51
|
Liu S, Gao S, Yang Z, Zhang P. miR-128-3p reduced acute lung injury induced by sepsis via targeting PEL12. Open Med (Wars) 2021; 16:1109-1120. [PMID: 34430706 PMCID: PMC8345018 DOI: 10.1515/med-2021-0258] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 02/08/2021] [Accepted: 02/25/2021] [Indexed: 01/01/2023] Open
Abstract
Objective Acute lung injury (ALI) caused by sepsis is clinically a syndrome, which is featured by damage to the alveolar epithelium and endothelium. In this study, we employed mice models of cecal ligation and puncture (CLP) and primary mice pulmonary microvascular endothelial cells (MPVECs) in vitro to investigate the effect of miR-128-3p in ALI caused by sepsis. Methods miR-128-3p agomir or randomized control were injected into adult male C57BL/6 mice 1 week before the CLP surgery. We used miR-128-3p agomir or scrambled control to transfect MPVECs and then employed lipopolysaccharide (LPS) stimulation on the cells. Pellino homolog 2 (PELI2) was predicted to be a direct target of miR-128-3p via luciferase reporter assay. MPVECs were cotransfected with lentiviral vector that expressed PELI2 (or empty vector) as well as miR-128-3p-mimics 1 day before LPS stimulation in rescue experiment. Transcriptional activity of caspase-3, cell apoptosis rate, and the expression levels of miR-128-3p, interleukin-1β (IL-1β), interleukin-6 (IL-6), and PELI2 were analyzed. Results Compared with the sham group, the lung of mice in the CLP group showed pulmonary morphological abnormalities, and the expression of IL-6 and IL-1β, caspase-3 activity, and apoptosis rate were significantly upregulated in the CLP group. Inflammatory factor levels and apoptosis rate were also significantly induced by LPS stimulation on MPVECs. Upregulation of miR-128-3p effectively inhibited sepsis-induced ALI, apoptosis as well as inflammation. miR-128-3p also played a role in antiapoptosis and anti-inflammation in MPVECs with LPS treatment. PEL12 upregulation in MPVECs alleviated miR-128-3p-induced caspase-3 activity inhibition and pro-inflammatory factor production. Conclusions miR-128-3p enabled to alleviate sepsis-induced ALI by inhibiting PEL12 expression, indicating a novel treatment strategy of miR-128-3p for sepsis-induced ALI.
Collapse
Affiliation(s)
- Shinan Liu
- Department of Thoracic Surgery, China Tianjin Medical University General Hospital, Tianjin, China
| | - Shuai Gao
- Department of Thoracic Surgery, China Tianjin Medical University General Hospital, Tianjin, China
| | - Zhaoyu Yang
- Department of Thoracic Surgery, China Tianjin Medical University General Hospital, Tianjin, China
| | - Peng Zhang
- Department of Thoracic Surgery, China Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
52
|
Inhibition of microsomal prostaglandin E synthase-1 ameliorates acute lung injury in mice. J Transl Med 2021; 19:340. [PMID: 34372885 PMCID: PMC8351447 DOI: 10.1186/s12967-021-03016-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 07/29/2021] [Indexed: 11/23/2022] Open
Abstract
Background To examine the effects of BI 1029539 (GS-248), a novel selective human microsomal prostaglandin E synthase-1 (mPGES-1) inhibitor, in experimental models of acute lung injury (ALI) and sepsis in transgenic mice constitutively expressing the mPGES1 (Ptges) humanized allele. Methods Series 1: Lipopolysaccharide (LPS)-induced ALI. Mice were randomized to receive vehicle, BI 1029539, or celecoxib. Series 2: Cecal ligation and puncture-induced sepsis. Mice were randomized to receive vehicle or BI 1029539. Results Series 1: BI 1029539 or celecoxib reduced LPS-induced lung injury, with reduction in neutrophil influx, protein content, TNF-ɑ, IL-1β and PGE2 levels in bronchoalveolar lavage (BAL), myeloperoxidase activity, expression of mPGES-1, cyclooxygenase (COX)-2 and intracellular adhesion molecule in lung tissue compared with vehicle-treated mice. Notably, prostacyclin (PGI2) BAL concentration was only lowered in celecoxib-treated mice. Series 2: BI 1029539 significantly reduced sepsis-induced BAL inflammatory cell recruitment, lung injury score and lung expression of mPGES-1 and inducible nitric oxide synthase. Treatment with BI 1029539 also significantly prolonged survival of mice with severe sepsis. Anti-inflammatory and anti-migratory effect of BI 1029539 was confirmed in peripheral blood leukocytes from healthy volunteers. Conclusions BI 1029539 ameliorates leukocyte infiltration and lung injury resulting from both endotoxin-induced and sepsis-induced lung injury. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03016-9.
Collapse
|
53
|
Rocha NN, Samary CS, Antunes MA, Oliveira MV, Hemerly MR, Santos PS, Capelozzi VL, Cruz FF, Marini JJ, Silva PL, Pelosi P, Rocco PRM. The impact of fluid status and decremental PEEP strategy on cardiac function and lung and kidney damage in mild-moderate experimental acute respiratory distress syndrome. Respir Res 2021; 22:214. [PMID: 34330283 PMCID: PMC8323327 DOI: 10.1186/s12931-021-01811-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 07/26/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND We evaluated the effects of abrupt versus gradual PEEP decrease, combined with standard versus high-volume fluid administration, on cardiac function, as well as lung and kidney damage in an established model of mild-moderate acute respiratory distress syndrome (ARDS). METHODS Wistar rats received endotoxin intratracheally. After 24 h, they were treated with Ringer's lactate at standard (10 mL/kg/h) or high (30 mL/kg/h) dose. For 30 min, all animals were mechanically ventilated with tidal volume = 6 mL/kg and PEEP = 9 cmH2O (to keep alveoli open), then randomized to undergo abrupt or gradual (0.2 cmH2O/min for 30 min) PEEP decrease from 9 to 3 cmH2O. Animals were then further ventilated for 10 min at PEEP = 3 cmH2O, euthanized, and their lungs and kidneys removed for molecular biology analysis. RESULTS At the end of the experiment, left and right ventricular end-diastolic areas were greater in animals treated with high compared to standard fluid administration, regardless of PEEP decrease rate. However, pulmonary arterial pressure, indicated by the pulmonary acceleration time (PAT)/pulmonary ejection time (PET) ratio, was higher in abrupt compared to gradual PEEP decrease, independent of fluid status. Animals treated with high fluids and abrupt PEEP decrease exhibited greater diffuse alveolar damage and higher expression of interleukin-6 (a pro-inflammatory marker) and vascular endothelial growth factor (a marker of endothelial cell damage) compared to the other groups. The combination of standard fluid administration and gradual PEEP decrease increased zonula occludens-1 expression, suggesting epithelial cell preservation. Expression of club cell-16 protein, an alveolar epithelial cell damage marker, was higher in abrupt compared to gradual PEEP decrease groups, regardless of fluid status. Acute kidney injury score and gene expression of kidney injury molecule-1 were higher in the high versus standard fluid administration groups, regardless of PEEP decrease rate. CONCLUSION In the ARDS model used herein, decreasing PEEP abruptly increased pulmonary arterial hypertension, independent of fluid status. The combination of abrupt PEEP decrease and high fluid administration led to greater lung and kidney damage. This information adds to the growing body of evidence that supports gradual transitioning of ventilatory patterns and warrants directing additional investigative effort into vascular and deflation issues that impact lung protection.
Collapse
Affiliation(s)
- Nazareth N Rocha
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Physiology and Pharmacology, Biomedical Institute, Niteroi, Brazil
| | - Cynthia S Samary
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Physiotherapy, Faculty of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mariana A Antunes
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Milena V Oliveira
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Matheus R Hemerly
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patrine S Santos
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vera L Capelozzi
- Department of Pathology, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Fernanda F Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - John J Marini
- Division of Pulmonary and Critical Care Medicine, Regions Hospital, University of Minnesota, St. Paul, MN, USA
| | - Pedro L Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paolo Pelosi
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
- San Martino Policlinico Hospital, IRCCS for Oncology and Neurosciences, Genoa, Italy
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
- Laboratory of Pulmonary Investigation, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.
| |
Collapse
|
54
|
Critical Role of Mortalin/GRP75 in Endothelial Cell Dysfunction Associated with Acute Lung Injury. Shock 2021; 54:245-255. [PMID: 31490354 DOI: 10.1097/shk.0000000000001445] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Mortalin/GRP75 (glucose regulated protein 75), a member of heat shock protein 70 family of chaperones, is involved in several cellular processes including proliferation and signaling, and plays a pivotal role in cancer and neurodegenerative disorders. In this study, we sought to determine the role of mortalin/GRP75 in mediating vascular inflammation and permeability linked to the pathogenesis of acute lung injury (ALI). In an aerosolized bacterial lipopolysaccharide inhalation mouse model of ALI, we found that administration of mortalin/GRP75 inhibitor mean kinetic temperature-077, both prophylactically and therapeutically, protected against polymorphonuclear leukocytes influx into alveolar airspaces, microvascular leakage, and expression of pro-inflammatory mediators such as interleukin-1β, E-selectin, and tumor necrosis factor TNFα. Consistent with this, thrombin-induced inflammation in cultured human endothelial cells (EC) was also protected upon before and after treatment with mean kinetic temperature-077. Similar to pharmacological inhibition of mortalin/GRP75, siRNA-mediated depletion of mortalin/GRP75 also blocked thrombin-induced expression of proinflammatory mediators such as intercellular adhesion molecule-1 and vascular adhesion molecule-1. Mechanistic analysis in EC revealed that inactivation of mortalin/GRP75 interfered with the binding of the liberated NF-κB to the DNA, thereby leading to inhibition of downstream expression of adhesion molecules, cytokines, and chemokines. Importantly, thrombin-induced Ca signaling and EC permeability were also prevented upon mortalin/GRP75 inactivation/depletion. Thus, this study provides evidence for a novel role of mortalin/GRP75 in mediating EC inflammation and permeability associated with ALI.
Collapse
|
55
|
Denstaedt SJ, Bustamante AC, Newstead MW, Moore BB, Standiford TJ, Zemans RL, Singer BH. Long-term survivors of murine sepsis are predisposed to enhanced LPS-induced lung injury and proinflammatory immune reprogramming. Am J Physiol Lung Cell Mol Physiol 2021; 321:L451-L465. [PMID: 34161747 DOI: 10.1152/ajplung.00123.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Millions of people who survive sepsis each year are rehospitalized and die due to late pulmonary complications. To prevent and treat these complications, biomarkers and molecular mediators must be identified. Persistent immune reprogramming in the form of immunoparalysis and impaired host defense is proposed to mediate late pulmonary complications after sepsis, particularly new pulmonary infections. However, immune reprogramming may also involve enhanced/primed responses to secondary stimuli, although their contribution to long-term sepsis complications remains understudied. We hypothesize that enhanced/primed immune responses in the lungs of sepsis survivors are associated with late pulmonary complications. To this end, we developed a murine sepsis model using cecal ligation and puncture (CLP) followed 3 wk later by administration of intranasal lipopolysaccharide to induce inflammatory lung injury. Mice surviving sepsis exhibit enhanced lung injury with increased alveolar permeability, neutrophil recruitment, and enhanced Ly6Chi monocyte Tnf expression. To determine the mediators of enhanced lung injury, we performed flow cytometry and RNA sequencing of lungs 3 wk after CLP, prior to lipopolysaccharide. Sepsis survivor mice showed expanded Ly6Chi monocytes populations and increased expression of many inflammatory genes. Of these, S100A8/A9 was also elevated in the circulation of human sepsis survivors for months after sepsis, validating our model and identifying S100A8/A9 as a potential biomarker and therapeutic target for long-term pulmonary complications after sepsis. These data provide new insight into the importance of enhanced/primed immune responses in survivors of sepsis and establish a foundation for additional investigation into the mechanisms mediating this response.
Collapse
Affiliation(s)
- Scott J Denstaedt
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Angela C Bustamante
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Michael W Newstead
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Bethany B Moore
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan.,Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan
| | - Theodore J Standiford
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Rachel L Zemans
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan.,Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, Michigan
| | - Benjamin H Singer
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
56
|
Lin Q, Liang Q, Qin C, Li Y. CircANKRD36 Knockdown Suppressed Cell Viability and Migration of LPS-Stimulated RAW264.7 Cells by Sponging MiR-330. Inflammation 2021; 44:2044-2053. [PMID: 34041646 DOI: 10.1007/s10753-021-01480-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 11/30/2022]
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is an independent risk factor for mortality in patients with sepsis. In this study, we attempt to investigate the molecular mechanism of circANKRD36 underlying sepsis-induced ALI/ARDS in vitro. We first detected the altered circRNAs in serums of patients with sepsis-induced ARDS using circRNAs microarray. CircANKRD36 expression in serums and LPS-stimulated RAW264.7 cells was measured using qRT-PCR. CCK-8, cell migration, ELISA, and qRT-PCR were applied to the evaluation of cell biological behavior and inflammation reaction. The results showed that circANKRD36 expression was significantly elevated in serum of patients with sepsis-induced ARDS. Knockdown of circANKRD36 inhibited cell viability and migration and alleviated inflammation of lipopolysaccharide-stimulated (LPS-stimulated) RAW264.7 cells. Bioinformatic analysis demonstrated that circANKRD36 serves as a sponge for miR-330 and ROCK1 was directly targeted by miR-330. Furthermore, knockdown of circANKRD36 repressed ROCK1 expression by targeting miR-330. In short, circANKRD36 knockdown suppressed cell viability and migration of LPS-stimulated RAW264.7 cells in vitro via sponging miR-330, which may provide new ideas for the treatment of sepsis-induced ARDS.
Collapse
Affiliation(s)
- Qiqing Lin
- Department of Emergency Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, No. 18, Zhongshan Second Road, Yooujiang District, Baise City, 533000, China.
| | - Qiong Liang
- Respiratory Department, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise City, 533000, China
| | - Chunyan Qin
- Respiratory Department, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise City, 533000, China
| | - Yueyong Li
- Department of Interventional Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise City, 533000, China
| |
Collapse
|
57
|
Wang M, Zhong H, Zhang X, Huang X, Wang J, Li Z, Chen M, Xiao Z. EGCG promotes PRKCA expression to alleviate LPS-induced acute lung injury and inflammatory response. Sci Rep 2021; 11:11014. [PMID: 34040072 PMCID: PMC8154949 DOI: 10.1038/s41598-021-90398-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 05/10/2021] [Indexed: 12/11/2022] Open
Abstract
Acute lung injury (ALI), which could be induced by multiple factors such as lipopolysaccharide (LPS), refer to clinical symptoms of acute respiratory failure, commonly with high morbidity and mortality. Reportedly, active ingredients from green tea have anti-inflammatory and anticancer properties, including epigallocatechin-3-gallate (EGCG). In the present study, protein kinase C alpha (PRKCA) is involved in EGCG protection against LPS-induced inflammation and ALI. EGCG treatment attenuated LPS-stimulated ALI in mice as manifested as improved lung injury scores, decreased total cell amounts, neutrophil amounts and macrophage amounts, inhibited the activity of MPO, decreased wet-to-dry weight ratio of lung tissues, and inhibited release of inflammatory cytokines TNF-α, IL-1β, and IL-6. PRKCA mRNA and protein expression showed to be dramatically decreased by LPS treatment while reversed by EGCG treatment. Within LPS-stimulated ALI mice, PRKCA silencing further aggravated, while PRKCA overexpression attenuated LPS-stimulated inflammation and ALI through MAPK signaling pathway. PRKCA silencing attenuated EGCG protection. Within LPS-induced RAW 264.7 macrophages, EGCG could induce PRKCA expression. Single EGCG treatment or Lv-PRKCA infection attenuated LPS-induced increases in inflammatory factors; PRKCA silencing could reverse the suppressive effects of EGCG upon LPS-stimulated inflammatory factor release. In conclusion, EGCG pretreatment inhibits LPS-induced ALI in mice. The protective mechanism might be associated with the inhibitory effects of PRKCA on proinflammatory cytokine release via macrophages and MAPK signaling pathway.
