreased GSTP1, p-ERK1/2, and BCL2 protein expression, but significantly increased p-JNK, p-p38, and BAX protein expression in INS-1 cells-all compared to control. Importantly, imatinib significantly ameliorated the effect of dexamethasone on the expression of GSTP1, p-ERK1/2, p-JNK, p-p38 MAPK, BAX, and BCL2. Furthermore-6-(7-nitro-2,1,3-benzoxadiazol-4-ylthio) hexanol (NBDHEX), which is a GSTP1 inhibitor, neutralized the protective effect of imatinib against pancreatic β-cell apoptosis induced by dexamethasone. In conclusion, imatinib decreases pancreatic β-cell apoptosis induced by dexamethasone via increased GSTP1 expression and reduced oxidative stress.
Collapse
Affiliation(s)
- Namoiy Semprasert
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Prannok Road, Bangkoknoi, Bangkok, 10700, Thailand
| | - Petcharee Maneethorn
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Prannok Road, Bangkoknoi, Bangkok, 10700, Thailand
| | - Suwattanee Kooptiwut
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Prannok Road, Bangkoknoi, Bangkok, 10700, Thailand.
| |
Collapse
|
2
|
McMullan RR, McAuley DF, O'Kane CM, Silversides JA. Vascular leak in sepsis: physiological basis and potential therapeutic advances. Crit Care 2024; 28:97. [PMID: 38521954 PMCID: PMC10961003 DOI: 10.1186/s13054-024-04875-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 03/14/2024] [Indexed: 03/25/2024] Open
Abstract
Sepsis is a life-threatening condition characterised by endothelial barrier dysfunction and impairment of normal microcirculatory function, resulting in a state of hypoperfusion and tissue oedema. No specific pharmacological therapies are currently used to attenuate microvascular injury. Given the prominent role of endothelial breakdown and microcirculatory dysfunction in sepsis, there is a need for effective strategies to protect the endothelium. In this review we will discuss key mechanisms and putative therapeutic agents relevant to endothelial barrier function.
Collapse
Affiliation(s)
- Ross R McMullan
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Lisburn Road, Belfast, BT9 7BL, UK.
| | - Daniel F McAuley
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Lisburn Road, Belfast, BT9 7BL, UK
- Department of Critical Care, Belfast Health and Social Care Trust, Belfast, UK
| | - Cecilia M O'Kane
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Lisburn Road, Belfast, BT9 7BL, UK
| | - Jonathan A Silversides
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Lisburn Road, Belfast, BT9 7BL, UK
- Department of Critical Care, Belfast Health and Social Care Trust, Belfast, UK
| |
Collapse
|
3
|
Dechtman ID, Ankory R, Sokolinsky K, Krasner E, Weiss L, Gal Y. Clinically Evaluated COVID-19 Drugs with Therapeutic Potential for Biological Warfare Agents. Microorganisms 2023; 11:1577. [PMID: 37375079 PMCID: PMC10304720 DOI: 10.3390/microorganisms11061577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/10/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) outbreak resulted in hundreds of millions of coronavirus cases, as well as millions of deaths worldwide. Coronavirus Disease 2019 (COVID-19), the disease resulting from exposure to this pathogen, is characterized, among other features, by a pulmonary pathology, which can progress to "cytokine storm", acute respiratory distress syndrome (ARDS), respiratory failure and death. Vaccines are the unsurpassed strategy for prevention and protection against the SARS-CoV-2 infection. However, there is still an extremely high number of severely ill people from at-risk populations. This may be attributed to waning immune response, variant-induced breakthrough infections, unvaccinated population, etc. It is therefore of high importance to utilize pharmacological-based treatments, despite the progression of the global vaccination campaign. Until the approval of Paxlovid, an efficient and highly selective anti-SARS-CoV-2 drug, and the broad-spectrum antiviral agent Lagevrio, many pharmacological-based countermeasures were, and still are, being evaluated in clinical trials. Some of these are host-directed therapies (HDTs), which modulate the endogenic response against the virus, and therefore may confer efficient protection against a wide array of pathogens. These could potentially include Biological Warfare Agents (BWAs), exposure to which may lead to mass casualties due to disease severity and a possible lack of efficient treatment. In this review, we assessed the recent literature on drugs under advanced clinical evaluation for COVID-19 with broad spectrum activity, including antiviral agents and HDTs, which may be relevant for future coping with BWAs, as well as with other agents, in particular respiratory infections.
Collapse
Affiliation(s)
- Ido-David Dechtman
- Pulmonology Department, Edith Wolfson Medical Center, 62 Halochamim Street, Holon 5822012, Israel;
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ran Ankory
- The Israel Defense Force Medical Corps, Tel Hashomer, Ramat Gan, Military Post 02149, Israel;
| | - Keren Sokolinsky
- Chemical, Biological, Radiological and Nuclear Defense Division, Ministry of Defense, HaKirya, Tel Aviv 61909, Israel; (K.S.); (E.K.)
| | - Esther Krasner
- Chemical, Biological, Radiological and Nuclear Defense Division, Ministry of Defense, HaKirya, Tel Aviv 61909, Israel; (K.S.); (E.K.)
| | - Libby Weiss
- Chemical, Biological, Radiological and Nuclear Defense Division, Ministry of Defense, HaKirya, Tel Aviv 61909, Israel; (K.S.); (E.K.)
| | - Yoav Gal
- Chemical, Biological, Radiological and Nuclear Defense Division, Ministry of Defense, HaKirya, Tel Aviv 61909, Israel; (K.S.); (E.K.)
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness Ziona 74100, Israel
| |
Collapse
|
4
|
Atmowihardjo LN, Schippers JR, Duijvelaar E, Bartelink IH, Bet PM, Swart NEL, van Rein N, Purdy K, Cavalla D, McElroy A, Fritchley S, Vonk Noordegraaf A, Endeman H, van Velzen P, Koopmans M, Bogaard HJ, Heunks L, Juffermans N, Schultz MJ, Tuinman PR, Bos LDJ, Aman J. Efficacy and safety of intravenous imatinib in COVID-19 ARDS: a randomized, double-blind, placebo-controlled clinical trial. Crit Care 2023; 27:226. [PMID: 37291677 DOI: 10.1186/s13054-023-04516-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/31/2023] [Indexed: 06/10/2023] Open
Abstract
PURPOSE A hallmark of acute respiratory distress syndrome (ARDS) is hypoxaemic respiratory failure due to pulmonary vascular hyperpermeability. The tyrosine kinase inhibitor imatinib reversed pulmonary capillary leak in preclinical studies and improved clinical outcomes in hospitalized COVID-19 patients. We investigated the effect of intravenous (IV) imatinib on pulmonary edema in COVID-19 ARDS. METHODS This was a multicenter, randomized, double-blind, placebo-controlled trial. Invasively ventilated patients with moderate-to-severe COVID-19 ARDS were randomized to 200 mg IV imatinib or placebo twice daily for a maximum of seven days. The primary outcome was the change in extravascular lung water index (∆EVLWi) between days 1 and 4. Secondary outcomes included safety, duration of invasive ventilation, ventilator-free days (VFD) and 28-day mortality. Posthoc analyses were performed in previously identified biological subphenotypes. RESULTS 66 patients were randomized to imatinib (n = 33) or placebo (n = 33). There was no difference in ∆EVLWi between the groups (0.19 ml/kg, 95% CI - 3.16 to 2.77, p = 0.89). Imatinib treatment did not affect duration of invasive ventilation (p = 0.29), VFD (p = 0.29) or 28-day mortality (p = 0.79). IV imatinib was well-tolerated and appeared safe. In a subgroup of patients characterized by high IL-6, TNFR1 and SP-D levels (n = 20), imatinib significantly decreased EVLWi per treatment day (- 1.17 ml/kg, 95% CI - 1.87 to - 0.44). CONCLUSIONS IV imatinib did not reduce pulmonary edema or improve clinical outcomes in invasively ventilated COVID-19 patients. While this trial does not support the use of imatinib in the general COVID-19 ARDS population, imatinib reduced pulmonary edema in a subgroup of patients, underscoring the potential value of predictive enrichment in ARDS trials. Trial registration NCT04794088 , registered 11 March 2021. European Clinical Trials Database (EudraCT number: 2020-005447-23).