Collapse
Affiliation(s)
- Mian Wang
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Xiangya RD 110, Changsha, 410078, China
- Department of Epidemiology and Health Statistics, School of Public Health, University of South China, Hengyang, 421001, China
| | - Hua Zhong
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xian Zhang
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Xiangya RD 110, Changsha, 410078, China
| | - Xin Huang
- Department of Epidemiology and Health Statistics, Hunan Normal University, Changsha, 410006, China
| | - Jing Wang
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Xiangya RD 110, Changsha, 410078, China
| | - Zihao Li
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Xiangya RD 110, Changsha, 410078, China
| | - Mengshi Chen
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Xiangya RD 110, Changsha, 410078, China.
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Central South University, Changsha, 410078, China.
| | - Zhenghui Xiao
- Hunan Provincial Key Laboratory of Pediatric Emergency, Hunan Children's Hospital, Changsha, 410006, China
| |
Collapse
|
58
|
Pulakurthi YS, Pederson JM, Saravu K, Gupta N, Balasubramanian P, Kamrowski S, Schmidt M, Vegivinti CTR, Dibas M, Reierson NL, Pisipati S, Joseph BA, Selvan PT, Dmytriw AA, Keesari PR, Sriram V, Chittajallu S, Brinjikji W, Katamreddy RR, Chibbar R, Davis AR, Malpe M, Mishra HK, Kallmes KM, Hassan AE, Evanson KW. Corticosteroid therapy for COVID-19: A systematic review and meta-analysis of randomized controlled trials. Medicine (Baltimore) 2021; 100:e25719. [PMID: 34011029 PMCID: PMC8137023 DOI: 10.1097/md.0000000000025719] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 04/11/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Corticosteroid treatment is an effective and common therapeutic strategy for various inflammatory lung pathologies and may be an effective treatment for coronavirus disease 2019 (COVID-19). The purpose of this systematic review and meta-analysis of current literature was to investigate the clinical outcomes associated with corticosteroid treatment of COVID-19. METHODS We systematically searched PubMed, medRxiv, Web of Science, and Scopus databases through March 10, 2021 to identify randomized controlled trials (RCTs) that evaluated the effects of corticosteroid therapies for COVID-19 treatment. Outcomes of interest were mortality, need for mechanical ventilation, serious adverse events (SAEs), and superinfection. RESULTS A total of 7737 patients from 8 RCTs were included in the quantitative meta-analysis, of which 2795 (36.1%) patients received corticosteroids plus standard of care (SOC) while 4942 (63.9%) patients received placebo and/or SOC alone. The odds of mortality were significantly lower in patients that received corticosteroids as compared to SOC (odds ratio [OR] = 0.85 [95% CI: 0.76; 0.95], P = .003). Corticosteroid treatment reduced the odds of a need for mechanical ventilation as compared to SOC (OR = 0.76 [95% CI: 0.59; 0.97], P = .030). There was no significant difference between the corticosteroid and SOC groups with regards to SAEs and superinfections. CONCLUSION Corticosteroid treatment can reduce the odds for mortality and the need for mechanical ventilation in severe COVID-19 patients.
Collapse
Affiliation(s)
| | | | - Kavitha Saravu
- Department of Infectious Diseases, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Nitin Gupta
- Department of Infectious Diseases, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Prasanth Balasubramanian
- Department of Internal Medicine, Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY
| | | | | | - Charan Thej Reddy Vegivinti
- Department of General Medicine, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Mahmoud Dibas
- Sulaiman Al Rajhi University, College of Medicine, Saudi Arabia
| | | | | | | | - Pragadeesh Thamarai Selvan
- Department of General Medicine, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Adam A. Dmytriw
- Neuroradiology & Neurointervention Service, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Varsha Sriram
- Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | | | | | - Rewanth R. Katamreddy
- Department of General Surgery, Kempegowda Institute of Medical Sciences, Bangalore, Karnataka, India
| | - Richa Chibbar
- Department of Medicine, Lakeridge Health, Oshawa, ON, Canada
| | | | | | | | | | - Ameer E. Hassan
- Department of Neuroradiology, Valley Baptist Medical Center-Harlingen, Texas
| | | |
Collapse
|
59
|
Sparrow NA, Anwar F, Covarrubias AE, Rajput PS, Rashid MH, Nisson PL, Gezalian MM, Toossi S, Ayodele MO, Karumanchi SA, Ely EW, Lahiri S. Interleukin-6 Inhibition Reduces Neuronal Injury In A Murine Model of Ventilator-Induced Lung Injury. Am J Respir Cell Mol Biol 2021; 65:403-412. [PMID: 34014798 DOI: 10.1165/rcmb.2021-0072oc] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Mechanical ventilation is a known risk factor for delirium, a cognitive impairment characterized by frontal cortex and hippocampal dysfunction. Although interleukin-6 (IL-6) is upregulated in mechanical ventilation-induced lung injury (VILI) and may contribute to delirium, it is not known whether inhibition of systemic IL-6 mitigates delirium-relevant neuropathology. To histologically define neuropathological effects of IL-6 inhibition in an experimental VILI model. VILI was simulated in anesthetized adult mice using a 35cc/kg tidal volume mechanical ventilation model. There were two controls groups: 1) spontaneously breathing, or 2) anesthetized and mechanically ventilated with 10cc/kg tidal volume to distinguish effects of anesthesia from VILI. Two hours prior to inducing VILI, mice were treated with either anti-IL-6 antibody, anti-IL-6 receptor antibody, or saline. Neuronal injury, stress, and inflammation were assessed using immunohistochemistry. Cleaved caspase-3 (CC3), a neuronal apoptosis marker, was significantly increased in the frontal (p<0.001) and hippocampal (p<0.0001) brain regions and accompanied by significant increases in c-Fos and heat shock protein-90 in the frontal cortices of VILI mice compared to controls (p<0.001). These findings were not related to cerebral hypoxia and there was no evidence of irreversible neuronal death. Frontal and hippocampal neuronal CC3 were significantly reduced with anti-IL-6 antibody (p<0.01 and p<0.0001, respectively), anti-IL-6 receptor antibody (p<0.05 and p<0.0001, respectively) compared to saline VILI mice. VILI induces potentially reversible neuronal injury and inflammation in the frontal cortex and hippocampus, which is mitigated with IL-6 inhibition. These data suggest a potentially novel neuroprotective role of systemic IL-6 inhibition that justifies further investigation.
Collapse
Affiliation(s)
- Nicklaus A Sparrow
- Cedars-Sinai Medical Center, 22494, Neurology, West Hollywood, California, United States
| | - Faizan Anwar
- Cedars-Sinai Medical Center, 22494, Los Angeles, California, United States
| | | | - Padmesh S Rajput
- Cedars-Sinai Health System, 5149, Neurology, Los Angeles, California, United States
| | | | - Peyton L Nisson
- Cedars-Sinai Medical Center, 22494, West Hollywood, California, United States
| | - Michael M Gezalian
- Cedars-Sinai Medical Center, 22494, Neurology, West Hollywood, California, United States
| | - Shahed Toossi
- Cedars-Sinai Medical Center, 22494, West Hollywood, California, United States
| | - Maranatha O Ayodele
- Cedars-Sinai Medical Center, 22494, West Hollywood, California, United States
| | | | - E Wesley Ely
- Vanderbilt University School of Medicine, 12327, Nashville, Tennessee, United States
| | - Shouri Lahiri
- Cedars-Sinai Health System, 5149, Neurology and Neurosurgery, Los Angeles, California, United States;
| |
Collapse
|
60
|
Munoz K, Wasnik S, Abdipour A, Bi H, Wilson SM, Tang X, Ghahramanpouri M, Baylink DJ. The Effects of Insulin-Like Growth Factor I and BTP-2 on Acute Lung Injury. Int J Mol Sci 2021; 22:ijms22105244. [PMID: 34063554 PMCID: PMC8170877 DOI: 10.3390/ijms22105244] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/06/2021] [Accepted: 05/11/2021] [Indexed: 12/24/2022] Open
Abstract
Acute lung injury (ALI) afflicts approximately 200,000 patients annually and has a 40% mortality rate. The COVID-19 pandemic has massively increased the rate of ALI incidence. The pathogenesis of ALI involves tissue damage from invading microbes and, in severe cases, the overexpression of inflammatory cytokines such as tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β). This study aimed to develop a therapy to normalize the excess production of inflammatory cytokines and promote tissue repair in the lipopolysaccharide (LPS)-induced ALI. Based on our previous studies, we tested the insulin-like growth factor I (IGF-I) and BTP-2 therapies. IGF-I was selected, because we and others have shown that elevated inflammatory cytokines suppress the expression of growth hormone receptors in the liver, leading to a decrease in the circulating IGF-I. IGF-I is a growth factor that increases vascular protection, enhances tissue repair, and decreases pro-inflammatory cytokines. It is also required to produce anti-inflammatory 1,25-dihydroxyvitamin D. BTP-2, an inhibitor of cytosolic calcium, was used to suppress the LPS-induced increase in cytosolic calcium, which otherwise leads to an increase in proinflammatory cytokines. We showed that LPS increased the expression of the primary inflammatory mediators such as toll like receptor-4 (TLR-4), IL-1β, interleukin-17 (IL-17), TNF-α, and interferon-γ (IFN-γ), which were normalized by the IGF-I + BTP-2 dual therapy in the lungs, along with improved vascular gene expression markers. The histologic lung injury score was markedly elevated by LPS and reduced to normal by the combination therapy. In conclusion, the LPS-induced increases in inflammatory cytokines, vascular injuries, and lung injuries were all improved by IGF-I + BTP-2 combination therapy.
Collapse
Affiliation(s)
- Kevin Munoz
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.M.); (S.W.); (A.A.); (X.T.); (M.G.)
| | - Samiksha Wasnik
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.M.); (S.W.); (A.A.); (X.T.); (M.G.)
| | - Amir Abdipour
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.M.); (S.W.); (A.A.); (X.T.); (M.G.)
- Division of Nephrology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA
| | - Hongzheng Bi
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, China;
| | - Sean M. Wilson
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA;
| | - Xiaolei Tang
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.M.); (S.W.); (A.A.); (X.T.); (M.G.)
- Department of Veterinary Biomedical Sciences, College of Veterinary Medicine, Long Island University, Brookville, NY 11548, USA
| | - Mahdis Ghahramanpouri
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.M.); (S.W.); (A.A.); (X.T.); (M.G.)
| | - David J. Baylink
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.M.); (S.W.); (A.A.); (X.T.); (M.G.)
- Correspondence: ; Tel.: +909-558-4000-49796; Fax: +(909)-558-0428
| |
Collapse
|
61
|
Ai M, Lin S, Zhang M, Wu T, Yang N, Li Y, Li L. Cirsilineol attenuates LPS-induced inflammation in both in vivo and in vitro models via inhibiting TLR-4/NFkB/IKK signaling pathway. J Biochem Mol Toxicol 2021; 35:e22799. [PMID: 33949057 DOI: 10.1002/jbt.22799] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/25/2021] [Accepted: 04/22/2021] [Indexed: 01/03/2023]
Abstract
The anti-inflammatory activity of cirsilineol in in vivo condition was assessed by measuring the relative organ weight, lung dry/wet weight ratio, protein concentration, and infiltration of inflammatory cells in bronchoalveolar lavage fluid. We estimated the myeloperoxidase activity and levels of cytokines, chemokines, and inflammatory markers to analyze the efficacy of cirsilineol against lipopolysaccharide (LPS)-induced lung inflammation. Furthermore, we quantified the gene expression of NFkB/IKK signaling molecules in cirsilineol-treated and untreated acute lung injury mice to confirm the anti-inflammatory property of cirsilineol. The lung histology was assessed with hematoxylin and eosin staining. Apart from in vivo experiments, in vitro tests with LPS-stimulated RAW 264.7 macrophages were also performed. Cell viability assay was performed in the presence and absence of LPS in RAW 264.7 macrophages to determine the cytotoxic effect of cirsilineol against macrophages. Reverse-transcription polymerase chain reaction (RT-PCR) analysis was done to analyze the gene expression of inflammatory markers in LPS-treated RAW 264.7 macrophages to prove that cirsilineol effectively inhibits inflammation in vitro. The results of our study prove that cirsilineol effectively inhibits inflammation in both in vivo and in vitro conditions. RT-PCR analysis results of NFkB/IKK signaling molecules clearly illustrate that cirsilineol inhibited the expression of NFkB/IKK signaling protein and thereby prevented inflammation in in vivo condition, and it is further confirmed with the results of inflammatory protein expression in vitro model. The lung histopathological studies authentically confirm that cirsilineol potentially prevented the mice from LPS-induced lung inflammation.
Collapse
Affiliation(s)
- Meimei Ai
- Department of Emergency, First Affiliated Hospital of Air Force Military Medical University (Xijing Hospital), Xian, Shaanxi, China
| | - Shusheng Lin
- Department of Intensive Care Unit, Huaian Hospital of Huai'an City, Huai'an, Jiangsu, China
| | - Meixia Zhang
- Department of Nursing, First Affiliated Hospital of Air Force Military Medical University (Xijing Hospital), Xian, Shaanxi, China
| | - Tieli Wu
- Pneumology Department, Jilin Medical College Affiliated Hospital, Jilin, China
| | - Nan Yang
- Day Observation Ward, First Hospital of Jilin University, Changchun, Jilin, China
| | - Yu Li
- Department of Health Examination, Peace Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Li Li
- Department of Emergency, Dongzhimen Hospital, Beijing University of Traditional Chinese Medicine, Beijing, China.,Institute of Sepsis, Beijing University of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
62
|
α1-Antitrypsin: Key Player or Bystander in Acute Respiratory Distress Syndrome? Anesthesiology 2021; 134:792-808. [PMID: 33721888 DOI: 10.1097/aln.0000000000003727] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Acute respiratory distress syndrome is characterized by hypoxemia, altered alveolar-capillary permeability, and neutrophil-dominated inflammatory pulmonary edema. Despite decades of research, an effective drug therapy for acute respiratory distress syndrome remains elusive. The ideal pharmacotherapy for acute respiratory distress syndrome should demonstrate antiprotease activity and target injurious inflammatory pathways while maintaining host defense against infection. Furthermore, a drug with a reputable safety profile, low possibility of off-target effects, and well-known pharmacokinetics would be desirable. The endogenous 52-kd serine protease α1-antitrypsin has the potential to be a novel treatment option for acute respiratory distress syndrome. The main function of α1-antitrypsin is as an antiprotease, targeting neutrophil elastase in particular. However, studies have also highlighted the role of α1-antitrypsin in the modulation of inflammation and bacterial clearance. In light of the current SARS-CoV-2 pandemic, the identification of a treatment for acute respiratory distress syndrome is even more pertinent, and α1-antitrypsin has been implicated in the inflammatory response to SARS-CoV-2 infection.