Collapse
Affiliation(s)
- Leila N Atmowihardjo
- Intensive Care, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Job R Schippers
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Location VUmc, Room number 5A-074, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Erik Duijvelaar
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Location VUmc, Room number 5A-074, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Imke H Bartelink
- Department of Clinical Pharmacology and Pharmacy, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1117, Amsterdam, The Netherlands
| | - Pierre M Bet
- Department of Clinical Pharmacology and Pharmacy, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1117, Amsterdam, The Netherlands
| | - Noortje E L Swart
- Department of Clinical Pharmacology and Pharmacy, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1117, Amsterdam, The Netherlands
- Department of Clinical Pharmacology and Pharmacy, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Nienke van Rein
- Department of Clinical Pharmacology and Pharmacy, Leiden UMC, Albinusdreef 2, Leiden, The Netherlands
| | | | | | | | | | - Anton Vonk Noordegraaf
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Location VUmc, Room number 5A-074, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Henrik Endeman
- Intensive Care, Erasmus University Medical Centre, Doctor Molewaterplein 40, Rotterdam, The Netherlands
| | - Patricia van Velzen
- Intensive Care, Dijklander Hospital, Location Purmerend, Waterlandlaan 250, Purmerend, The Netherlands
| | - Matty Koopmans
- Intensive Care, OLVG Hospital Location Oost, Oosterpark 9, Amsterdam, The Netherlands
| | - Harm Jan Bogaard
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Location VUmc, Room number 5A-074, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Leo Heunks
- Intensive Care, Erasmus University Medical Centre, Doctor Molewaterplein 40, Rotterdam, The Netherlands
| | - Nicole Juffermans
- Intensive Care, OLVG Hospital Location Oost, Oosterpark 9, Amsterdam, The Netherlands
- Laboratory of Translational Intensive Care, Erasmus University, Rotterdam, The Netherlands
| | - Marcus J Schultz
- Intensive Care, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Mahidol Oxford Tropical Medicine Research Unit (MORU), Mahidol University, Bangkok, Thailand
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Research and Development, Hamilton Medical AG, Chur, Switzerland
| | - Pieter R Tuinman
- Intensive Care, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1117, Amsterdam, The Netherlands
| | - Lieuwe D J Bos
- Intensive Care, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Location VUmc, Room number 5A-074, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Jurjan Aman
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Location VUmc, Room number 5A-074, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
5
|
Xin Y, Cereda M, Yehya N, Humayun S, Delvecchio P, Thompson JM, Martin K, Hamedani H, Martorano P, Duncan I, Kadlecek S, Makvandi M, Brenner JS, Rizi RR. Imatinib alleviates lung injury and prolongs survival in ventilated rats. Am J Physiol Lung Cell Mol Physiol 2022; 322:L866-L872. [PMID: 35438574 PMCID: PMC9142156 DOI: 10.1152/ajplung.00006.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/28/2022] [Accepted: 04/13/2022] [Indexed: 11/22/2022] Open
Abstract
Imatinib, a tyrosine kinase inhibitor, attenuates pulmonary edema and inflammation in lung injury. However, the physiological effects of this drug and their impact on outcomes are poorly characterized. Using serial computed tomography (CT), we tested the hypothesis that imatinib reduces injury severity and improves survival in ventilated rats. Hydrochloric acid (HCl) was instilled in the trachea (pH 1.5, 2.5 mL/kg) of anesthetized, intubated supine rats. Animals were randomized (n = 17 each group) to receive intraperitoneal imatinib or vehicle immediately prior to HCl. All rats then received mechanical ventilation. CT was performed hourly for 4 h. Images were quantitatively analyzed to assess the progression of radiological abnormalities. Injury severity was confirmed via hourly blood gases, serum biomarkers, bronchoalveolar lavage (BAL), and histopathology. Serial blood drug levels were measured in a subset of rats. Imatinib reduced mortality while delaying functional and radiological injury progression: out of 17 rats per condition, 2 control vs. 8 imatinib-treated rats survived until the end of the experiment (P = 0.02). Imatinib attenuated edema after lung injury (P < 0.05), and survival time in both groups was negatively correlated with increased lung mass (R2 = 0.70) as well as other physiological and CT parameters. Capillary leak (BAL protein concentration) was significantly lower in the treated group (P = 0.04). Peak drug concentration was reached after 70 min, and the drug half-life was 150 min. Imatinib decreased both mortality and lung injury severity in mechanically ventilated rats. Pharmacological inhibition of edema could be used during mechanical ventilation to improve the severity and outcome of lung injury.
Collapse
Affiliation(s)
- Yi Xin
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Maurizio Cereda
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nadir Yehya
- Pediatric Sepsis Program and Division of Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Shiraz Humayun
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Paolo Delvecchio
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jill M Thompson
- Pediatric Sepsis Program and Division of Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Kevin Martin
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hooman Hamedani
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Paul Martorano
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ian Duncan
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Stephen Kadlecek
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mehran Makvandi
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jacob S Brenner
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rahim R Rizi
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
6
|
Lapuente JP, Gómez G, Marco-Brualla J, Fernández P, Desportes P, Sanz J, García-Gil M, Bermejo F, San Martín JV, Algaba A, De Gregorio JC, Lapuente D, De Gregorio A, Lapuente B, Gómez S, Andrés MDLV, Anel A. Evaluation in a Cytokine Storm Model in Vivo of the Safety and Efficacy of Intravenous Administration of PRS CK STORM (Standardized Conditioned Medium Obtained by Coculture of Monocytes and Mesenchymal Stromal Cells). Biomedicines 2022; 10:biomedicines10051094. [PMID: 35625831 PMCID: PMC9138962 DOI: 10.3390/biomedicines10051094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/02/2022] [Accepted: 05/05/2022] [Indexed: 11/16/2022] Open
Abstract
Our research group has been developing a series of biological drugs produced by coculture techniques with M2-polarized macrophages with different primary tissue cells and/or mesenchymal stromal cells (MSC), generally from fat, to produce anti-inflammatory and anti-fibrotic effects, avoiding the overexpression of pro-inflammatory cytokines by the innate immune system at a given time. One of these products is the drug PRS CK STORM, a medium conditioned by allogenic M2-polarized macrophages, from coculture, with those macrophages M2 with MSC from fat, whose composition, in vitro safety, and efficacy we studied. In the present work, we publish the results obtained in terms of safety (pharmacodynamics and pharmacokinetics) and efficacy of the intravenous application of this biological drug in a murine model of cytokine storm associated with severe infectious processes, including those associated with COVID-19. The results demonstrate the safety and high efficacy of PRS CK STORM as an intravenous drug to prevent and treat the cytokine storm associated with infectious processes, including COVID-19.