Collapse
|
63
|
Silva de França F, Villas-Boas IM, Cogliati B, Woodruff TM, Reis EDS, Lambris JD, Tambourgi DV. C5a-C5aR1 Axis Activation Drives Envenomation Immunopathology by the Snake Naja annulifera. Front Immunol 2021; 12:652242. [PMID: 33936074 PMCID: PMC8082402 DOI: 10.3389/fimmu.2021.652242] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/22/2021] [Indexed: 12/03/2022] Open
Abstract
Systemic complement activation drives a plethora of pathological conditions, but its role in snake envenoming remains obscure. Here, we explored complement's contribution to the physiopathogenesis of Naja annulifera envenomation. We found that N. annulifera venom promoted the generation of C3a, C4a, C5a, and the soluble Terminal Complement Complex (sTCC) mediated by the action of snake venom metalloproteinases. N. annulifera venom also induced the release of lipid mediators and chemokines in a human whole-blood model. This release was complement-mediated, since C3/C3b and C5a Receptor 1 (C5aR1) inhibition mitigated the effects. In an experimental BALB/c mouse model of envenomation, N. annulifera venom promoted lipid mediator and chemokine production, neutrophil influx, and swelling at the injection site in a C5a-C5aR1 axis-dependent manner. N. annulifera venom induced systemic complementopathy and increased interleukin and chemokine production, leukocytosis, and acute lung injury (ALI). Inhibition of C5aR1 with the cyclic peptide antagonist PMX205 rescued mice from these systemic reactions and abrogated ALI development. These data reveal hitherto unrecognized roles for complement in envenomation physiopathogenesis, making complement an interesting therapeutic target in envenomation by N. annulifera and possibly by other snake venoms.
Collapse
Affiliation(s)
| | | | - Bruno Cogliati
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Trent M. Woodruff
- Neuroinflammation Laboratory, School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia
| | - Edimara da Silva Reis
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - John D. Lambris
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | | |
Collapse
|
64
|
Bayraktutan Z, Dincer B, Keskin H, Kose D, Bilen A, Toktay E, Sirin B, Halici Z. Roflumilast as a Potential Therapeutic Agent for Cecal Ligation and Puncture-Induced Septic Lung Injury. J INVEST SURG 2021; 35:605-613. [PMID: 33843406 DOI: 10.1080/08941939.2021.1908462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
PURPOSE/AIMS This study focused on delineating the possible effects of roflumilast (ROF), a selective phosphodiesterase 4 (PDE4) inhibitor, in rats with cecal ligation and puncture (CLP)-induced polymicrobial sepsis, and investigated whether ROF can act as a protective agent in sepsis-induced lung damage. MATERIAL AND METHODS Four experimental groups were organized, each comprising eight rats: Control, Sepsis, Sepsis + ROF 0.5 mgkg-1, and Sepsis + ROF 1 mgkg-1 groups. A polymicrobial sepsis model was induced in the rats by cecal ligation and puncture under anesthesia. Twelve hours after sepsis induction, the lungs were obtained for biochemical, molecular, and histopathological analyses. RESULTS In the sepsis group's lungs, the TNF-α, IL-1β, and IL-6 mRNA expression levels peaked in the sepsis group's lung tissues, and ROF significantly decreased these levels compared with the sepsis group dose-dependently. ROF also significantly decreased MDA levels in septic lungs and increased antioxidant parameters (SOD and GSH) compared with the sepsis group. Histopathological analysis results supported biochemical and molecular results. CONCLUSIONS ROF, a PDE4 inhibitor, suppressed the expression levels of pro-inflammatory cytokines, alleviated lung damage (probably by blocking neutrophil infiltration), and increased the capacity of the antioxidant system.
Collapse
Affiliation(s)
| | - Busra Dincer
- Department of Pharmacology, Erzincan Binali Yildirim University, Erzincan, Turkey
| | - Halil Keskin
- Department of Child Health and Diseases, Ataturk University, Erzurum, Turkey
| | - Duygu Kose
- Department of Pharmacology, Ataturk University, Erzurum, Turkey
| | - Arzu Bilen
- Department of Internal Medicine, Ataturk University, Erzurum, Turkey
| | - Erdem Toktay
- Department of Histology and Embryology, Kafkas University, Kars, Turkey
| | - Busra Sirin
- Clinical Research, Development and Design Application and Research Center, Ataturk University, Erzurum, Turkey
| | - Zekai Halici
- Department of Pharmacology, Ataturk University, Erzurum, Turkey.,Department of Internal Medicine, Ataturk University, Erzurum, Turkey.,Department of Histology and Embryology, Kafkas University, Kars, Turkey.,Clinical Research, Development and Design Application and Research Center, Ataturk University, Erzurum, Turkey
| |
Collapse
|
65
|
Kumari M, Mathur P, Aggarwal R, Madan K, Sagar S, Gupta A, Khurana S, Sreenivas V, Kumar S. Changes in extracellular cytokines in predicting disease severity and final clinical outcome of patients with blunt chest trauma. Immunobiology 2021; 226:152087. [PMID: 33857690 DOI: 10.1016/j.imbio.2021.152087] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 03/10/2021] [Accepted: 03/10/2021] [Indexed: 01/04/2023]
Abstract
BACKGROUND Chest trauma causes substantial morbidity and mortality and its severity is assessed using clinical diagnosis or scoring systems like Injury severity score (ISS) and thoracic trauma severity score (TTSS). Association of inflammatory cytokines with severity of disease and final clinical outcome is not clearly defined in patients with chest trauma. In this study, we thought to evaluate the inflammatory response in serum and bronchoalveolar lavage fluid (BALF) in chest trauma patients and correlate the level of extracellular cytokines with diseases severity and final outcome. METHODS A total of 65 patients with blunt chest trauma and 30 healthy controls were enrolled in this prospective observational study. Assessment of inflammatory cytokines such as Interleukin (s) - IL-5, IL-13, IL-2, IL-6, IL-9, IL-1β, IFN-γ, TNF-α, IL-17A, IL-17F,IL-4, IL-21 and IL-22 was performed in both serum and bronchoalveolar lavage fluid using 13-plex multiplex kit using fluorescence-encoded bead based immunoassays. RESULTS A significantly higher level of IL-13, IL-2, IL-6, IL-9, IL-1β, IFN-γ, TNF-α, IL-17A, IL-17F, IL-21 and IL-22 cytokines were observed in patients with blunt chest trauma compared to healthy controls. Level of IL-2, IL-6, IL-1β and IL-17A was significantly raised in the patients with blunt chest trauma who had a fatal outcome during the hospital stay. An elevated cytokine response of IL-13, IL-4, and IL-21 was noted in the group of patients with high (>5) thoracic trauma severity score. CONCLUSION Routine monitoring of the inflammatory cytokine level in patients with chest trauma may be used routinely. Longer prospective studies should be encouraged to determine the role of cytokines in patients with chest trauma in predicting the patient final clinical outcome.
Collapse
Affiliation(s)
- Minu Kumari
- Division of Trauma Surgery & Critical Care, JPN Apex Trauma Center, All India Institute of Medical Sciences, New Delhi, India
| | - Purva Mathur
- Department of Laboratory Medicine, JPN Apex Trauma Center, All India Institute of Medical Sciences, New Delhi, India
| | - Richa Aggarwal
- Division of Trauma Surgery & Critical Care, JPN Apex Trauma Center, All India Institute of Medical Sciences, New Delhi, India
| | - Karan Madan
- Department of Pulmonary Medicine & Sleep Disorders, All India Institute of Medical Sciences, New Delhi, India
| | - Sushma Sagar
- Division of Trauma Surgery & Critical Care, JPN Apex Trauma Center, All India Institute of Medical Sciences, New Delhi, India
| | - Amit Gupta
- Division of Trauma Surgery & Critical Care, JPN Apex Trauma Center, All India Institute of Medical Sciences, New Delhi, India
| | - Surbhi Khurana
- Department of Laboratory Medicine, JPN Apex Trauma Center, All India Institute of Medical Sciences, New Delhi, India
| | | | - Subodh Kumar
- Division of Trauma Surgery & Critical Care, JPN Apex Trauma Center, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
66
|
Lin CK, Huang TH, Yang CT, Shi CS. Roles of lung-recruited monocytes and pulmonary Vascular Endothelial Growth Factor (VEGF) in resolving Ventilator-Induced Lung Injury (VILI). PLoS One 2021; 16:e0248959. [PMID: 33740009 PMCID: PMC7978382 DOI: 10.1371/journal.pone.0248959] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 03/09/2021] [Indexed: 01/31/2023] Open
Abstract
Monocytes and vascular endothelial growth factor (VEGF) have profound effects on tissue injury and repair. In ventilator-induced lung injury (VILI), monocytes, the majority of which are Ly6C+high, and VEGF are known to initiate lung injury. However, their roles in post-VILI lung repair remain unclear. In this study, we used a two-hit mouse model of VILI to identify the phenotypes of monocytes recruited to the lungs during the resolution of VILI and investigated the contributions of monocytes and VEGF to lung repair. We found that the lung-recruited monocytes were predominantly Ly6C+low from day 1 after the insult. Meanwhile, contrary to inflammatory cytokines, pulmonary VEGF decreased upon VILI but subsequently increased significantly on days 7 and 14 after the injury. There was a strong positive correlation between VEGF expression and proliferation of alveolar epithelial cells in lung sections. The expression pattern of VEGF mRNA in lung-recruited monocytes was similar to that of pulmonary VEGF proteins, and the depletion of monocytes significantly suppressed the increase of pulmonary VEGF proteins on days 7 and 14 after VILI. In conclusion, during recovery from VILI, the temporal expression patterns of pulmonary growth factors are different from those of inflammatory cytokines, and the restoration of pulmonary VEGF by monocytes, which are mostly Ly6C+low, is associated with pulmonary epithelial proliferation. Lung-recruited monocytes and pulmonary VEGF may play crucial roles in post-VILI lung repair.
Collapse
Affiliation(s)
- Chin-Kuo Lin
- Division of Pulmonary Infection and Critical Care, Department of Pulmonary and Critical Care Medicine, Chiayi Chang Gung Memorial Hospital, Puzi City, Taiwan
- Graduate Institute of Clinical Medicine Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Tzu-Hsiung Huang
- Department of Respiratory Therapy, Chiayi Chang Gung Memorial Hospital, Puzi City, Taiwan
| | - Cheng-Ta Yang
- Department of Thoracic Medicine, Taoyuan Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Respiratory Therapy, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chung-Sheng Shi
- Graduate Institute of Clinical Medicine Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Colon and Rectal Surgery, Department of Surgery, Chiayi Chang Gung Memorial Hospital, Puzi City, Taiwan
| |
Collapse
|
67
|
Tharmarajah E, Buazon A, Patel V, Hannah JR, Adas M, Allen VB, Bechman K, Clarke BD, Nagra D, Norton S, Russell MD, Rutherford AI, Yates M, Galloway JB. IL-6 inhibition in the treatment of COVID-19: A meta-analysis and meta-regression. J Infect 2021; 82:178-185. [PMID: 33745918 PMCID: PMC7970418 DOI: 10.1016/j.jinf.2021.03.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 03/13/2021] [Indexed: 12/29/2022]
Abstract
Objectives Multiple RCTs of interleukin-6 (IL-6) inhibitors in COVID-19 have been published, with conflicting conclusions. We performed a meta-analysis to assess the impact of IL-6 inhibition on mortality from COVID-19, utilising meta-regression to explore differences in study results. Methods Systematic database searches were performed to identify RCTs comparing IL-6 inhibitors (tocilizumab and sarilumab) to placebo or standard of care in adults with COVID-19. Meta-analysis was used to estimate the relative risk of mortality at 28 days between arms, expressed as a risk ratio. Within-study mortality rates were compared, and meta-regression was used to investigate treatment effect modification. Results Data from nine RCTs were included. The combined mortality rate across studies was 19% (95% CI: 18, 20%), ranging from 2% to 31%. The overall risk ratio for 28-day mortality was 0.90 (95% CI: 0.81, 0.99), in favour of benefit for IL-6 inhibition over placebo or standard of care, with low treatment effect heterogeneity: I2 0% (95% CI: 0, 53%). Meta-regression showed no evidence of treatment effect modification by patient characteristics. Trial-specific mortality rates were explained by known patient-level predictors of COVID-19 outcome (male sex, CRP, hypertension), and country-level COVID-19 incidence. Conclusions IL-6 inhibition is associated with clinically meaningful improvements in outcomes for patients admitted with COVID-19. Long-term benefits of IL-6 inhibition, its effectiveness across healthcare systems, and implications for differing standards of care are currently unknown.