Collapse
Affiliation(s)
- Juan Pedro Lapuente
- R4T Molecular and Cell Biology Research Laboratories, Fuenlabrada Hospital, 28942 Madrid, Spain; (G.G.); (P.F.); (J.C.D.G.); (D.L.); (A.D.G.); (B.L.); (S.G.); (M.d.l.V.A.)
- Correspondence: (J.P.L.); (A.A.)
| | - Gonzalo Gómez
- R4T Molecular and Cell Biology Research Laboratories, Fuenlabrada Hospital, 28942 Madrid, Spain; (G.G.); (P.F.); (J.C.D.G.); (D.L.); (A.D.G.); (B.L.); (S.G.); (M.d.l.V.A.)
| | - Joaquín Marco-Brualla
- Group Immunity, Cancer and Stem Cells, Faculty of Sciences, University of Zaragoza, 50009 Zaragoza, Spain;
| | - Pablo Fernández
- R4T Molecular and Cell Biology Research Laboratories, Fuenlabrada Hospital, 28942 Madrid, Spain; (G.G.); (P.F.); (J.C.D.G.); (D.L.); (A.D.G.); (B.L.); (S.G.); (M.d.l.V.A.)
| | - Paula Desportes
- GMP Facility, Peaches Biotech, 28050 Madrid, Spain; (P.D.); (J.S.)
| | - Jara Sanz
- GMP Facility, Peaches Biotech, 28050 Madrid, Spain; (P.D.); (J.S.)
| | | | - Fernando Bermejo
- Digestive Department, Fuenlabrada Hospital, 28942 Madrid, Spain;
- Medicine Department, University Rey Juan Carlos, 28942 Madrid, Spain
| | | | - Alicia Algaba
- Clinical Assay Department, Fuelabrada Hospital, 28942 Madrid, Spain;
| | - Juan Carlos De Gregorio
- R4T Molecular and Cell Biology Research Laboratories, Fuenlabrada Hospital, 28942 Madrid, Spain; (G.G.); (P.F.); (J.C.D.G.); (D.L.); (A.D.G.); (B.L.); (S.G.); (M.d.l.V.A.)
| | - Daniel Lapuente
- R4T Molecular and Cell Biology Research Laboratories, Fuenlabrada Hospital, 28942 Madrid, Spain; (G.G.); (P.F.); (J.C.D.G.); (D.L.); (A.D.G.); (B.L.); (S.G.); (M.d.l.V.A.)
| | - Almudena De Gregorio
- R4T Molecular and Cell Biology Research Laboratories, Fuenlabrada Hospital, 28942 Madrid, Spain; (G.G.); (P.F.); (J.C.D.G.); (D.L.); (A.D.G.); (B.L.); (S.G.); (M.d.l.V.A.)
| | - Belén Lapuente
- R4T Molecular and Cell Biology Research Laboratories, Fuenlabrada Hospital, 28942 Madrid, Spain; (G.G.); (P.F.); (J.C.D.G.); (D.L.); (A.D.G.); (B.L.); (S.G.); (M.d.l.V.A.)
| | - Sergio Gómez
- R4T Molecular and Cell Biology Research Laboratories, Fuenlabrada Hospital, 28942 Madrid, Spain; (G.G.); (P.F.); (J.C.D.G.); (D.L.); (A.D.G.); (B.L.); (S.G.); (M.d.l.V.A.)
| | - María de las Viñas Andrés
- R4T Molecular and Cell Biology Research Laboratories, Fuenlabrada Hospital, 28942 Madrid, Spain; (G.G.); (P.F.); (J.C.D.G.); (D.L.); (A.D.G.); (B.L.); (S.G.); (M.d.l.V.A.)
| | - Alberto Anel
- Group Immunity, Cancer and Stem Cells, Faculty of Sciences, University of Zaragoza, 50009 Zaragoza, Spain;
- Correspondence: (J.P.L.); (A.A.)
| |
Collapse
|
7
|
Hawez A, Ding Z, Taha D, Madhi R, Rahman M, Thorlacius H. c-Abl kinase regulates neutrophil extracellular trap formation and lung injury in abdominal sepsis. J Transl Med 2022; 102:263-271. [PMID: 34732849 PMCID: PMC8860741 DOI: 10.1038/s41374-021-00683-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 08/23/2021] [Accepted: 10/07/2021] [Indexed: 11/14/2022] Open
Abstract
Sepsis is associated with exaggerated neutrophil responses although mechanisms remain elusive. The aim of this study was to investigate the role of c-Abelson (c-Abl) kinase in neutrophil extracellular trap (NET) formation and inflammation in septic lung injury. Abdominal sepsis was induced by cecal ligation and puncture (CLP). NETs were detected by electron microscopy in the lung and by confocal microscopy in vitro. Plasma levels of DNA-histone complexes, interleukin-6 (IL-6) and CXC chemokines were quantified. CLP-induced enhanced phosphorylation of c-Abl kinase in circulating neutrophils. Administration of the c-Abl kinase inhibitor GZD824 not only abolished activation of c-Abl kinase in neutrophils but also reduced NET formation in the lung and plasma levels of DNA-histone complexes in CLP mice. Moreover, inhibition of c-Abl kinase decreased CLP-induced lung edema and injury. Administration of GDZ824 reduced CLP-induced increases in the number of alveolar neutrophils. Inhibition of c-Abl kinase also markedly attenuated levels of CXC chemokines in the lung and plasma as well as IL-6 levels in the plasma of septic animals. Taken together, this study demonstrates that c-Abl kinase is a potent regulator of NET formation and we conclude that c-Abl kinase might be a useful target to ameliorate lung damage in abdominal sepsis.
Collapse
Affiliation(s)
- Avin Hawez
- Department of Clinical Sciences, Malmö, Section for Surgery, Skåne University Hospital, Lund University, 205 02, Malmö, Sweden
| | - Zhiyi Ding
- Department of Clinical Sciences, Malmö, Section for Surgery, Skåne University Hospital, Lund University, 205 02, Malmö, Sweden
| | - Dler Taha
- Department of Clinical Sciences, Malmö, Section for Surgery, Skåne University Hospital, Lund University, 205 02, Malmö, Sweden
| | - Raed Madhi
- Department of Clinical Sciences, Malmö, Section for Surgery, Skåne University Hospital, Lund University, 205 02, Malmö, Sweden
| | - Milladur Rahman
- Department of Clinical Sciences, Malmö, Section for Surgery, Skåne University Hospital, Lund University, 205 02, Malmö, Sweden
| | - Henrik Thorlacius
- Department of Clinical Sciences, Malmö, Section for Surgery, Skåne University Hospital, Lund University, 205 02, Malmö, Sweden.
| |
Collapse
|
8
|
Atmowihardjo L, Schippers JR, Bartelink IH, Bet PM, van Rein N, Purdy K, Cavalla D, Comberiati V, McElroy A, Snape SD, Bogaard HJ, Heunks L, Juffermans N, Schultz M, Tuinman PR, Bos LDJ, Aman J. The INVENT COVID trial: a structured protocol for a randomized controlled trial investigating the efficacy and safety of intravenous imatinib mesylate (Impentri®) in subjects with acute respiratory distress syndrome induced by COVID-19. Trials 2022; 23:158. [PMID: 35172891 PMCID: PMC8848942 DOI: 10.1186/s13063-022-06055-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 01/27/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) pandemic has led to a disruptive increase in the number of intensive care unit (ICU) admissions with acute respiratory distress syndrome (ARDS). ARDS is a severe, life-threatening medical condition characterized by widespread inflammation and vascular leak in the lungs. Although there is no proven therapy to reduce pulmonary vascular leak in ARDS, recent studies demonstrated that the tyrosine kinase inhibitor imatinib reinforces the endothelial barrier and prevents vascular leak in inflammatory conditions, while leaving the immune response intact. METHODS This is a randomized, double-blind, parallel-group, placebo-controlled, multicenter clinical trial of intravenous (IV) imatinib mesylate in 90 mechanically ventilated subjects with COVID-19-induced ARDS. Subjects are 18 years or older, admitted to the ICU for mechanical ventilation, meeting the Berlin criteria for moderate-severe ARDS with a positive polymerase chain reaction test for SARS-CoV2. Participants will be randomized in a 1:1 ratio to either imatinib (as mesylate) 200 mg bis in die (b.i.d.) or placebo IV infusion for 7 days, or until ICU discharge or death. The primary study outcome is the change in Extravascular Lung Water Index (EVLWi) between day 1 and day 4. Secondary outcome parameters include changes in oxygenation and ventilation parameters, duration of invasive mechanical ventilation, number of ventilator-free days during the 28-day study period, length of ICU stay, and mortality during 28 days after randomization. Additional secondary parameters include safety, tolerability, and pharmacokinetics. DISCUSSION The current study aims to investigate the efficacy and safety of IV imatinib in mechanically ventilated subjects with COVID-19-related ARDS. We hypothesize that imatinib decreases pulmonary edema, as measured by extravascular lung water using a PiCCO catheter. The reduction in pulmonary edema may reverse hypoxemic respiratory failure and hasten recovery. As pulmonary edema is an important contributor to ARDS, we further hypothesize that imatinib reduces disease severity, reflected by a reduction in 28-day mortality, duration of mechanical ventilation, and ICU length of stay. TRIAL STATUS Protocol version and date: V3.1, 16 April 2021. Recruitment started on 09 March 2021. Estimated recruitment period of approximately 40 weeks. TRIAL REGISTRATION ClinicalTrials.gov NCT04794088 . Registered on 11 March 2021.
Collapse
Affiliation(s)
- Leila Atmowihardjo
- Dept. of Intensive Care, Amsterdam UMC location AMC, Amsterdam, The Netherlands
| | - Job R. Schippers
- Dept. of Pulmonology, Amsterdam UMC location VUMC, Amsterdam, The Netherlands
| | - Imke H. Bartelink
- Hospital Pharmacy, Amsterdam UMC location VUMC, Amsterdam, The Netherlands
| | - Pierre M. Bet
- Hospital Pharmacy, Amsterdam UMC location VUMC, Amsterdam, The Netherlands
| | - Nienke van Rein
- Hospital Pharmacy, Amsterdam UMC location VUMC, Amsterdam, The Netherlands
| | | | | | | | | | | | - Harm Jan Bogaard
- Dept. of Pulmonology, Amsterdam UMC location VUMC, Amsterdam, The Netherlands
| | - Leo Heunks
- Dept. of Intensive Care, Amsterdam UMC, location VUMC, Amsterdam, The Netherlands
| | - Nicole Juffermans
- Dept. of Intensive Care, Onze Lieve Vrouwe Gasthuis (OLVG), Amsterdam, The Netherlands
| | - Marcus Schultz
- Dept. of Intensive Care, Amsterdam UMC location AMC, Amsterdam, The Netherlands
| | - Pieter R. Tuinman
- Dept. of Intensive Care, Amsterdam UMC, location VUMC, Amsterdam, The Netherlands
| | - Lieuwe D. J. Bos
- Dept. of Pulmonology, Amsterdam UMC location VUMC, Amsterdam, The Netherlands
| | - Jurjan Aman
- Dept. of Pulmonology, Amsterdam UMC location VUMC, Amsterdam, The Netherlands
| |
Collapse
|
9
|
Morales-Ortega A, García de Tena J, Frutos-Pérez B, Jaenes-Barrios B, Farfán-Sedano AI, Canales-Albendea MÁ, Bernal-Bello D. COVID-19 in patients with hematological malignancies: Considering the role of tyrosine kinase inhibitors. Cancer 2021; 127:1937-1938. [PMID: 33721325 PMCID: PMC8250378 DOI: 10.1002/cncr.33432] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 12/22/2020] [Indexed: 01/09/2023]
Affiliation(s)
| | | | - Begoña Frutos-Pérez
- Department of Internal Medicine, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | | | | | | | - David Bernal-Bello
- Department of Internal Medicine, Hospital Universitario de Fuenlabrada, Madrid, Spain
| |
Collapse
|
10
|
Nejat R, Sadr AS. Are losartan and imatinib effective against SARS-CoV2 pathogenesis? A pathophysiologic-based in silico study. In Silico Pharmacol 2020; 9:1. [PMID: 33294307 PMCID: PMC7716628 DOI: 10.1007/s40203-020-00058-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/04/2020] [Indexed: 12/13/2022] Open
Abstract
Proposing a theory about the pathophysiology of cytokine storm in COVID19, we were to find the potential drugs to treat this disease and to find any effect of these drugs on the virus infectivity through an in silico study. COVID-19-induced ARDS is linked to a cytokine storm phenomenon not explainable solely by the virus infectivity. Knowing that ACE2, the hydrolyzing enzyme of AngII and SARS-CoV2 receptor, downregulates when the virus enters the host cells, we hypothesize that hyperacute AngII upregulation is the eliciting factor of this ARDS. We were to validate this theory through reviewing previous studies to figure out the role of overzealous activation of AT1R in ARDS. According to this theory losartan may attenuate ARDS in this disease. Imatinib, has previously been elucidated to be promising in modulating lung inflammatory reactions and virus infectivity in SARS and MERS. We did an in silico study to uncover any probable other unconsidered inhibitory effects of losartan and imatinib against SARS-CoV2 pathogenesis. Reviewing the literature, we could find that over-activation of AT1R could explain precisely the mechanism of cytokine storm in COVID19. Our in silico study revealed that losartan and imatinib could probably: (1) decline SARS-CoV2 affinity to ACE2. (2) inhibit the main protease and furin, (3) disturb papain-like protease and p38MAPK functions. Our reviewing on renin-angiotensin system showed that overzealous activation of AT1R by hyper-acute excess of AngII due to acute downregulation of ACE2 by SARS-CoV2 explains precisely the mechanism of cytokine storm in COVID-19. Besides, based on our in silico study we concluded that losartan and imatinib are promising in COVID19.