Collapse
Affiliation(s)
- Emmanuel Tharmarajah
- Centre for Rheumatic Diseases, Weston Education Centre, King's College London, 10 Cutcombe Road, London SE5 9RJ, UK
| | - April Buazon
- Centre for Rheumatic Diseases, Weston Education Centre, King's College London, 10 Cutcombe Road, London SE5 9RJ, UK
| | - Vishit Patel
- Centre for Rheumatic Diseases, Weston Education Centre, King's College London, 10 Cutcombe Road, London SE5 9RJ, UK
| | - Jennifer R Hannah
- Centre for Rheumatic Diseases, Weston Education Centre, King's College London, 10 Cutcombe Road, London SE5 9RJ, UK
| | - Maryam Adas
- Centre for Rheumatic Diseases, Weston Education Centre, King's College London, 10 Cutcombe Road, London SE5 9RJ, UK
| | - Victoria B Allen
- Centre for Rheumatic Diseases, Weston Education Centre, King's College London, 10 Cutcombe Road, London SE5 9RJ, UK
| | - Katie Bechman
- Centre for Rheumatic Diseases, Weston Education Centre, King's College London, 10 Cutcombe Road, London SE5 9RJ, UK
| | - Benjamin D Clarke
- Centre for Rheumatic Diseases, Weston Education Centre, King's College London, 10 Cutcombe Road, London SE5 9RJ, UK
| | - Deepak Nagra
- Centre for Rheumatic Diseases, Weston Education Centre, King's College London, 10 Cutcombe Road, London SE5 9RJ, UK
| | - Sam Norton
- Centre for Rheumatic Diseases, Weston Education Centre, King's College London, 10 Cutcombe Road, London SE5 9RJ, UK
| | - Mark D Russell
- Centre for Rheumatic Diseases, Weston Education Centre, King's College London, 10 Cutcombe Road, London SE5 9RJ, UK.
| | - Andrew I Rutherford
- Centre for Rheumatic Diseases, Weston Education Centre, King's College London, 10 Cutcombe Road, London SE5 9RJ, UK
| | - Mark Yates
- Centre for Rheumatic Diseases, Weston Education Centre, King's College London, 10 Cutcombe Road, London SE5 9RJ, UK
| | - James B Galloway
- Centre for Rheumatic Diseases, Weston Education Centre, King's College London, 10 Cutcombe Road, London SE5 9RJ, UK
| |
Collapse
|
68
|
Yoga-An Alternative Form of Therapy in Patients with Blunt Chest Trauma: A Randomized Controlled Trial. World J Surg 2021; 45:2015-2026. [PMID: 33738521 PMCID: PMC7972014 DOI: 10.1007/s00268-021-06057-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2021] [Indexed: 11/03/2022]
Abstract
BACKGROUND Yoga as alternative form of therapy has shown positive impact on pulmonary functions, exercise capacity, behavioral changes, and inflammation in non-trauma patients. However, the efficacy of Yoga has not been studied in chest trauma patients. METHODS This randomized controlled trial was conducted at level-1 Trauma Centre. Isolated chest injury patients were randomized into either standard physiotherapy or Yogatherapy groups. Patients in physiotherapy group received conventional chest physiotherapy and Yogatherapy group received a set of Yogic exercises in addition to conventional chest physiotherapy. Primary outcome measure was changes in pulmonary function tests (PFT) at 4 weeks of discharge. Secondary outcomes were changes in quality of life (QoL), respiratory muscle strength and endurance, chest wall mobility, and levels of cytokines at 4 weeks. Data were analyzed using STATA v14.0. RESULTS A total of 89 eligible patients were randomized to physiotherapy (n = 46) and Yoga therapy (n = 43) groups. Demographic characteristics were comparable in both the groups. There were statistically significant improvements in PFT in the Yogatherapy group compared with physiotherapy with an increase in Forced vital capacity (p = 0.02) and Forced expiratory volume (p = 0.01) at 4 weeks. In addition, there were significant improvement in physical component of QoL, respiratory muscle endurance (p = 0.003) and axillary cirtometry (p = 0.009) in the Yogatherapy group. However, no statistically significant difference in the trends of cytokine markers seen between the groups. CONCLUSION Yoga was found to be effective in improving pulmonary functions and QoL in patients with chest trauma. (Trial registered at ctri.nic.in/clinicaltrials/login.php, numberREF/2016/05/011,287).
Collapse
|
69
|
Mokra D, Mokry J. Phosphodiesterase Inhibitors in Acute Lung Injury: What Are the Perspectives? Int J Mol Sci 2021; 22:1929. [PMID: 33669167 PMCID: PMC7919656 DOI: 10.3390/ijms22041929] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/12/2021] [Accepted: 02/14/2021] [Indexed: 12/14/2022] Open
Abstract
Despite progress in understanding the pathophysiology of acute lung damage, currently approved treatment possibilities are limited to lung-protective ventilation, prone positioning, and supportive interventions. Various pharmacological approaches have also been tested, with neuromuscular blockers and corticosteroids considered as the most promising. However, inhibitors of phosphodiesterases (PDEs) also exert a broad spectrum of favorable effects potentially beneficial in acute lung damage. This article reviews pharmacological action and therapeutical potential of nonselective and selective PDE inhibitors and summarizes the results from available studies focused on the use of PDE inhibitors in animal models and clinical studies, including their adverse effects. The data suggest that xanthines as representatives of nonselective PDE inhibitors may reduce acute lung damage, and decrease mortality and length of hospital stay. Various (selective) PDE3, PDE4, and PDE5 inhibitors have also demonstrated stabilization of the pulmonary epithelial-endothelial barrier and reduction the sepsis- and inflammation-increased microvascular permeability, and suppression of the production of inflammatory mediators, which finally resulted in improved oxygenation and ventilatory parameters. However, the current lack of sufficient clinical evidence limits their recommendation for a broader use. A separate chapter focuses on involvement of cyclic adenosine monophosphate (cAMP) and PDE-related changes in its metabolism in association with coronavirus disease 2019 (COVID-19). The chapter illuminates perspectives of the use of PDE inhibitors as an add-on treatment based on actual experimental and clinical trials with preliminary data suggesting their potential benefit.
Collapse
Affiliation(s)
- Daniela Mokra
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Juraj Mokry
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia;
| |
Collapse
|
70
|
Local administration of p-coumaric acid decreases lipopolysaccharide-induced acute lung injury in mice: In vitro and in silico studies. Eur J Pharmacol 2021; 897:173929. [PMID: 33561444 DOI: 10.1016/j.ejphar.2021.173929] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 01/22/2021] [Accepted: 01/29/2021] [Indexed: 01/21/2023]
Abstract
Acute lung injury (ALI) remains to cause a high rate of mortality in critically ill patients. It is known that inflammation is a key factor in the pathogenesis of lipopolysaccharide (LPS)-induced ALI, which makes it a relevant approach to the treatment of ALI. In this study, we evaluated the potential of nasally instilled p-coumaric acid to prevent LPS-induced ALI in mice, by evaluating its effects on cellular and molecular targets involved in inflammatory response via in vitro and in silico approaches. Our results demonstrated that p-coumaric acid reduced both neutrophil accumulation and pro-inflammatory cytokine abundance, and simultaneously increased IL-10 production at the site of inflammation, potentially contributing to protection against LPS-induced ALI in mice. In the in vitro experiments, we observed inhibitory effects of p-coumaric acid against IL-6 and IL-8 production in stimulated A549 cells, as well as reactive oxygen species generation by neutrophils. In addition, p-coumaric acid treatment decreased neutrophil adhesion on the TNF-α-stimulated endothelial cells. According to the in silico predictions, p-coumaric acid reached stable interactions with both the ATP-binding site of IKKβ as well as the regions within LFA-1, critical for interaction with ICAM-1, thereby suppressing the production of proinflammatory mediators and hindering the neutrophil infiltration, respectively. Collectively, these findings indicate that p-coumaric acid is a promising anti-inflammatory agent that can be used for developing a pharmaceutical drug for the treatment of ALI and other inflammatory disorders.
Collapse
|
71
|
Pan P, Su L, Wang X, Chai W, Liu D, Song L, Xie L. Vimentin regulation of autophagy activation in lung fibroblasts in response to lipopolysaccharide exposure in vitro. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:304. [PMID: 33708931 PMCID: PMC7944268 DOI: 10.21037/atm-20-5129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Background The activation and assembly of the NLRP3 inflammasome is dependent on the interaction between NLRP3 and the intermediate filament protein vimentin in an acute respiratory distress syndrome (ARDS) model. We investigated the role of vimentin in this process using human fetal lung (HFL-1) fibroblasts with vimentin transfer genes or gene knockdown and lipopolysaccharide (LPS) intervention. Methods HFL-1 cells [con-vector + LPS, vimentin-pCMV3 (VIM-pCMV3), con-siRNA, and vimentin siRNA (VIM-siRNA)] were treated with LPS. An oxidative stress damage assessment, apoptosis analysis, and quantification of tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, IL-6, and IL-10 by enzyme linked immunosorbent assay (ELISA) were performed. Immunoblotting was used to reveal the autophagy pathway. Results We demonstrated that in response to LPS vimentin expression was lower in the HFL-1 cells with the vimentin gene knocked down. Specifically, an increase in oxidative stress, a decrease in mitochondrial membrane potential, or an increase in calcium ion permeability resulted in an increase in the fibroblast apoptosis rate. In addition, the inflammatory response after vimentin gene knockout was upregulated, as indicated by higher levels of TNF-a, IL-1β, IL-6, and IL-10. Importantly, the mechanism of suppression of vimentin in the lung fibroblasts was caused by a decrease in autophagy, an increase in mitochondrial membrane protein, and a decrease in mitochondrial function, which may contribute to the augmented cellular injury generated during the response to LPS. Conclusions This study provides insights into whether vimentin may interfere with the inflammatory cascade by activating the autophagy pathway of mitochondrial lung fibroblasts in the early stage of acute lung injury (ALI).
Collapse
Affiliation(s)
- Pan Pan
- College of Pulmonary and Critical Care Medicine, Chinese PLA General Hospital, Beijing, China
| | - Longxiang Su
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoting Wang
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Wenzhao Chai
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Dawei Liu
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Licheng Song
- College of Pulmonary and Critical Care Medicine, Chinese PLA General Hospital, Beijing, China
| | - Lixin Xie
- College of Pulmonary and Critical Care Medicine, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
72
|
α-Ketoglutarate Modulates Macrophage Polarization Through Regulation of PPARγ Transcription and mTORC1/p70S6K Pathway to Ameliorate ALI/ARDS. Shock 2021; 53:103-113. [PMID: 31841452 DOI: 10.1097/shk.0000000000001333] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
As tissue-resident cells in the lung, alveolar macrophages display remarkable heterogeneity and play a crucial role in the development and control of septic acute lung injury/acute respiratory distress syndrome (ALI/ARDS). Recent evidence suggests that α-ketoglutarate (α-KG) plays an important role in alternative activation of macrophage (M2) through metabolic and epigenetic reprogramming, and thus possesses anti-inflammatory properties. However, the underlying mechanisms of α-KG's effect on alveolar macrophage polarization and the potential effects of α-KG in ALI/ARDS remain unclear. Here, we examined the effects and mechanisms of α-KG on alveolar macrophage polarization, and investigated the possible effects of α-KG on lipopolysaccharide (LPS)-induced ALI/ARDS in a mouse model. We found that α-KG inhibited M1 macrophage polarization and promoted IL-4-induced M2 macrophage polarization in MH-S cells (a murine alveolar macrophage cell line). Further experiments showed that α-KG down-regulated the expression of M1-polarized marker genes and inhibited the activities of mammalian target of rapamycin complex 1 (mTORC1)/p70 ribosomal protein S6 kinase (p70S6K) signaling pathway in M1-polarized MH-S cells. Moreover, our results showed that α-KG promoted IL-4-induced M2 polarization of MH-S cells by augmenting nuclear translocation of peroxisome proliferator-activated receptor γ (PPARγ) and increasing expression of relevant fatty acid metabolic genes. Finally, using an LPS-induced ALI/ARDS mouse model, we found that α-KG ameliorated the LPS-induced inflammation and lung pathological damage, as well as α-KG pretreated mice had better clinical scores compared with the LPS group. These findings reveal new mechanisms of α-KG in regulating macrophage polarization which may provide novel strategies for the prevention and treatment of inflammatory diseases, including sepsis and septic ALI/ARDS.
Collapse
|
73
|
Lin Y, Lu Q, Chen C, Wang B, Guo L, Xie J, Chen C, Huang L, Dong L. A synthetic chalcone derivative, compound 39, alleviates lipopolysaccharide-induced acute lung injury in mice. Eur J Pharmacol 2021; 891:173730. [PMID: 33188742 DOI: 10.1016/j.ejphar.2020.173730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 11/06/2020] [Accepted: 11/06/2020] [Indexed: 11/22/2022]
Abstract
Our research group has previously synthesized various chalcone analogues. Of these analogues, compound 39 has been shown to exhibit potent antioxidative activities but its anti-inflammatory and anti-apoptosis effects remain unclear. Thus, the present study investigated the in vivo and in vitro effects and mechanisms of compound 39 in lipopolysaccharide (LPS)-induced acute lung injury (ALI). To induce ALI, the mice received LPS via a tracheal instillation 6 h after intragastric administration of compound 39 or vehicle. Histological changes, the lung wet/dry weight ratio, and the amounts of protein and inflammatory cells in the broncho-alveolar lavage (BAL) fluid were assessed after 24 h. Additionally, to determine its underlying mechanisms, Western blot and immunofluorescence analyses were used. Moreover, the in vitro effects of compound 39 were also investigated. In the in vivo experiment, compound 39 markedly alleviated histopathological alterations, lung edema, and protein leakage, and exhibited potent anti-inflammatory effects. In the in vitro experiments, compound 39 dose-dependently reduced the levels of pro-inflammatory cytokines and reactive oxygen species. The results suggested that the anti-inflammatory effects of compound 39 were due to suppression of the mitogen-activated protein kinase (MAPK)/nuclear factor κB (NF-κB) pathway. Compound 39 also enhanced the protein levels of Bcl-2 and reduced the protein levels of Bax and cleaved caspase-3. The present study confirmed the anti-inflammatory, oxy-radical prohibitive, and anti-apoptosis activities of compound 39 against LPS-induced tissue and cell damage, and revealed the mechanisms underlying those processes.
Collapse
Affiliation(s)
- Yuting Lin
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qingdi Lu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chaolei Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Beibei Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lisha Guo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jingwen Xie
- Department of Pharmacy, Pharmacy School, Wenzhou Medical University, Wenzhou, China
| | - Chengshui Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lili Huang
- Department of Pharmacy, Ningbo Medical Centre Lihuili Hospital, Ningbo, China.
| | - Li Dong
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
74
|
Celastrol alleviates LPS-induced inflammation in BMDMs and acute lung injury in mice via inhibition of p-38 MAPK/MK2 signaling. EUR J INFLAMM 2021. [DOI: 10.1177/20587392211020569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Objective: Celastrol is a compound extracted from a medicinal plant Tripterygium wilfordii which has a broad-spectrum anti-inflammatory effect in traditional medicine. However, the effect of celastrol on acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) is still unknown. Methods: We reported that celastrol alleviated LPS-induced acute lung injury by H&E staining, MPO activity and the expression of cytokines in broncho-alveolar lavage fluid. The effect of celastrol on bone marrow-derived macrophages (BMDMs) after LPS treatment was measured by ELISA and Western blotting. Results: In vivo, celastrol reduced the LPS-induced lung edema and MPO activity of lung tissue. Furthermore, the production of inflammatory cytokines IL-6, TNF-α, and KC in bronchoalveolar lavage was reduced. In vitro, upon treatment of LPS, celastrol dose-dependently inhibited the expression of iNOS in BMDMs. Meanwhile, the expression of IL-6, TNF-α, and KC in BMDMs were also inhibited by celastrol treatment. Furthermore, we found that celastrol attenuated the phosphorylation of p38 MAPK and MK2, and inhibited the interaction between p38 MAPK and MK2. Conclusion: Our data indicate that celastrol has an anti-inflammatory effect on LPS-induced inflammatory response in vivo and in vitro, suggesting celastrol is a promising compound for the treatment of ALI and ARDS.