Collapse
Affiliation(s)
- Reza Nejat
- Department of Anesthesiology and Critical Care Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ahmad Shahir Sadr
- Bioinformatics Research Center, Cheragh Medical Institute and Hospital, Kabul, Afghanistan
- Department of Computer Science, Faculty of Mathematical Sciences, Shahid Beheshti University, Tehran, Iran
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran, Iran
- School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| |
Collapse
|
11
|
Horie S, McNicholas B, Rezoagli E, Pham T, Curley G, McAuley D, O'Kane C, Nichol A, Dos Santos C, Rocco PRM, Bellani G, Laffey JG. Emerging pharmacological therapies for ARDS: COVID-19 and beyond. Intensive Care Med 2020; 46:2265-2283. [PMID: 32654006 PMCID: PMC7352097 DOI: 10.1007/s00134-020-06141-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 05/26/2020] [Indexed: 02/06/2023]
Abstract
ARDS, first described in 1967, is the commonest form of acute severe hypoxemic respiratory failure. Despite considerable advances in our knowledge regarding the pathophysiology of ARDS, insights into the biologic mechanisms of lung injury and repair, and advances in supportive care, particularly ventilatory management, there remains no effective pharmacological therapy for this syndrome. Hospital mortality at 40% remains unacceptably high underlining the need to continue to develop and test therapies for this devastating clinical condition. The purpose of the review is to critically appraise the current status of promising emerging pharmacological therapies for patients with ARDS and potential impact of these and other emerging therapies for COVID-19-induced ARDS. We focus on drugs that: (1) modulate the immune response, both via pleiotropic mechanisms and via specific pathway blockade effects, (2) modify epithelial and channel function, (3) target endothelial and vascular dysfunction, (4) have anticoagulant effects, and (5) enhance ARDS resolution. We also critically assess drugs that demonstrate potential in emerging reports from clinical studies in patients with COVID-19-induced ARDS. Several therapies show promise in earlier and later phase clinical testing, while a growing pipeline of therapies is in preclinical testing. The history of unsuccessful clinical trials of promising therapies underlines the challenges to successful translation. Given this, attention has been focused on the potential to identify biologically homogenous subtypes within ARDS, to enable us to target more specific therapies 'precision medicines.' It is hoped that the substantial number of studies globally investigating potential therapies for COVID-19 will lead to the rapid identification of effective therapies to reduce the mortality and morbidity of this devastating form of ARDS.
Collapse
Affiliation(s)
- Shahd Horie
- Lung Biology Group, Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland, Galway, Ireland
| | - Bairbre McNicholas
- Department of Anaesthesia and Intensive Care Medicine, Galway University Hospitals, Galway, Ireland
| | - Emanuele Rezoagli
- Lung Biology Group, Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland, Galway, Ireland
- Department of Medicine and Surgery, University of Milano - Bicocca, Monza, Italy
- Department of Emergency and Intensive Care, San Gerardo Hospital, Monza, Italy
| | - Tài Pham
- Service de médecine Intensive-Réanimation, AP-HP, Hôpital de Bicêtre, Hôpitaux Universitaires Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Ger Curley
- Department of Anaesthesiology, Beaumont Hospital, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Danny McAuley
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland, UK
- Department of Intensive Care Medicine, Royal Victoria Hospital, Belfast, Northern Ireland, UK
| | - Cecilia O'Kane
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Alistair Nichol
- Clinical Research Centre at St Vincent's University Hospital, University College Dublin, Dublin, Ireland
- Australian and New Zealand Intensive Care Research Centre, Monash University, Melbourne, Australia
- Intensive Care Unit, Alfred Hospital, Melbourne, Australia
| | - Claudia Dos Santos
- Keenan Research Centre and Interdepartmental Division of Critical Care, University of Toronto, Toronto, ON, Canada
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Giacomo Bellani
- Department of Medicine and Surgery, University of Milano - Bicocca, Monza, Italy
- Department of Emergency and Intensive Care, San Gerardo Hospital, Monza, Italy
| | - John G Laffey
- Lung Biology Group, Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland, Galway, Ireland.
- Department of Anaesthesia and Intensive Care Medicine, Galway University Hospitals, Galway, Ireland.
| |
Collapse
|
12
|
Huidobro C, Martín-Vicente P, López-Martínez C, Alonso-López I, Amado-Rodríguez L, Crespo I, M Albaiceta G. Cellular and molecular features of senescence in acute lung injury. Mech Ageing Dev 2020; 193:111410. [PMID: 33249191 DOI: 10.1016/j.mad.2020.111410] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 11/20/2020] [Indexed: 12/14/2022]
Abstract
A wide range of insults can trigger acute injury in the lungs, which eventually may lead to respiratory failure and death of patients. Current treatment relies mainly on supportive measures and mechanical ventilation. Even so, survivors frequently develop important sequels that compromise quality of life. In the search for new approaches to prevent and treat acute lung injury, many investigations have focused on molecular and cellular pathways which could exert a pathogenic role in this disease. Herein, we review recent findings in the literature suggesting that cellular senescence could be involved in lung injury and discuss the potential use of senotherapies to prevent disease progression.
Collapse
Affiliation(s)
- Covadonga Huidobro
- Instituto de Investigación Sanitaria del Principado de Asturias, Avenida de Roma s/n, 33011, Oviedo, Spain.
| | - Paula Martín-Vicente
- Instituto de Investigación Sanitaria del Principado de Asturias, Avenida de Roma s/n, 33011, Oviedo, Spain; Centro de Investigación Biomédica en Red (CIBER)-Enfermedades Respiratorias, Calle de Melchor Fernández Almagro, 3, 28029, Madrid, Spain; Instituto Universitario de Oncología del Principado de Asturias, Calle Fernando Bongera s/n, 33006, Oviedo, Spain
| | - Cecilia López-Martínez
- Instituto de Investigación Sanitaria del Principado de Asturias, Avenida de Roma s/n, 33011, Oviedo, Spain; Centro de Investigación Biomédica en Red (CIBER)-Enfermedades Respiratorias, Calle de Melchor Fernández Almagro, 3, 28029, Madrid, Spain; Instituto Universitario de Oncología del Principado de Asturias, Calle Fernando Bongera s/n, 33006, Oviedo, Spain
| | - Inés Alonso-López
- Instituto de Investigación Sanitaria del Principado de Asturias, Avenida de Roma s/n, 33011, Oviedo, Spain; Centro de Investigación Biomédica en Red (CIBER)-Enfermedades Respiratorias, Calle de Melchor Fernández Almagro, 3, 28029, Madrid, Spain; Instituto Universitario de Oncología del Principado de Asturias, Calle Fernando Bongera s/n, 33006, Oviedo, Spain
| | - Laura Amado-Rodríguez
- Instituto de Investigación Sanitaria del Principado de Asturias, Avenida de Roma s/n, 33011, Oviedo, Spain; Unidad de Cuidados Intensivos Cardiológicos, Hospital Universitario Central de Asturias, Oviedo, Avenida de Roma s/n, 33011, Oviedo, Spain
| | - Irene Crespo
- Departamento de Biología Funcional. Universidad de Oviedo, C/ Julián Clavería s/n, 33006, Oviedo, Spain
| | - Guillermo M Albaiceta
- Instituto de Investigación Sanitaria del Principado de Asturias, Avenida de Roma s/n, 33011, Oviedo, Spain; Centro de Investigación Biomédica en Red (CIBER)-Enfermedades Respiratorias, Calle de Melchor Fernández Almagro, 3, 28029, Madrid, Spain; Instituto Universitario de Oncología del Principado de Asturias, Calle Fernando Bongera s/n, 33006, Oviedo, Spain; Unidad de Cuidados Intensivos Cardiológicos, Hospital Universitario Central de Asturias, Oviedo, Avenida de Roma s/n, 33011, Oviedo, Spain; Departamento de Biología Funcional. Universidad de Oviedo, C/ Julián Clavería s/n, 33006, Oviedo, Spain
| |
Collapse
|
13
|
Morales-Ortega A, Bernal-Bello D, Llarena-Barroso C, Frutos-Pérez B, Duarte-Millán MÁ, García de Viedma-García V, Farfán-Sedano AI, Canalejo-Castrillero E, Ruiz-Giardín JM, Ruiz-Ruiz J, San Martín-López JV. Imatinib for COVID-19: A case report. Clin Immunol 2020; 218:108518. [PMID: 32599278 PMCID: PMC7319919 DOI: 10.1016/j.clim.2020.108518] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 06/22/2020] [Accepted: 06/23/2020] [Indexed: 12/21/2022]
|
14
|
Bernal-Bello D, Jaenes-Barrios B, Morales-Ortega A, Ruiz-Giardin JM, García-Bermúdez V, Frutos-Pérez B, Farfán-Sedano AI, de Ancos-Aracil C, Bermejo F, García-Gil M, Zapatero-Gaviria A, San Martín-López JV. Imatinib might constitute a treatment option for lung involvement in COVID-19. Autoimmun Rev 2020; 19:102565. [PMID: 32376403 PMCID: PMC7252139 DOI: 10.1016/j.autrev.2020.102565] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 12/26/2022]
Affiliation(s)
- David Bernal-Bello
- Department of Internal Medicine, Hospital Universitario de Fuenlabrada, Madrid, Spain..
| | | | | | | | | | - Begoña Frutos-Pérez
- Department of Internal Medicine, Hospital Universitario de Fuenlabrada, Madrid, Spain..
| | | | | | - Fernando Bermejo
- Department of Gastroenterology, Hospital Universitario de Fuenlabrada, Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), Madrid, Spain..
| | - Mario García-Gil
- Department of Hospital Pharmacy, Hospital Universitario de Fuenlabrada, Madrid, Spain..
| | | | | |
Collapse
|
15
|
Rizzo AN, Belvitch P, Demeritte R, Garcia JGN, Letsiou E, Dudek SM. Arg mediates LPS-induced disruption of the pulmonary endothelial barrier. Vascul Pharmacol 2020; 128-129:106677. [PMID: 32240815 DOI: 10.1016/j.vph.2020.106677] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 02/23/2020] [Accepted: 03/21/2020] [Indexed: 12/12/2022]
Abstract
Acute Respiratory Distress Syndrome (ARDS) is a devastating disease process that involves dysregulated inflammation and decreased alveolar-capillary barrier function. Despite increased understanding of the pathophysiology, no effective targeted therapies exist to treat ARDS. Recent preclinical studies suggest that the multi-tyrosine kinase inhibitor, imatinib, which targets the Abl kinases c-Abl and Arg, has the potential to restore endothelial dysfunction caused by inflammatory agonists. Prior work demonstrates that imatinib attenuates LPS (lipopolysaccharide)-induced vascular leak and inflammation; however, the mechanisms underlying these effects remain incompletely understood. In the current study, we demonstrate that imatinib inhibits LPS-induced increase in the phosphorylation of CrkL, a specific substrate of Abl kinases, in human pulmonary endothelial cells. Specific silencing of Arg, and not c-Abl, attenuated LPS-induced pulmonary vascular permeability as measured by electrical cellular impedance sensing (ECIS) and gap formation assays. In addition, direct activation of Abl family kinases with the small molecule activator DPH resulted in endothelial barrier disruption that was attenuated by Arg siRNA. In complementary studies to characterize the mechanisms by which Arg mediates endothelial barrier function, Arg silencing was found to inhibit LPS-induced disruption of adherens junctions and phosphorylation of myosin light chains (MLC). Overall, these results characterize the mechanisms by which imatinib protects against LPS-induced endothelial barrier disruption and suggest that Arg inhibition may represent a novel strategy to enhance endothelial barrier function.
Collapse
Affiliation(s)
- Alicia N Rizzo
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Patrick Belvitch
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Regaina Demeritte
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Joe G N Garcia
- Department of Medicine, University of Arizona, Tucson, AZ, United States of America
| | - Eleftheria Letsiou
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Steven M Dudek
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, United States of America.
| |
Collapse
|
16
|
Depression of lncRNA NEAT1 Antagonizes LPS-Evoked Acute Injury and Inflammatory Response in Alveolar Epithelial Cells via HMGB1-RAGE Signaling. Mediators Inflamm 2020; 2020:8019467. [PMID: 32089649 PMCID: PMC7025070 DOI: 10.1155/2020/8019467] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/31/2019] [Accepted: 01/16/2020] [Indexed: 12/18/2022] Open
Abstract
Sepsis-evoked acute lung injury (ALI) and its extreme manifestation, acute respiratory distress syndrome (ARDS), constitute a major cause of mortality in intensive care units. High levels of the long noncoding RNA nuclear paraspeckle assembly transcript 1 (NEAT1) have been positively correlated with increased severity and unfavorable prognoses in patients with sepsis. Nevertheless, the function and molecular mechanism of NEAT1 in ALI remain elusive. In the current study, high levels of NEAT1 were confirmed in lipopolysaccharide- (LPS-) induced ALI mice models and in LPS-stimulated cells from the alveolar epithelial A549 cell line. Intriguingly, cessation of NEAT1 led to increased cell viability and decreased lactate dehydrogenase release, apoptosis, and caspase-3/9 activity, which conferred protection against LPS-induced injury in these cells. NEAT1 inhibition also restrained LPS-evoked transcripts and production of inflammatory cytokines IL-6, IL-1β, and TNF-α. A mechanism analysis corroborated the activation of high-mobility group box1 (HMGB1)/receptors for advanced glycation end products (RAGE) and NF-κB signaling in LPS-treated A549 cells. NEAT1 suppression reversed the activation of this pathway. Notably, reactivating HMGB1/RAGE signaling via HMGB1 overexpression blunted the anti-injury and anti-inflammation effects of NEAT1 knockdown. These findings suggest that NEAT1 may aggravate the progression of ALI and ARDS by inducing alveolar epithelial cell injury and inflammation via HMGB1/RAGE signaling, implying a promising treatment target for these conditions.