Collapse
|
75
|
Martucci G, Arcadipane A, Tuzzolino F, Occhipinti G, Panarello G, Carcione C, Bonicolini E, Vitiello C, Lorusso R, Conaldi PG, Miceli V. Identification of a Circulating miRNA Signature to Stratify Acute Respiratory Distress Syndrome Patients. J Pers Med 2020; 11:jpm11010015. [PMID: 33375484 PMCID: PMC7824233 DOI: 10.3390/jpm11010015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/07/2020] [Accepted: 12/25/2020] [Indexed: 02/07/2023] Open
Abstract
There is a need to improve acute respiratory distress syndrome (ARDS) diagnosis and management, particularly with extracorporeal membrane oxygenation (ECMO), and different biomarkers have been tested to implement a precision-focused approach. We included ARDS patients on veno-venous (V-V) ECMO in a prospective observational pilot study. Blood samples were obtained before cannulation, and screened for the expression of 754 circulating microRNA (miRNAs) using high-throughput qPCR and hierarchical cluster analysis. The miRNet database was used to predict target genes of deregulated miRNAs, and the DIANA tool was used to identify significant enrichment pathways. A hierarchical cluster of 229 miRNAs (identified after quality control screening) produced a clear separation of 11 patients into two groups: considering the baseline SAPS II, SOFA, and RESP score cluster A (n = 6) showed higher severity compared to cluster B (n = 5); p values < 0.05. After analysis of differentially expressed miRNAs between the two clusters, 95 deregulated miRNAs were identified, and reduced to 13 by in silico analysis. These miRNAs target genes implicated in tissue remodeling, immune system, and blood coagulation pathways. The blood levels of 13 miRNAs are altered in severe ARDS. Further investigations will have to match miRNA results with inflammatory biomarkers and clinical data.
Collapse
Affiliation(s)
- Gennaro Martucci
- Anesthesia and Intensive Care Department, IRCCS-ISMETT, 90133 Palermo, Italy; (G.M.); (G.O.); (G.P.); (E.B.); (C.V.)
| | - Antonio Arcadipane
- Anesthesia and Intensive Care Department, IRCCS-ISMETT, 90133 Palermo, Italy; (G.M.); (G.O.); (G.P.); (E.B.); (C.V.)
- Correspondence: ; Tel.: +39-091-2192332
| | - Fabio Tuzzolino
- Research Department, IRCCS-ISMETT, 90133 Palermo, Italy; (F.T.); (P.G.C.); (V.M.)
| | - Giovanna Occhipinti
- Anesthesia and Intensive Care Department, IRCCS-ISMETT, 90133 Palermo, Italy; (G.M.); (G.O.); (G.P.); (E.B.); (C.V.)
| | - Giovanna Panarello
- Anesthesia and Intensive Care Department, IRCCS-ISMETT, 90133 Palermo, Italy; (G.M.); (G.O.); (G.P.); (E.B.); (C.V.)
| | | | - Eleonora Bonicolini
- Anesthesia and Intensive Care Department, IRCCS-ISMETT, 90133 Palermo, Italy; (G.M.); (G.O.); (G.P.); (E.B.); (C.V.)
| | - Chiara Vitiello
- Anesthesia and Intensive Care Department, IRCCS-ISMETT, 90133 Palermo, Italy; (G.M.); (G.O.); (G.P.); (E.B.); (C.V.)
| | - Roberto Lorusso
- Cardio-Thoracic Surgery Department Heart and Vascular Centre, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands;
- Cardiovascular Research Institute Maastricht (CARIM), 6229HX Maastricht, The Netherlands
| | - Pier Giulio Conaldi
- Research Department, IRCCS-ISMETT, 90133 Palermo, Italy; (F.T.); (P.G.C.); (V.M.)
| | - Vitale Miceli
- Research Department, IRCCS-ISMETT, 90133 Palermo, Italy; (F.T.); (P.G.C.); (V.M.)
| |
Collapse
|
76
|
Zhao J, Xie F, Chen R, Zhang Z, Dai R, Zhao N, Wang R, Sun Y, Chen Y. Transcription factor NF-κB promotes acute lung injury via microRNA-99b-mediated PRDM1 down-regulation. J Biol Chem 2020; 295:18638-18648. [PMID: 33109608 PMCID: PMC7939479 DOI: 10.1074/jbc.ra120.014861] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/08/2020] [Indexed: 01/12/2023] Open
Abstract
Acute lung injury (ALI), is a rapidly progressing heterogenous pulmonary disorder that possesses a high risk of mortality. Accumulating evidence has implicated the activation of the p65 subunit of NF-κB [NF-κB(p65)] activation in the pathological process of ALI. microRNAs (miRNAs), a group of small RNA molecules, have emerged as major governors due to their post-transcriptional regulation of gene expression in a wide array of pathological processes, including ALI. The dysregulation of miRNAs and NF-κB activation has been implicated in human diseases. In the current study, we set out to decipher the convergence of miR-99b and p65 NF-κB activation in ALI pathology. We measured the release of pro-inflammatory cytokines (IL-1β, IL-6, and TNFα) in bronchoalveolar lavage fluid using ELISA. MH-S cells were cultured and their viability were detected with cell counting kit 8 (CCK8) assays. The results showed that miR-99b was up-regulated, while PRDM1 was down-regulated in a lipopolysaccharide (LPS)-induced murine model of ALI. Mechanistic investigations showed that NF-κB(p65) was enriched at the miR-99b promoter region, and further promoted its transcriptional activity. Furthermore, miR-99b targeted PRDM1 by binding to its 3'UTR, causing its down-regulation. This in-creased lung injury, as evidenced by increased wet/dry ratio of mouse lung, myeloperoxidase activity and pro-inflammatory cytokine secretion, and enhanced infiltration of inflammatory cells in lung tissues. Together, our findings indicate that NF-κB(p65) promotion of miR-99b can aggravate ALI in mice by down-regulating the expression of PRDM1.
Collapse
Affiliation(s)
- Jie Zhao
- The Second Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China.
| | - Fei Xie
- The Six Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China
| | - Ruidong Chen
- The Six Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China
| | - Zhen Zhang
- The Second Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China
| | - Rujun Dai
- The Second Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China
| | - Na Zhao
- The Second Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China
| | - Rongxin Wang
- The Second Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China
| | - Yanhong Sun
- The Second Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China
| | - Yue Chen
- The Second Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China
| |
Collapse
|
77
|
Solorio-Rodríguez A, Escamilla-Rivera V, Uribe-Ramírez M, González-Pozos S, Hernández-Soto J, Rafael-Vázquez L, De Vizcaya-Ruiz A. In vitro cytotoxicity study of superparamagnetic iron oxide and silica nanoparticles on pneumocyte organelles. Toxicol In Vitro 2020; 72:105071. [PMID: 33352256 DOI: 10.1016/j.tiv.2020.105071] [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: 08/14/2020] [Revised: 11/24/2020] [Accepted: 12/12/2020] [Indexed: 11/30/2022]
Abstract
Inhalation is the main route of nanoparticles (NP) exposure during manufacturing. Although many mechanisms of toxicity have been described, the interaction of NP with relevant pneumocytes organelles is not widely understood. Considering that the physicochemical properties of NP influence their toxicological responses, the objective of this study was to evaluate whether exposure to different NP, crystalline Fe3O4 NP and amorphous SiO2 NP could alter pneumocytes organelles in alveolar epithelial cells. To achieve this goal, cell viability, ultrastructural changes, lysosomal damage, mitochondrial membrane potential (MMP), lipid droplets (LD) formation and cytokines production were evaluated by MTT, electron microscopy, lysotracker red staining, JC-1, Oil Red staining and Milliplex® assay respectively. Both NP were observed within lamellar bodies (LB), lysosomes, and cytoplasm causing morphological changes. Exposure to SiO2 NP at 6 h induced lysosomal activation, but not Fe3O4 NP. MMP decreased and LD increased at the highest concentrations after both NP exposure. Pro-inflammatory cytokines were released only after SiO2 NP exposure at 48 h. These results indicate that SiO2 NP have a greater impact than Fe3O4 NP on organelles responsible for energy, secretion, degradation and metabolism in pneumocytes leading to the development of respiratory disorders or the exacerbation of preexisting conditions. Therefore, the established biocompatibility for amorphous NP has to be reconsidered.
Collapse
Affiliation(s)
- A Solorio-Rodríguez
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Ciudad de México, Mexico
| | - V Escamilla-Rivera
- Yale University School of Medicine, Department of Laboratory Medicine, New Haven, Connecticut, USA
| | - M Uribe-Ramírez
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Ciudad de México, Mexico
| | - S González-Pozos
- Unidad de Microscopía Electrónica (LanSE), CINVESTAV-IPN, Ciudad de México, Mexico
| | - J Hernández-Soto
- Departamento de Fisiología, Biofísica y Neurociencias, CINVESTAV-IPN, Ciudad de México, Mexico
| | - L Rafael-Vázquez
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Ciudad de México, Mexico
| | - A De Vizcaya-Ruiz
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Ciudad de México, Mexico.
| |
Collapse
|
78
|
Chimenti L, Morales-Quinteros L, Puig F, Camprubi-Rimblas M, Guillamat-Prats R, Gómez MN, Tijero J, Blanch L, Matute-Bello G, Artigas A. Comparison of direct and indirect models of early induced acute lung injury. Intensive Care Med Exp 2020; 8:62. [PMID: 33336290 PMCID: PMC7746791 DOI: 10.1186/s40635-020-00350-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 10/02/2020] [Indexed: 12/29/2022] Open
Abstract
Background The animal experimental counterpart of human acute respiratory distress syndrome (ARDS) is acute lung injury (ALI). Most models of ALI involve reproducing the clinical risk factors associated with human ARDS, such as sepsis or acid aspiration; however, none of these models fully replicates human ARDS. Aim To compare different experimental animal models of ALI, based on direct or indirect mechanisms of lung injury, to characterize a model which more closely could reproduce the acute phase of human ARDS. Materials and methods Adult male Sprague-Dawley rats were subjected to intratracheal instillations of (1) HCl to mimic aspiration of gastric contents; (2) lipopolysaccharide (LPS) to mimic bacterial infection; (3) HCl followed by LPS to mimic aspiration of gastric contents with bacterial superinfection; or (4) cecal ligation and puncture (CLP) to induce peritonitis and mimic sepsis. Rats were sacrificed 24 h after instillations or 24 h after CLP. Results At 24 h, rats instilled with LPS or HCl-LPS had increased lung permeability, alveolar neutrophilic recruitment and inflammatory markers (GRO/KC, TNF-α, MCP-1, IL-1β, IL-6). Rats receiving only HCl or subjected to CLP had no evidence of lung injury. Conclusions Rat models of ALI induced directly by LPS or HCl-LPS more closely reproduced the acute phase of human ARDS than the CLP model of indirectly induced ALI.
Collapse
Affiliation(s)
- Laura Chimenti
- Critical Care Centre, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Spain
| | - Luis Morales-Quinteros
- Critical Care Centre, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Spain. .,Hospital Universitari Sagrat Cor., Grupo Quirón Salud, Barcelona, Spain.
| | - Ferranda Puig
- Critical Care Centre, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Spain.,CIBER de Enfermedades Respiratorias, Instituto de Investigación Carlos III, Madrid, Spain
| | - Marta Camprubi-Rimblas
- Critical Care Centre, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Spain.,CIBER de Enfermedades Respiratorias, Instituto de Investigación Carlos III, Madrid, Spain
| | - Raquel Guillamat-Prats
- CIBER de Enfermedades Respiratorias, Instituto de Investigación Carlos III, Madrid, Spain
| | - Maria Nieves Gómez
- Critical Care Centre, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Spain
| | - Jessica Tijero
- Critical Care Centre, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Spain
| | - Lluis Blanch
- Critical Care Centre, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Spain.,CIBER de Enfermedades Respiratorias, Instituto de Investigación Carlos III, Madrid, Spain
| | - Gustavo Matute-Bello
- Medical Research Service of the Veterans Affairs/Puget Sound Health Care System, Seattle, WA, USA.,Centre for Lung Biology, Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Antonio Artigas
- Critical Care Centre, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Spain.,Hospital Universitari Sagrat Cor., Grupo Quirón Salud, Barcelona, Spain.,CIBER de Enfermedades Respiratorias, Instituto de Investigación Carlos III, Madrid, Spain
| |
Collapse
|
79
|
Bime C, Camp SM, Casanova N, Oita RC, Ndukum J, Lynn H, Garcia JGN. The acute respiratory distress syndrome biomarker pipeline: crippling gaps between discovery and clinical utility. Transl Res 2020; 226:105-115. [PMID: 32599095 PMCID: PMC7319618 DOI: 10.1016/j.trsl.2020.06.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/17/2020] [Accepted: 06/23/2020] [Indexed: 12/14/2022]
Abstract
Recent innovations in translational research have ushered an exponential increase in the discovery of novel biomarkers, thereby elevating the hope for deeper insights into "personalized" medicine approaches to disease phenotyping and care. However, a critical gap exists between the fast pace of biomarker discovery and the successful translation to clinical use. This gap underscores the fundamental biomarker conundrum across various acute and chronic disorders: how does a biomarker address a specific unmet need? Additionally, the gap highlights the need to shift the paradigm from a focus on biomarker discovery to greater translational impact and the need for a more streamlined drug approval process. The unmet need for biomarkers in acute respiratory distress syndrome (ARDS) is for reliable and validated biomarkers that minimize heterogeneity and allow for stratification of subject selection for enrollment in clinical trials of tailored therapies. This unmet need is particularly highlighted by the ongoing SARS-CoV-2/COVID-19 pandemic. The unprecedented numbers of COVID-19-induced ARDS cases has strained health care systems across the world and exposed the need for biomarkers that would accelerate drug development and the successful phenotyping of COVID-19-infected patients at risk for development of ARDS and ARDS mortality. Accordingly, this review discusses the current state of ARDS biomarkers in the context of the drug development pipeline and highlight gaps between biomarker discovery and clinical implementation while proposing potential paths forward. We discuss potential ARDS biomarkers by category and by context of use, highlighting progress in the development continuum. We conclude by discussing challenges to successful translation of biomarker candidates to clinical impact and proposing possible novel strategies.
Collapse
Affiliation(s)
- Christian Bime
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona.
| | - Sara M Camp
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Nancy Casanova
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Radu C Oita
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Juliet Ndukum
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Heather Lynn
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Joe G N Garcia
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| |
Collapse
|
80
|
Chen T, Zhu G, Meng X, Zhang X. Recent developments of small molecules with anti-inflammatory activities for the treatment of acute lung injury. Eur J Med Chem 2020; 207:112660. [DOI: 10.1016/j.ejmech.2020.112660] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/05/2020] [Accepted: 07/10/2020] [Indexed: 12/22/2022]
|
81
|
Baer B, McCaig L, Yamashita C, Veldhuizen R. Exogenous Surfactant as a Pulmonary Delivery Vehicle for Budesonide In Vivo. Lung 2020; 198:909-916. [PMID: 33106891 PMCID: PMC7587541 DOI: 10.1007/s00408-020-00399-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/14/2020] [Indexed: 12/19/2022]
Abstract
Background Lung inflammation is associated with many respiratory conditions. Consequently, anti-inflammatory medications, like glucocorticoids, have become mainstay intrapulmonary therapeutics. However, their effectiveness for treating inflammation occurring in the alveolar regions of the lung is limited by suboptimal delivery. To improve the pulmonary distribution of glucocorticoids, such as budesonide to distal regions of the lung, exogenous surfactant has been proposed as an ideal delivery vehicle for such therapies. It was therefore hypothesized that fortifying an exogenous surfactant (BLES) with budesonide would enhance efficacy for treating pulmonary inflammation in vivo. Methods An intratracheal instillation of heat-killed bacteria was used to elicit an inflammatory response in the lungs of male and female rats. Thirty minutes after this initial instillation, either budesonide or BLES combined with budesonide was administered intratracheally. To evaluate the efficacy of surfactant delivery, various markers of inflammation were measured in the bronchoalveolar lavage and lung tissue. Results Although budesonide exhibited anti-inflammatory effects when administered alone, delivery with BLES enhanced those effects by lowering the lavage neutrophil counts and myeloperoxidase activity in lung tissue. Combining budesonide with BLES was also shown to reduce several other pro-inflammatory mediators. These results were shown across both sexes, with no observed sex differences. Conclusion Based on these findings, it was concluded that exogenous surfactant can enhance the delivery and efficacy of budesonide in vivo.
Collapse
Affiliation(s)
- Brandon Baer
- Department of Physiology and Pharmacology, Western University, London, ON, Canada.
| | - Lynda McCaig
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Cory Yamashita
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
- Department of Medicine, Western University, London, ON, Canada
| | - Ruud Veldhuizen
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
- Department of Medicine, Western University, London, ON, Canada
| |
Collapse
|
82
|
Tian LX, Tang X, Ma W, Wang J, Zhang W, Liu K, Chen T, Zhu JY, Liang HP. Knockout of cytochrome P450 1A1 enhances lipopolysaccharide-induced acute lung injury in mice by targeting NF-κB activation. FEBS Open Bio 2020; 10:2316-2328. [PMID: 32935470 PMCID: PMC7609787 DOI: 10.1002/2211-5463.12977] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 08/19/2020] [Accepted: 09/09/2020] [Indexed: 12/24/2022] Open
Abstract
Acute lung injury (ALI) is accompanied by overactivation of multiple pro-inflammatory factors. Cytochrome P450 1A1 (CYP1A1) has been shown to aggravate lung injury in response to hyperoxia. However, the relationship between CYP1A1 and lipopolysaccharide (LPS)-induced ALI is unknown. In this study, CYP1A1 was shown to be upregulated in mouse lung in response to LPS. Using CYP1A1-deficient (CYP1A1-/-) mice, we found that CYP1A1 knockout enhanced LPS-induced ALI, as evidenced by increased TNF-α, IL-1β, IL-6, and nitric oxide in lung; these effects were mediated by overactivation of NF-κB and iNOS. Furthermore, we found that aspartate aminotransferase, lactate dehydrogenase, creatine kinase, and creatinine levels were elevated in serum of LPS-induced CYP1A1-/- mice. Altogether, these data provide novel insights into the involvement of CYP1A1 in LPS-induced lung injury.
Collapse
Affiliation(s)
- Li-Xing Tian
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, China
| | - Xin Tang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, China.,Department of Intensive Care Unit, the Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Wei Ma
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, China
| | - Jing Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, China.,Department of Emergency, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Zhang
- Emergency and Trauma College, Hainan Medical University, Haikou, China
| | - Kuan Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, China.,Department of Intensive Care Unit, the Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Tao Chen
- Department of Intensive Care Unit, the Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jun-Yu Zhu
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, China
| | - Hua-Ping Liang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
83
|
Ku TJY, Ribeiro RVP, Ferreira VH, Galasso M, Keshavjee S, Kumar D, Cypel M, Humar A. Ex-vivo delivery of monoclonal antibody (Rituximab) to treat human donor lungs prior to transplantation. EBioMedicine 2020; 60:102994. [PMID: 32950000 PMCID: PMC7501077 DOI: 10.1016/j.ebiom.2020.102994] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/18/2020] [Accepted: 08/25/2020] [Indexed: 01/14/2023] Open
Abstract
Background Ex-vivo lung perfusion (EVLP) is an innovative platform for assessing donor lungs in the pre-transplant window. In this study, we demonstrate an extension of its utility by administering the anti-CD20 monoclonal antibody, Rituximab, during EVLP. We hypothesized that this would lead to targeted depletion of allograft B-cells which may provide significant clinical benefit, including the potential to reduce latent Epstein-Barr virus (EBV) and decrease the incidence of post-transplant lymphoproliferative malignancies. Methods Twenty human donor lungs rejected for transplantation were placed on EVLP with (n = 10) or without (n = 10) 500 mg of Rituximab. Safety parameters such as lung physiology and inflammatory cytokines were evaluated. We measured the delivery efficacy through flow cytometry, immunohistochemistry and ELISA. An in-vitro culture assay, in the presence of complement, was further conducted to monitor whether B-cell depletion would occur in Rituximab-perfused samples. Findings Rituximab was successfully delivered to human lungs during EVLP as evidenced by flow cytometric binding assays where lung tissue and lymph node biopsies demonstrated occupied CD20 epitopes after perfusion with the antibody. Lymph nodes from Rituximab perfusions demonstrated a 10.9 fold-reduction in CD20+ staining compared to controls (p = 0.0003). In lung tissue, Rituximab resulted in an 8.75 fold-reduction in CD20+ staining relative to controls (p = 0.0002). This decrease in CD20+ binding illustrates the successful delivery and occupation of epitopes after perfusion with the Rituximab. No apparent safety concerns were seen as exhibited by markers associated with acute cell injury (e.g., proinflammatory cytokines), cell death (e.g., TUNEL staining), or pulmonary physiology. In a post-perfusion tissue culture model, the addition of complement (human serum) resulted in evidence of B-cell depletion consistent with what would be expected with posttransplant activation of bound Rituximab. Interpretation Our experiments illustrate the potential of EVLP as a platform to deliver monoclonal antibody therapies to treat donor lungs pretransplant with the goal of eliminating a latent virus responsible for considerable morbidity after lung transplantation. Funding Supported by the University Health Network Transplant Center.
Collapse
Affiliation(s)
- Terrance J Y Ku
- Ajmera Transplant Center, University Health Network, PMB 11-175, 585 University Avenue, Toronto, Ontario M5G 2N2, Canada
| | - Rafaela V P Ribeiro
- Latner Thoracic Surgery Research Laboratories, University Health Network, Canada
| | - Victor H Ferreira
- Ajmera Transplant Center, University Health Network, PMB 11-175, 585 University Avenue, Toronto, Ontario M5G 2N2, Canada
| | - Marcos Galasso
- Latner Thoracic Surgery Research Laboratories, University Health Network, Canada
| | - Shaf Keshavjee
- Latner Thoracic Surgery Research Laboratories, University Health Network, Canada
| | - Deepali Kumar
- Ajmera Transplant Center, University Health Network, PMB 11-175, 585 University Avenue, Toronto, Ontario M5G 2N2, Canada
| | - Marcelo Cypel
- Ajmera Transplant Center, University Health Network, PMB 11-175, 585 University Avenue, Toronto, Ontario M5G 2N2, Canada; Latner Thoracic Surgery Research Laboratories, University Health Network, Canada
| | - Atul Humar
- Ajmera Transplant Center, University Health Network, PMB 11-175, 585 University Avenue, Toronto, Ontario M5G 2N2, Canada.
| |
Collapse
|
84
|
Wang Y, Huang W, He M, Peng L, Cai M, Yuan C, Hu Z, Li K. [Inverse ratio ventilation combined with PEEP in infants undergoing thoracoscopic surgery with one lung ventilation for lung cystadenomas: a randomized control trial of 63 cases]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:1008-1012. [PMID: 32895160 DOI: 10.12122/j.issn.1673-4254.2020.07.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the effect of inverse ratio ventilation (IRV) combined with positive end-expiratory pressure (PEEP) in infants undergoing thoracoscopic surgery with single lung ventilation (OLV) for lung cystadenomas. METHODS A total of 66 infants undergoing thoracoscopic surgery with OLV for lung cystadenomas in our hospital from February, 2018 to February, 2019 were randomized into conventional ventilation groups (group N, n=33) and inverse ventilation group (group R, n=33). Hemodynamics and respiratory parameters of the infants were recorded and arterial blood gas analysis was performed at 15 min after two lung ventilation (TLV) (T1), OLV30 min (T2), OLV60 min (T3), and 15 min after recovery of TLV (T4). Bronchoalveolar lavage fluid was collected before and after surgery to detect the expression level of advanced glycation end product receptor (RAGE). RESULTS Sixty-three infants were finally included in this study. At T2 and T3, Cdyn, PaO2 and OI in group R were significantly higher (P < 0.05) and Ppeak, PaCO2 and PA-aO2 were significantly lower than those in group N (P < 0.05). There was no significant difference in HR or MAP between the two groups at T2 and T3 (P > 0.05). The level of RAGE significantly increased after the surgery in both groups (P < 0.05), and was significantly lower in R group than in N group (P < 0.05). CONCLUSIONS In infants undergoing thoracoscopic surgery with OLV for pulmonary cystadenoma, appropriate IRV combined with PEEP does not affect hemodynamic stability and can increases pulmonary compliance, reduce the peak pressure, and improve oxygenation to provide pulmonary protection.
Collapse
Affiliation(s)
- Yun Wang
- Department of Anesthesiology, Guangdong Women and Children's Hospital, Guangzhou 511400, China
| | - Weijian Huang
- Department of Anesthesiology, Guangdong Women and Children's Hospital, Guangzhou 511400, China
| | - Mudan He
- Department of Anesthesiology, Guangdong Women and Children's Hospital, Guangzhou 511400, China
| | - Lingli Peng
- Department of Anesthesiology, Guangdong Women and Children's Hospital, Guangzhou 511400, China
| | - Mingyang Cai
- Department of Anesthesiology, Guangdong Women and Children's Hospital, Guangzhou 511400, China
| | - Chao Yuan
- Department of Anesthesiology, Guangdong Women and Children's Hospital, Guangzhou 511400, China
| | - Zurong Hu
- Department of Anesthesiology, Guangdong Women and Children's Hospital, Guangzhou 511400, China
| | - Kunwei Li
- Department of Anesthesiology, Guangdong Women and Children's Hospital, Guangzhou 511400, China
| |
Collapse
|
85
|
Haddad J, Latoche JD, Nigam S, Bellavia MC, Day KE, Zhu Q, Edwards WB, Anderson CJ, Tavakoli S. Molecular Imaging of Very Late Antigen-4 in Acute Lung Injury. J Nucl Med 2020; 62:280-286. [DOI: 10.2967/jnumed.120.242347] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 06/23/2020] [Indexed: 11/16/2022] Open
|
86
|
Suzuki K, Okada H, Takemura G, Takada C, Tomita H, Yano H, Muraki I, Zaikokuji R, Kuroda A, Fukuda H, Nishio A, Takashima S, Suzuki A, Miyazaki N, Fukuta T, Yamada N, Watanabe T, Doi T, Yoshida T, Kumada K, Ushikoshi H, Yoshida S, Ogura S. Recombinant thrombomodulin protects against LPS-induced acute respiratory distress syndrome via preservation of pulmonary endothelial glycocalyx. Br J Pharmacol 2020; 177:4021-4033. [PMID: 32497259 PMCID: PMC7429482 DOI: 10.1111/bph.15153] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 04/15/2020] [Accepted: 05/28/2020] [Indexed: 01/05/2023] Open
Abstract
Background and Purpose Disruption of the endothelial glycocalyx is causally related to microvascular endothelial dysfunction, a characteristic of sepsis‐induced acute respiratory distress syndrome (ARDS). Recombinant human thrombomodulin (rhTM) attenuates vascular endothelial injuries, but the underlying mechanism remains elusive. Here, we investigated the structural basis and molecular mechanisms of rhTM effects on vascular endothelial injury in a model of sepsis. Experimental Approach LPS (20 mg·kg−1) was intraperitoneally injected into 10‐week‐old male C57BL6 mice, and saline or rhTM was intraperitoneally injected 3 and 24 h after LPS injection. Using serum and/or lung tissue, histological, ultrastructural, and microarray analyses were performed. Key Results Survival rate of rhTM‐treated mice was significantly higher than that of control mice 48 h after LPS injection. Serum concentrations of IL‐6 and high‐mobility group box 1 were lower in the rhTM‐treated group than in the control. Injury to the endothelial glycocalyx in pulmonary capillaries was attenuated by rhTM treatment. Gene set enrichment analysis revealed up‐regulation of gene sets corresponding to cell proliferation/differentiation and anti‐inflammation, such as the TGF‐β pathway, and negative regulation of IL‐6, upon rhTM treatment. Gene expression of heparan sulfate 6‐O‐sulfotransferase 1 and endothelial cell‐specific molecule 1 (components of the endothelial glycocalyx) was significantly preserved by rhTM treatment, and their protein expression levels were maintained in endothelial cells. Conclusion and Implications Our findings show that rhTM treatment affected inflammation, cell proliferation/differentiation, and glycocalyx synthesis in serum and lung tissue, subsequently attenuating ARDS caused by endothelial injury.
Collapse
Affiliation(s)
- Kodai Suzuki
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hideshi Okada
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Genzou Takemura
- Department of Internal Medicine, Asahi University School of Dentistry, Mizuho, Japan
| | - Chihiro Takada
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hiroyuki Tomita
- Department of Tumour Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hirohisa Yano
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Isamu Muraki
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Ryogen Zaikokuji
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Ayumi Kuroda
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hirotsugu Fukuda
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Ayane Nishio
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Shigeo Takashima
- Division of Genomics Research, Life Science Research Center, Gifu University, Gifu, Japan
| | - Akio Suzuki
- Department of Pharmacy, Gifu University Hospital, Gifu, Japan
| | - Nagisa Miyazaki
- Department of Internal Medicine, Asahi University School of Dentistry, Mizuho, Japan
| | - Tetsuya Fukuta
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Noriaki Yamada
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Takatomo Watanabe
- Division of Clinical Laboratory, Gifu University Hospital, Gifu, Japan
| | - Tomoaki Doi
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Takahiro Yoshida
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Keisuke Kumada
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hiroaki Ushikoshi
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Shozo Yoshida
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Shinji Ogura
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| |
Collapse
|
87
|
Litvin DG, Denstaedt SJ, Borkowski LF, Nichols NL, Dick TE, Smith CB, Jacono FJ. Peripheral-to-central immune communication at the area postrema glial-barrier following bleomycin-induced sterile lung injury in adult rats. Brain Behav Immun 2020; 87:610-633. [PMID: 32097765 PMCID: PMC8895345 DOI: 10.1016/j.bbi.2020.02.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 02/02/2020] [Accepted: 02/13/2020] [Indexed: 02/07/2023] Open
Abstract
The pathways for peripheral-to-central immune communication (P → C I-comm) following sterile lung injury (SLI) are unknown. SLI evokes systemic and central inflammation, which alters central respiratory control and viscerosensory transmission in the nucleus tractus solitarii (nTS). These functional changes coincide with increased interleukin-1 beta (IL-1β) in the area postrema, a sensory circumventricular organ that connects P → C I-comm to brainstem circuits that control homeostasis. We hypothesize that IL-1β and its downstream transcriptional target, cyclooxygenase-2 (COX-2), mediate P → C I-comm in the nTS. In a rodent model of SLI induced by intratracheal bleomycin (Bleo), the sigh frequency and duration of post-sigh apnea increased in Bleo- compared to saline- treated rats one week after injury. This SLI-dependent change in respiratory control occurred concurrently with augmented IL-1β and COX-2 immunoreactivity (IR) in the funiculus separans (FS), a barrier between the AP and the brainstem. At this barrier, increases in IL-1β and COX-2 IR were confined to processes that stained for glial fibrillary acidic protein (GFAP) and that projected basolaterally to the nTS. Further, FS radial-glia did not express TNF-α or IL-6 following SLI. To test our hypothesis, we blocked central COX-1/2 activity by intracerebroventricular (ICV) infusion of Indomethacin (Ind). Continuous ICV Ind treatment prevented Bleo-dependent increases in GFAP + and IL-1β + IR, and restored characteristics of sighs that reset the rhythm. These data indicate that changes in sighs following SLI depend partially on activation of a central COX-dependent P → C I-comm via radial-glia of the FS.