Collapse
|
17
|
Meegan JE, Shaver CM, Putz ND, Jesse JJ, Landstreet SR, Lee HNR, Sidorova TN, McNeil JB, Wynn JL, Cheung-Flynn J, Komalavilas P, Brophy CM, Ware LB, Bastarache JA. Cell-free hemoglobin increases inflammation, lung apoptosis, and microvascular permeability in murine polymicrobial sepsis. PLoS One 2020; 15:e0228727. [PMID: 32012200 PMCID: PMC6996826 DOI: 10.1371/journal.pone.0228727] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 01/22/2020] [Indexed: 12/28/2022] Open
Abstract
Increased endothelial permeability is central to the pathogenesis of sepsis and leads to organ dysfunction and death but the endogenous mechanisms that drive increased endothelial permeability are not completely understood. We previously reported that cell-free hemoglobin (CFH), elevated in 80% of patients with sepsis, increases lung microvascular permeability in an ex vivo human lung model and cultured endothelial cells. In this study, we augmented a murine model of polymicrobial sepsis with elevated circulating CFH to test the hypothesis that CFH increases microvascular endothelial permeability by inducing endothelial apoptosis. Mice were treated with an intraperitoneal injection of cecal slurry with or without a single intravenous injection of CFH. Severity of illness, mortality, systemic and lung inflammation, endothelial injury and dysfunction and lung apoptosis were measured at selected time points. We found that CFH added to CS increased sepsis mortality, plasma inflammatory cytokines as well as lung apoptosis, edema and inflammation without affecting large vessel reactivity or vascular injury marker concentrations. These results suggest that CFH is an endogenous mediator of increased endothelial permeability and apoptosis in sepsis and may be a promising therapeutic target.
Collapse
Affiliation(s)
- Jamie E. Meegan
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Ciara M. Shaver
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Nathan D. Putz
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Jordan J. Jesse
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Stuart R. Landstreet
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Han Noo Ri Lee
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Tatiana N. Sidorova
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - J. Brennan McNeil
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - James L. Wynn
- Departments of Pediatrics, Pathology, Immunology, and Experimental Medicine, University of Florida Health, Gainesville, FL, United States of America
| | - Joyce Cheung-Flynn
- Division of Vascular Surgery, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Padmini Komalavilas
- Division of Vascular Surgery, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Colleen M. Brophy
- Division of Vascular Surgery, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Lorraine B. Ware
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Julie A. Bastarache
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States of America
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States of America
| |
Collapse
|
18
|
Ma X, Zhang Y, Jiang D, Yang Y, Wu G, Wu Z. Protective Effects of Functional Amino Acids on Apoptosis, Inflammatory Response, and Pulmonary Fibrosis in Lipopolysaccharide-Challenged Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:4915-4922. [PMID: 31001980 DOI: 10.1021/acs.jafc.9b00942] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Lung injury is a complicated and lethal condition characterized by alveolar barrier disruption, pulmonary edema, enhanced inflammation, and apoptosis in alveoli. However, therapeutic strategies to ameliorate lung injury without exerting side effects are not available. Functional amino acids have been shown to have anti-inflammatory and anti-apoptotic effects under various conditions. The objective of this study was to test the hypothesis that arginine, glutamine, or glycine supplementation ameliorated lipopolysaccharide (LPS)-induced lung injury in mice. Mice pretreated with aerosolized arginine, glutamine, or glycine were exposed to aerosolized LPS to induce lung injury. Results showed that arginine or glycine pretreatment beneficially reduced LPS-induced collagen deposition, apoptosis of alveolar cells, expression of inflammatory cytokines and chemokines, and accumulation of neutrophils and macrophages in lung tissues of mice, thus contributing to improved alveolar integrity and function. Glutamine administration reduced LPS-induced collagen deposition and inflammatory cytokines without affecting any other parameters examined in the study. Our findings indicated that arginine or glycine pretreatment effectively alleviated LPS-induced lung injury by inhibiting the accumulation of lymphocytes, the release of inflammatory cytokines and chemokines, and the apoptosis of alveolar cells. Supplementation of arginine or glycine may be a novel nutritional strategy to reduce deleterious effects of bacterial infection on alveolar function.
Collapse
Affiliation(s)
- Xiaoshi Ma
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science , China Agricultural University , Beijing 100193 , China
| | - Yunchang Zhang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science , China Agricultural University , Beijing 100193 , China
| | - Da Jiang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science , China Agricultural University , Beijing 100193 , China
| | - Ying Yang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science , China Agricultural University , Beijing 100193 , China
| | - Guoyao Wu
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science , China Agricultural University , Beijing 100193 , China
- Department of Animal Science , Texas A&M University , College Station , Texas 77843 , United States
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science , China Agricultural University , Beijing 100193 , China
| |
Collapse
|
19
|
Zaki OS, Safar MM, Ain-Shoka AA, Rashed LA. A Novel Role of a Chemotherapeutic Agent in a Rat Model of Endotoxemia: Modulation of the STAT-3 Signaling Pathway. Inflammation 2018; 41:20-32. [PMID: 28871508 DOI: 10.1007/s10753-017-0659-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Sepsis caused by lipopolysaccharide (LPS) is a life-threatening disease accompanied by multiple organ failure. This study investigated the curative effects of imatinib (IMA) against hepatic, renal, and pulmonary responses caused by a single administration of LPS (10 mg/kg, i.p.) in rats. Treatment with IMA (15 mg/kg, i.p.) 30 min after LPS antagonized the LPS-induced boost of liver enzymes (ALT, AST), kidney functions (BUN, sCr) as well as the elevated pulmonary vascular permeability and edema. IMA declined tissue contents of NF-κB, STAT-3, P38-MAPK, TNF-α, IL-1β, and iNOS. It also amplified the anti-inflammatory cytokine IL-10 as well as the Bcl-2/Bax ratio, a cardinal indicator of the anti-apoptotic effect. Meanwhile, the rats exhibited marked reduction of the broncho-alveolar lavage fluid (BALF) contents of TNF-α, IL-1β, IFN-γ, and neutrophil count; however, they revealed prominent augmentation of the BALF content IL-10. In conclusion, these findings suggest that IMA is endowed with anti-inflammatory, anti-oxidant, and anti-apoptotic properties and hence may provide a novel agent for the management of sepsis.
Collapse
Affiliation(s)
- Omnia S Zaki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for technology & information (MTI), Cairo, Egypt.
| | - Marwa M Safar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, The British University in Egypt (BUE), Cairo, Egypt
| | - Afaf A Ain-Shoka
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Laila A Rashed
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
20
|
Magruder JT, Grimm JC, Crawford TC, Johnston L, Santhanam L, Stephens RS, Berkowitz DE, Shah AS, Bush EL, Damarla M, Damico RL, Hassoun PM, Kim BS. Imatinib Is Protective Against Ischemia-Reperfusion Injury in an Ex Vivo Rabbit Model of Lung Injury. Ann Thorac Surg 2017; 105:950-956. [PMID: 29289364 DOI: 10.1016/j.athoracsur.2017.10.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 09/21/2017] [Accepted: 10/02/2017] [Indexed: 01/28/2023]
Abstract
BACKGROUND Ischemia-reperfusion injury is characterized by an increase in oxidative stress and leads to significant morbidity and death. The tyrosine kinase c-Abl is activated by oxidative stress and mediates processes that affect endothelial barrier function. We hypothesized treatment with the c-Abl inhibitor imatinib would be protective against ischemia-reperfusion injury in our ex vivo rabbit model. METHODS Heart-lung blocs were harvested from rabbits and stored in cold in Perfadex (Vitrolife, Englewood, CO) for 18 hours. Blocs were reperfused for 2 hours in an ex vivo circuit with donor rabbit blood alone (untreated group, n = 7) or donor rabbit blood and 4 mg imatinib (treatment group, n = 10). Serial clinical variables measured every 15 minutes (arterial oxygen and carbon dioxide tension and mean pulmonary artery pressures) and biochemistry of tissue samples before and after reperfusion were assessed. RESULTS Compared with untreated lungs, imatinib treatment improved physiologic parameters, including oxygen, carbon dioxide, and pulmonary artery pressures. Imatinib-treated lungs had less vascular barrier dysfunction as quantified by wet-to-dry weight ratios and bronchoalveolar lavage protein concentrations. Treated lungs showed less inflammation as measured by bronchoalveolar lavage myeloperoxidase assay, less mitochondrial reactive oxygen species production, and increased antioxidant catalase levels. Finally, imatinib protected lungs from DNA damage and p53 upregulation. CONCLUSIONS Imatinib treatment significantly improved the physiologic performance of reperfused lungs and biochemical indicators associated with reperfusion injury in this ex vivo model. Further study is necessary to elucidate the mechanism of tyrosine kinase inhibition in lungs exposed to ischemia and reperfusion.