Collapse
Affiliation(s)
- David G Litvin
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States; Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States; Department of Fundamental Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland
| | - Scott J Denstaedt
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States; Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - Lauren F Borkowski
- Department of Biomedical Sciences, University of Missouri College of Veterinary Medicine, Columbia, MO 65212, United States
| | - Nicole L Nichols
- Department of Biomedical Sciences, University of Missouri College of Veterinary Medicine, Columbia, MO 65212, United States
| | - Thomas E Dick
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States; Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States
| | - Corey B Smith
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States
| | - Frank J Jacono
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States; Division of Pulmonary, Critical Care and Sleep Medicine, Louis Stokes VA Medical Center, Cleveland, OH 44106, United States.
| |
Collapse
|
88
|
Muthumalage T, Lucas JH, Wang Q, Lamb T, McGraw MD, Rahman I. Pulmonary Toxicity and Inflammatory Response of E-Cigarette Vape Cartridges Containing Medium-Chain Triglycerides Oil and Vitamin E Acetate: Implications in the Pathogenesis of EVALI. TOXICS 2020; 8:toxics8030046. [PMID: 32605182 PMCID: PMC7560420 DOI: 10.3390/toxics8030046] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/20/2020] [Accepted: 06/24/2020] [Indexed: 12/11/2022]
Abstract
Recently, there has been an outbreak of a condition named e-cigarette or vaping products-associated lung injury (EVALI). The primary components of vaping products include tetrahydrocannabinol (THC), vitamin E acetate (VEA) and medium-chain triglycerides (MCT), may be responsible for acute lung toxicity. Currently, little information is available on the physiological and biological effects of exposure to these products. We hypothesized that these CBD/counterfeit vape cartridges and their constituents (VEA and MCT) induce pulmonary toxicity, mediated by oxidative damage and inflammatory responses, leading to acute lung injury. We studied the potential mechanisms of CBD/counterfeit vape cartridge aerosol induced inflammatory response by evaluating the generation of reactive oxygen species by MCT, VEA, and cartridges and their effects on the inflammatory state of pulmonary epithelium and immune cells both in vitro and in vivo. Cells exposed to these aerosols generated reactive oxygen species, caused cytotoxicity, induced epithelial barrier dysfunction, and elicited an inflammatory response. Using a murine model, the parameters of acute toxicity to aerosol inhalation were assessed. Infiltration of neutrophils and lymphocytes was accompanied by significant increases in IL-6, eotaxin, and G-CSF in the bronchoalveolar lavage fluid (BALF). In mouse BALF, eicosanoid inflammatory mediators, leukotrienes, were significantly increased. Plasma from e-cig users also showed increased levels of hydroxyeicosatetraenoic acid (HETEs) and various eicosanoids. Exposure to CBD/counterfeit vape cartridge aerosols showed the most significant effects and toxicity compared to MCT and VEA. In addition, we determined SARS-CoV-2 related proteins and found no impact associated with aerosol exposures from these tested cartridges. Overall, this study demonstrates acute exposure to specific CBD/counterfeit vape cartridges induces in vitro cytotoxicity, barrier dysfunction, and inflammation and in vivo mouse exposure induces acute inflammation with elevated proinflammatory markers in the pathogenesis of EVALI.
Collapse
Affiliation(s)
- Thivanka Muthumalage
- Department of Environmental Medicine, School of Medicine & Dentistry, University of Rochester Medical Center, Rochester, NY 14642, USA; (T.M.); (J.H.L.); (Q.W.); (T.L.)
| | - Joseph H. Lucas
- Department of Environmental Medicine, School of Medicine & Dentistry, University of Rochester Medical Center, Rochester, NY 14642, USA; (T.M.); (J.H.L.); (Q.W.); (T.L.)
| | - Qixin Wang
- Department of Environmental Medicine, School of Medicine & Dentistry, University of Rochester Medical Center, Rochester, NY 14642, USA; (T.M.); (J.H.L.); (Q.W.); (T.L.)
| | - Thomas Lamb
- Department of Environmental Medicine, School of Medicine & Dentistry, University of Rochester Medical Center, Rochester, NY 14642, USA; (T.M.); (J.H.L.); (Q.W.); (T.L.)
| | - Matthew D. McGraw
- Division of Pediatric Pulmonology, School of Medicine & Dentistry, University of Rochester Medical Center, Rochester, NY 14642, USA;
| | - Irfan Rahman
- Department of Environmental Medicine, School of Medicine & Dentistry, University of Rochester Medical Center, Rochester, NY 14642, USA; (T.M.); (J.H.L.); (Q.W.); (T.L.)
- Correspondence: ; Tel.: +1-(585)-275-6911
| |
Collapse
|
89
|
Muthumalage T, Lucas JH, Wang Q, Lamb T, McGraw MD, Rahman I. Pulmonary toxicity and inflammatory response of e-cigarettes containing medium-chain triglyceride oil and vitamin E acetate: Implications in the pathogenesis of EVALI but independent of SARS-COV-2 COVID-19 related proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 32587960 DOI: 10.1101/2020.06.14.151381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Recently, there has been an outbreak associated with the use of e-cigarette or vaping products, associated lung injury (EVALI). The primary components of vaping products, vitamin E acetate (VEA) and medium-chain triglycerides (MCT) may be responsible for acute lung toxicity. Currently, little information is available on the physiological and biological effects of exposure to these products. We hypothesized that these e-cig cartridges and their constituents (VEA and MCT) induce pulmonary toxicity, mediated by oxidative damage and inflammatory responses, leading to acute lung injury. We studied the potential mechanisms of cartridge aerosol induced inflammatory response by evaluating the generation of reactive oxygen species by MCT, VEA, and cartridges, and their effects on the inflammatory state of pulmonary epithelium and immune cells both in vitro and in vivo. Cells exposed to these aerosols generated reactive oxygen species, caused cytotoxicity, induced epithelial barrier dysfunction, and elicited an inflammatory response. Using a murine model, the parameters of acute toxicity to aerosol inhalation were assessed. Infiltration of neutrophils and lymphocytes was accompanied by significant increases in IL-6, eotaxin, and G-CSF in the bronchoalveolar lavage fluid (BALF). In mouse plasma, eicosanoid inflammatory mediators, leukotrienes, were significantly increased. Plasma from e-cig users also showed increased levels of hydroxyeicosatetraenoic acid (HETEs) and various eicosanoids. Exposure to e-cig cartridge aerosols showed the most significant effects and toxicity compared to MCT and VEA. In addition, we determined at SARS-COV-2 related proteins and found no impact associated with aerosol exposures from these tested cartridges. Overall, this study demonstrates acute exposure to specific e-cig cartridges induces in vitro cytotoxicity, barrier dysfunction, and inflammation and in vivo mouse exposure induces acute inflammation with elevated pro-inflammatory markers in the pathogenesis of EVALI.
Collapse
|
90
|
Abstract
INTRODUCTION The use of mechanical ventilation is an invaluable tool in caring for critically ill patients. Enhancing our capabilities in mechanical ventilation has been instrumental in the ability to support clinical conditions and diseases which were once associated with high mortality. Areas covered: Within this manuscript, we will look to discuss emerging approaches to improving the care of pediatric patients who require mechanical ventilation. After an extensive literature search, we will provide a brief review of the history and pathophysiology of acute respiratory distress syndrome, an assessment of several ventilator settings, a discussion on assisted ventilation, review of therapy used to rescue in severe respiratory failure, methods of monitoring the effects of mechanical ventilation, and nutrition. Expert opinion: As we have advanced in our care, we are seeing children survive illnesses that would have once claimed their lives. Given this knowledge, we must continue to advance the research in pediatric critical care to understand the means in which we can tailor the therapy to the patient in efforts to efficiently liberate them from mechanical ventilation once their illness has resolved.
Collapse
Affiliation(s)
- Duane C Williams
- a Division of Pediatric Critical Care Medicine, Department of Pediatrics , Penn State Hershey Children's Hospital , Hershey , PA , USA
| | - Ira M Cheifetz
- b Division of Pediatric Critical Care Medicine, Department of Pediatrics , Duke Children's Hospital , Durham , NC , USA
| |
Collapse
|
91
|
Bagaria V, Mathur P, Madan K, Kumari M, Sagar S, Gupta A, Soni KD, Bhattacharjee H, Kumar S. Predicting Outcomes After Blunt Chest Trauma-Utility of Thoracic Trauma Severity Score, Cytokines (IL-1β, IL-6, IL-8, IL-10, and TNF-α), and Biomarkers (vWF and CC-16). Indian J Surg 2020; 83:113-119. [PMID: 32837068 PMCID: PMC7275928 DOI: 10.1007/s12262-020-02407-4] [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: 11/30/2019] [Accepted: 05/22/2020] [Indexed: 11/18/2022] Open
Abstract
Thoracic trauma severity score (TTSS) has been used to assess severity and risk of pulmonary complications in patients with chest trauma. The role of cytokines and biomarkers in patients with chest trauma and its association with TTSS is not well elucidated. The aim of the study was to assess the cytokines (IL-1β, IL-6, IL-8, IL-10, and TNF-α) and biomarkers (vWF, CC-16) in patients of thoracic trauma and correlate it with TTSS and patient’s outcome. This was a prospective observational study. Serum and bronchoalveolar lavage fluid samples were collected from chest trauma patients. TTSS was calculated in all patients. Suitable controls for serum and bronchoalveolar lavage (BAL) sample were selected. The outcome parameters included patient discharge or death, duration of hospital, and intensive care unit (ICU) stay. Forty-three patients were included. There was no significant correlation between the measured cytokines and biomarkers and TTSS. The mean TTSS of patients who had a fatal outcome was significantly higher than the patients who recovered. Patients with a high TTSS score had a significant prolonged ICU stay. Patients with a prolonged hospital stay had lower values of CC-16. TTSS is a useful tool to predict severity of chest trauma and prolonged ICU stay. Lower levels of CC-16 in BAL fluid of chest trauma patients were associated with prolonged hospital stay suggestive of its protective role in the airway. Longer prospective studies are required to determine the role of cytokines and biomarkers in patients with thoracic trauma in predicting the patient’s outcome.
Collapse
Affiliation(s)
- Vivek Bagaria
- Department of Surgical Disciplines, All India Institute of Medical Sciences, New Delhi, India
| | - Purva Mathur
- Department of Laboratory Medicine, JPN Apex Trauma Center, All India Institute of Medical Sciences, New Delhi, India
| | - Karan Madan
- Department of Pulmonary Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Minu Kumari
- Division of Trauma Surgery & Critical Care, JPN Apex Trauma Center, All India Institute of Medical Sciences, New Delhi, India
| | - Sushma Sagar
- Division of Trauma Surgery & Critical Care, JPN Apex Trauma Center, All India Institute of Medical Sciences, New Delhi, India
| | - Amit Gupta
- Division of Trauma Surgery & Critical Care, JPN Apex Trauma Center, All India Institute of Medical Sciences, New Delhi, India
| | - Kapil Dev Soni
- Division of Trauma Surgery & Critical Care, JPN Apex Trauma Center, All India Institute of Medical Sciences, New Delhi, India
| | - Hemanga Bhattacharjee
- Department of Surgical Disciplines, All India Institute of Medical Sciences, New Delhi, India
| | - Subodh Kumar
- Division of Trauma Surgery & Critical Care, JPN Apex Trauma Center, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
92
|
Jiang ZF, Zhang L, Shen J. MicroRNA: Potential biomarker and target of therapy in acute lung injury. Hum Exp Toxicol 2020; 39:1429-1442. [PMID: 32495695 DOI: 10.1177/0960327120926254] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs stretching over 18-22 nucleotides and considered to be modifiers of many respiratory diseases. They are highly evolutionary conserved and have been implicated in several biological processes, including cell proliferation, apoptosis, differentiation, among others. Acute lung injury (ALI) is a fatal disease commonly caused by direct or indirect injury factors and has a high mortality rate in intensive care unit. Changes in expression of several types of miRNAs have been reported in patients with ALI. Some miRNAs suppress cellular injury and accelerate the recovery of ALI by targeting specific molecules and decreasing excessive immune response. For this reason, miRNAs are proposed as potential biomarkers for ALI and as therapeutic targets for this disease. This review summarizes current evidence supporting the role of miRNAs in ALI.
Collapse
Affiliation(s)
- Z-F Jiang
- Center of Emergency & Intensive Care Unit, Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - L Zhang
- Center of Emergency & Intensive Care Unit, Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - J Shen
- Center of Emergency & Intensive Care Unit, Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
93
|
Zhang V, Ganz T, Nemeth E, Kim A. Iron overload causes a mild and transient increase in acute lung injury. Physiol Rep 2020; 8:e14470. [PMID: 32596989 PMCID: PMC7322498 DOI: 10.14814/phy2.14470] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 05/05/2020] [Accepted: 05/07/2020] [Indexed: 12/27/2022] Open
Abstract
Recent studies have demonstrated a strong link between acute respiratory distress syndrome (ARDS) and the levels of iron and iron-related proteins in the lungs. However, the role of iron overload in ARDS development has yet to be characterized. In this study, we compared the highly iron-overloaded hepcidin knockout mice (HKO) to their iron-sufficient wild-type (WT) littermates in a model of sterile acute lung injury (ALI) induced by treatment with oropharyngeal (OP) LPS. There were no major differences in systemic inflammatory response or airway neutrophil infiltration between the two groups at the time of maximal injury (days 2 and 3) or during the recovery phase (day 7). Hepcidin knockout mice had transiently increased bronchoalveolar lavage fluid (BALF) protein and MPO activity in the lung and BALF on day 3, indicating worse vascular leakage and increased neutrophil activity, respectively. The increased ALI severity in iron-overloaded mice may be a result of increased apoptosis of lung tissue, as evidenced by an increase in cleaved capsase-3 protein in lung homogenates from HKO mice versus WT mice on day 3. Altogether, our data suggest that even severe iron overload has a relatively minor and transient effect in LPS-induced ALI.