Collapse
Affiliation(s)
- J Trent Magruder
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University, Baltimore, Maryland
| | - Joshua C Grimm
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University, Baltimore, Maryland
| | - Todd C Crawford
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University, Baltimore, Maryland
| | - Laura Johnston
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Lakshmi Santhanam
- Department of Anesthesia and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland
| | - R Scott Stephens
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Dan E Berkowitz
- Department of Cardiac Surgery, Vanderbilt University, Nashville, Tennessee
| | - Ashish S Shah
- Department of Cardiac Surgery, Vanderbilt University, Nashville, Tennessee
| | - Errol L Bush
- Division of Thoracic Surgery, Department of Surgery, Johns Hopkins University, Baltimore, Maryland
| | - Mahendra Damarla
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Rachel L Damico
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Paul M Hassoun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Bo S Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland.
| |
Collapse
|
21
|
Tu GW, Shi Y, Zheng YJ, Ju MJ, He HY, Ma GG, Hao GW, Luo Z. Glucocorticoid attenuates acute lung injury through induction of type 2 macrophage. J Transl Med 2017; 15:181. [PMID: 28851381 PMCID: PMC5576304 DOI: 10.1186/s12967-017-1284-7] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 08/22/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are severe inflammatory lung diseases. Methylprednisolone (MP) is a common drug against inflammation in clinic. In this study, we aim to investigate the protective effect of MP on ALI and potential mechanisms. METHODS Male BABL/c mice were injected through tail vein using lipopolysaccharide (LPS, 5 mg/kg) with or without 5 mg/kg MP. Lung mechanics, tissue injury and inflammation were examined. Macrophage subsets in the lung were identified by flow cytometry. Macrophages were cultured from bone marrow of mice with or without MP. Then, we analyzed and isolated the subsets of macrophages. These isolated macrophages were then co-cultured with CD4+ T cells, and the percentage of regulatory T cells (Tregs) was examined. The expression of IL-10 and TGF-β in the supernatant was measured. The Tregs immunosuppression function was examined by T cell proliferation assay. To disclose the mechanism of the induction of Tregs by M2c, we blocked IL-10 or/and TGF-β using neutralizing antibody. RESULTS Respiratory physiologic function was significantly improved by MP treatment. Tissue injury and inflammation were ameliorated in the MP-treated group. After MP treatment, the number of M1 decreased and M2 increased in the lung. In in vitro experiment, MP promoted M2 polarization rather than M1. We then induced M1, M2a and M2c from bone marrow cells. M1 induced more Th17 while M2 induced more CD4+CD25+Fxop3+ Tregs. Compared with M2a, M2c induced more Tregs, and this effect could be blocked by anti-IL-10 and anti-TGF-β antibodies. However, M2a and M2c have no impact on Tregs immunosuppression function. CONCLUSION In conclusion, MP ameliorated ALI by promoting M2 polarization. M2, especially M2c, induced Tregs without any influence on Tregs immunosuppression function.
Collapse
Affiliation(s)
- Guo-wei Tu
- 0000 0001 0125 2443grid.8547.eDepartment of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032 People’s Republic of China
| | - Yi Shi
- 0000 0001 0125 2443grid.8547.eBiomedical Research Center, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Yi-jun Zheng
- 0000 0001 0125 2443grid.8547.eDepartment of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032 People’s Republic of China
| | - Min-jie Ju
- 0000 0001 0125 2443grid.8547.eDepartment of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032 People’s Republic of China
| | - Hong-yu He
- 0000 0001 0125 2443grid.8547.eDepartment of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032 People’s Republic of China
| | - Guo-guang Ma
- 0000 0001 0125 2443grid.8547.eDepartment of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032 People’s Republic of China
| | - Guang-wei Hao
- 0000 0001 0125 2443grid.8547.eDepartment of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032 People’s Republic of China
| | - Zhe Luo
- 0000 0001 0125 2443grid.8547.eDepartment of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032 People’s Republic of China
| |
Collapse
|
22
|
Letsiou E, Sammani S, Wang H, Belvitch P, Dudek SM. Parkin regulates lipopolysaccharide-induced proinflammatory responses in acute lung injury. Transl Res 2017; 181:71-82. [PMID: 27693468 DOI: 10.1016/j.trsl.2016.09.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 08/18/2016] [Accepted: 09/03/2016] [Indexed: 12/12/2022]
Abstract
The acute respiratory distress syndrome (ARDS) is a serious condition resulting from direct or indirect lung injury that is associated with high mortality and morbidity. A key biological event in the pathogenesis of the acute lung injury (ALI) that causes acute respiratory distress syndrome is activation of the lung endothelium cells (ECs), which is triggered by a variety of inflammatory insults leading to barrier disruption and excessive accumulation of neutrophils. Recently, we demonstrated that imatinib protects against lipopolysaccharide (LPS)-induced EC activation by inhibiting c-Abl kinase. In the present study, we explored the role of parkin, a novel c-Abl substrate, in ALI. Parkin is an E3 ubiquitin ligase originally characterized in the pathogenesis of Parkinson disease; however, its potential role in acute inflammatory processes and lung EC function remains largely unknown. Using parkin deficient (PARK2-/-) mice, we now demonstrate that parkin mediates LPS-induced ALI. After LPS, PARK2-/- mice have reduced total protein and cell levels in bronchoalveolar lavage (BAL) compared to wild type. Moreover, in LPS-treated PARK2-/- lungs, the sequestration and activation of neutrophils and release of inflammatory cytokines (interleukin 6 [IL-6], tumor necrosis factor alpha [TNF-α]) are significantly reduced. The BAL levels of soluble VCAM-1 and ICAM-1 are also decreased in LPS-treated PARK2-/- mice compared to wild type. In cultured human lung endothelial cells, downregulation of parkin by small interfering RNA decreases LPS-induced VCAM-1 expression, IL-8 and IL-6 secretion, and NF-kB phosphorylation. These results suggest a previously unidentified role of parkin in mediating endotoxin-induced endothelial proinflammatory signaling and indicate that it may play a critical role in acute inflammation.
Collapse
Affiliation(s)
- Eleftheria Letsiou
- Division of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois, Chicago, Ill.
| | - Saad Sammani
- Arizona Health Sciences Center, University of Arizona, Ariz
| | - Huashan Wang
- Division of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois, Chicago, Ill
| | - Patrick Belvitch
- Division of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois, Chicago, Ill
| | - Steven M Dudek
- Division of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois, Chicago, Ill
| |
Collapse
|