Collapse
Affiliation(s)
- Vida Zhang
- Department of MedicineDavid Geffen School of MedicineUCLALos AngelesCAUSA
- Department of Molecular and Medical PharmacologyUCLALos AngelesCAUSA
| | - Tomas Ganz
- Department of MedicineDavid Geffen School of MedicineUCLALos AngelesCAUSA
| | - Elizabeta Nemeth
- Department of MedicineDavid Geffen School of MedicineUCLALos AngelesCAUSA
| | - Airie Kim
- Department of MedicineDavid Geffen School of MedicineUCLALos AngelesCAUSA
| |
Collapse
|
94
|
Kosutova P, Mikolka P, Balentova S, Adamkov M, Calkovska A, Mokra D. Effects of PDE3 Inhibitor Olprinone on the Respiratory Parameters, Inflammation, and Apoptosis in an Experimental Model of Acute Respiratory Distress Syndrome. Int J Mol Sci 2020; 21:E3382. [PMID: 32403267 PMCID: PMC7247002 DOI: 10.3390/ijms21093382] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/03/2020] [Accepted: 05/06/2020] [Indexed: 12/12/2022] Open
Abstract
This study aimed to investigate whether a selective phosphodiesterase-3 (PDE3) inhibitor olprinone can positively influence the inflammation, apoptosis, and respiratory parameters in animals with acute respiratory distress syndrome (ARDS) model induced by repetitive saline lung lavage. Adult rabbits were divided into 3 groups: ARDS without therapy (ARDS), ARDS treated with olprinone i.v. (1 mg/kg; ARDS/PDE3), and healthy ventilated controls (Control), and were oxygen-ventilated for the following 4 h. Dynamic lung-thorax compliance (Cdyn), mean airway pressure (MAP), arterial oxygen saturation (SaO2), alveolar-arterial gradient (AAG), ratio between partial pressure of oxygen in arterial blood to a fraction of inspired oxygen (PaO2/FiO2), oxygenation index (OI), and ventilation efficiency index (VEI) were evaluated every hour. Post mortem, inflammatory and oxidative markers (interleukin (IL)-6, IL-1β, a receptor for advanced glycation end products (RAGE), IL-10, total antioxidant capacity (TAC), 3-nitrotyrosine (3NT), and malondialdehyde (MDA) and apoptosis (apoptotic index and caspase-3) were assessed in the lung tissue. Treatment with olprinone reduced the release of inflammatory mediators and markers of oxidative damage decreased apoptosis of epithelial cells and improved respiratory parameters. The results indicate a future potential of PDE3 inhibitors also in the therapy of ARDS.
Collapse
Affiliation(s)
- Petra Kosutova
- Biomedical Center Martin and Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (P.K.); (P.M.); (A.C.)
| | - Pavol Mikolka
- Biomedical Center Martin and Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (P.K.); (P.M.); (A.C.)
| | - Sona Balentova
- Department of Histology and Embryology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (S.B.); (M.A.)
| | - Marian Adamkov
- Department of Histology and Embryology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (S.B.); (M.A.)
| | - Andrea Calkovska
- Biomedical Center Martin and Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (P.K.); (P.M.); (A.C.)
| | - Daniela Mokra
- Biomedical Center Martin and Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (P.K.); (P.M.); (A.C.)
| |
Collapse
|
95
|
Viswan A, Singh C, Kayastha AM, Azim A, Sinha N. An NMR based panorama of the heterogeneous biology of acute respiratory distress syndrome (ARDS) from the standpoint of metabolic biomarkers. NMR IN BIOMEDICINE 2020; 33:e4192. [PMID: 31733128 DOI: 10.1002/nbm.4192] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 08/16/2019] [Accepted: 09/05/2019] [Indexed: 06/10/2023]
Abstract
Acute respiratory distress syndrome (ARDS), manifested by intricate etiology and pathophysiology, demands careful clinical surveillance due to its high mortality and imminent life support measures. NMR based metabolomics provides an approach for ARDS which culminates from a wide spectrum of illness thereby confounding early manifestation and prognosis predictors. 1 H NMR with its manifold applications in critical disease settings can unravel the biomarker of ARDS thus holding potent implications by providing surrogate endpoints of clinical utility. NMR metabolomics which is the current apogee platform of omics trilogy is contributing towards the possible panacea of ARDS by subsequent validation of biomarker credential on larger datasets. In the present review, the physiological derangements that jeopardize the whole metabolic functioning in ARDS are exploited and the biomarkers involved in progression are addressed and substantiated. The following sections of the review also outline the clinical spectrum of ARDS from the standpoint of NMR based metabolomics which is an emerging element of systems biology. ARDS is the main premise of intensivists textbook, which has been thoroughly reviewed along with its incidence, progressive stages of severity, new proposed diagnostic definition, and the preventive measures and the current pitfalls of clinical management. The advent of new therapies, the need for biomarkers, the methodology and the contemporary promising approaches needed to improve survival and address heterogeneity have also been evaluated. The review has been stepwise illustrated with potent biometrics employed to selectively pool out differential metabolites as diagnostic markers and outcome predictors. The following sections have been drafted with an objective to better understand ARDS mechanisms with predictive and precise biomarkers detected so far on the basis of underlying physiological parameters having close proximity to diseased phenotype. The aim of this review is to stimulate interest in conducting more studies to help resolve the complex heterogeneity of ARDS with biomarkers of clinical utility and relevance.
Collapse
Affiliation(s)
- Akhila Viswan
- Centre of Biomedical Research, Sanjay Gandhi Post Graduate Institute of Medical Sciences (SGPGIMS) - Campus, Lucknow, Uttar Pradesh, India
- Faculty of Engineering and Technology, Dr. A. P. J Abdul Kalam Technical University, Lucknow, India
| | - Chandan Singh
- Centre of Biomedical Research, Sanjay Gandhi Post Graduate Institute of Medical Sciences (SGPGIMS) - Campus, Lucknow, Uttar Pradesh, India
- School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Arvind M Kayastha
- School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Afzal Azim
- Critical Care Medicine, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Neeraj Sinha
- Centre of Biomedical Research, Sanjay Gandhi Post Graduate Institute of Medical Sciences (SGPGIMS) - Campus, Lucknow, Uttar Pradesh, India
| |
Collapse
|
96
|
Overexpression of transcription factor EB regulates mitochondrial autophagy to protect lipopolysaccharide-induced acute lung injury. Chin Med J (Engl) 2019; 132:1298-1304. [PMID: 30946071 PMCID: PMC6629347 DOI: 10.1097/cm9.0000000000000243] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Acute lung injury (ALI) is characterized by an acute inflammatory process, and oxidative stress in the lung tissue leads to a lack of effective therapeutics. This study aimed to identify whether the overexpression of transcription factor EB (TFEB) regulates mitophagy to protect against lipopolysaccharide (LPS)-induced ALI. METHODS We detected the expression of inflammatory factors, cytochrome c (Cyt.c) and nicotinamide adenine dinucleotide phosphate (NADPH), and autophagy-related proteins and observed the changes in lung histopathology induced by ALI in rats and the changes in the cell ultrastructure of primary alveolar type II epithelial cells induced by changing the expression of TFEB in the context of ALI. RESULTS The overexpression of TFEB could reduce the expression of proinflammatory factors, such as IL-1 and IL-6, and increase the expression of anti-inflammatory factors, such as IL-10, both in vitro and in vivo. In addition, the overexpression of TFEB could reduce the Cyt.c and NADPH levels both in vivo and in vitro. The overexpression of TFEB could upregulate the expression of autophagy-related proteins, such as lysosomal-associated membrane protein 1 (LAMP1), microtubule-associated protein light chain 3B (LC3B), and Beclin both in vivo and in vitro, and promote mitochondrial autophagy. The overexpression of TFEB significantly improved the histopathologic changes induced by LPS-induced ALI in rats. However, low TFEB expression produced the opposite results. CONCLUSION TFEB overexpression can decrease inflammation and mitochondrial damage in the lung tissue and alveolar epithelial cells through regulating mitochondrial autophagy to protect against LPS-induced ALI. Therefore, TFEB is likely a potential therapeutic target in LPS-induced ALI.
Collapse
|
97
|
MHTP, a synthetic tetratetrahydroisoquinoline alkaloid, attenuates lipopolysaccharide-induced acute lung injury via p38MAPK/p65NF-κB signaling pathway-TLR4 dependent. Inflamm Res 2019; 68:1061-1070. [DOI: 10.1007/s00011-019-01291-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 07/24/2019] [Accepted: 10/02/2019] [Indexed: 01/07/2023] Open
|
98
|
Ying Y, Mao Y, Yao M. NLRP3 Inflammasome Activation by MicroRNA-495 Promoter Methylation May Contribute to the Progression of Acute Lung Injury. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:801-814. [PMID: 31734560 PMCID: PMC6861628 DOI: 10.1016/j.omtn.2019.08.028] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/01/2019] [Accepted: 08/17/2019] [Indexed: 12/18/2022]
Abstract
Acute lung injury (ALI) is a pulmonary disorder that causes acute respiratory failure, thus leading to relative high mortality worldwide. However, the molecular mechanisms of ALI remain largely unknown. MicroRNA (miRNA)-dependent control of gene expression at a post-transcriptional level has been recently reported. Herein, we identify a candidate miRNA, miR-495, that affects the progression of ALI. Alveolar macrophages (NR8383) were treated with 1 μg/mL lipopolysaccharide (LPS) to establish a cell-injury model. Combined with the data from western blot, methylation-specific PCR, methylated DNA immunoprecipitation, and chromatin immunoprecipitation assays, NLRP3 inflammasome activation and methylation-dependent repression of miR-495 were found in LPS-exposed NR8383 cells. Dual-luciferase reporter gene assay and miR-495 gain-of-function experiments confirmed that NLRP3 was a target of miR-495. Next, the expression of miR-495 and NLRP3 was overexpressed or silenced to assess their effects on NLRP3 inflammasome activation, alveolar macrophage inflammation, and pyroptosis in vitro. As demonstrated, overexpressed miR-495 alleviated alveolar macrophage inflammation and pyroptosis and inhibited NLRP3 inflammasome activation by negatively regulating the NLRP3 gene. Consistently, elevated miR-495 alleviated lung injury and reduced the neutrophil infiltration and inflammation in rat models of LPS-induced ALI. Taken together, the data in our study demonstrated that methylation of the miR-495 promoter could downregulate miR-495, whose elevation could attenuate the activation of the NLRP3 inflammasome to protect against ALI, which provides novel therapeutic targets for ALI treatment.
Collapse
Affiliation(s)
- Youguo Ying
- Department of Intensive Care Unit, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, P.R. China
| | - Yong Mao
- Department of Intensive Care Unit, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, P.R. China
| | - Min Yao
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, P.R. China.
| |
Collapse
|
99
|
Ding XF, Li JB, Liang HY, Wang ZY, Jiao TT, Liu Z, Yi L, Bian WS, Wang SP, Zhu X, Sun TW. Predictive model for acute respiratory distress syndrome events in ICU patients in China using machine learning algorithms: a secondary analysis of a cohort study. J Transl Med 2019; 17:326. [PMID: 31570096 PMCID: PMC6771100 DOI: 10.1186/s12967-019-2075-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 09/18/2019] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND To develop a machine learning model for predicting acute respiratory distress syndrome (ARDS) events through commonly available parameters, including baseline characteristics and clinical and laboratory parameters. METHODS A secondary analysis of a multi-centre prospective observational cohort study from five hospitals in Beijing, China, was conducted from January 1, 2011, to August 31, 2014. A total of 296 patients at risk for developing ARDS admitted to medical intensive care units (ICUs) were included. We applied a random forest approach to identify the best set of predictors out of 42 variables measured on day 1 of admission. RESULTS All patients were randomly divided into training (80%) and testing (20%) sets. Additionally, these patients were followed daily and assessed according to the Berlin definition. The model obtained an average area under the receiver operating characteristic (ROC) curve (AUC) of 0.82 and yielded a predictive accuracy of 83%. For the first time, four new biomarkers were included in the model: decreased minimum haematocrit, glucose, and sodium and increased minimum white blood cell (WBC) count. CONCLUSIONS This newly established machine learning-based model shows good predictive ability in Chinese patients with ARDS. External validation studies are necessary to confirm the generalisability of our approach across populations and treatment practices.
Collapse
Affiliation(s)
- Xian-Fei Ding
- Department of General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, 1 Jianshe East Road, Zhengzhou, 450052, China
| | - Jin-Bo Li
- Department of General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, 1 Jianshe East Road, Zhengzhou, 450052, China.,Department of Electrical & Computer Engineering, University of Alberta, Edmonton, Canada
| | - Huo-Yan Liang
- Department of General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, 1 Jianshe East Road, Zhengzhou, 450052, China
| | - Zong-Yu Wang
- Department of Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Ting-Ting Jiao
- Department of General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, 1 Jianshe East Road, Zhengzhou, 450052, China
| | - Zhuang Liu
- Intensive Care Unit, Beijing Friendship Hospital Affiliated with Capital Medical University, Beijing, China
| | - Liang Yi
- Intensive Care Unit, Xiyuan Hospital Affiliated with the China Academy of Chinese Medical Sciences, Beijing, China
| | - Wei-Shuai Bian
- Intensive Care Unit, Beijing Shijitan Hospital Affiliated with Capital Medical University, Beijing, China
| | - Shu-Peng Wang
- Intensive Care Unit, China-Japan Friendship Hospital, Beijing, China
| | - Xi Zhu
- Department of Critical Care Medicine, Peking University Third Hospital, Beijing, China.
| | - Tong-Wen Sun
- Department of General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, 1 Jianshe East Road, Zhengzhou, 450052, China.
| |
Collapse
|
100
|
Mokra D, Mikolka P, Kosutova P, Mokry J. Corticosteroids in Acute Lung Injury: The Dilemma Continues. Int J Mol Sci 2019; 20:ijms20194765. [PMID: 31557974 PMCID: PMC6801694 DOI: 10.3390/ijms20194765] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 09/21/2019] [Accepted: 09/25/2019] [Indexed: 12/19/2022] Open
Abstract
Acute lung injury (ALI) represents a serious heterogenous pulmonary disorder with high mortality. Despite improved understanding of the pathophysiology, the efficacy of standard therapies such as lung-protective mechanical ventilation, prone positioning and administration of neuromuscular blocking agents is limited. Recent studies have shown some benefits of corticosteroids (CS). Prolonged use of CS can shorten duration of mechanical ventilation, duration of hospitalization or improve oxygenation, probably because of a wide spectrum of potentially desired actions including anti-inflammatory, antioxidant, pulmonary vasodilator and anti-oedematous effects. However, the results from experimental vs. clinical studies as well as among the clinical trials are often controversial, probably due to differences in the designs of the trials. Thus, before the use of CS in ARDS can be definitively confirmed or refused, the additional studies should be carried on to determine the most appropriate dosing, timing and choice of CS and to analyse the potential risks of CS administration in various groups of patients with ARDS.
Collapse
Affiliation(s)
- Daniela Mokra
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK-03601 Martin, Slovakia; (P.M.); (P.K.)
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK-03601 Martin, Slovakia;
- Correspondence: ; Tel.: +421-43-263-3454
| | - Pavol Mikolka
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK-03601 Martin, Slovakia; (P.M.); (P.K.)
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK-03601 Martin, Slovakia;
| | - Petra Kosutova
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK-03601 Martin, Slovakia; (P.M.); (P.K.)
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK-03601 Martin, Slovakia;
| | - Juraj Mokry
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK-03601 Martin, Slovakia;
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK-03601 Martin, Slovakia
| |
Collapse
|