1
|
Wang LN, Shao YR, Wang PF, Lv J, He DK. Characteristics of phosgene aspiration lung injury analyzed based on transcriptomics and proteomics. Front Genet 2024; 15:1393665. [PMID: 38826806 PMCID: PMC11140124 DOI: 10.3389/fgene.2024.1393665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/26/2024] [Indexed: 06/04/2024] Open
Abstract
Background Phosgene is a chemical material widely used worldwide. No effective method has been developed to reverse its pathological injuries. Some studies have shown that neuronal inflammation in lung tissue is involved, but the specific mechanism has not been reported. Objective To analyze the expression alterations of whole transcriptome gene sequencing bioinformatics and protein expression profile in lung tissue after phosgene aspiration lung injury (P-ALI) and find the main factors and pathways affecting the prognosis of P-ALI. Methods Rat models of P-ALI were made by phosgene. Rats were divided into a P-ALI group and a blank group. Hematoxylin-eosin (HE) staining and lung wet/dry ratio measurement were used to evaluate the lung injury. The levels of inflammatory factors were measured by ELISA. High-throughput sequencing was used to measure the expression profile of each gene. Protein expression profiles were determined by label-free relative quantification of the differential proteome. Results Lung injury such as the disordered structure of alveolar wall and inflammatory factors (IL-1β, IL-18, and IL-33) were significantly increased in the P-ALI group (p < 0.05). There were 225 differentially expressed lncRNAs, including 85 upregulated and 140 downregulated genes. They were also the genomes with the most significant changes in transcriptome gene expression, mainly constituting cytoplasmic, synaptic structures and transporters, and involved in amino acid and carbon metabolism. There were 42 differentially expressed circRNAs, including 25 upregulated genes and 17 downregulated genes, mainly involved in cell composition, growth, differentiation, and division. There were only 10 differentially expressed miRNAs genes, all upregulated and mainly involved in the inflammatory response pathway. Proteome identification showed 79 differentially expressed proteins. KEGG enrichment analysis showed that it was mainly involved in the N-glycan biosynthesis pathway. Conclusion We discovered that differentially regulated genes (lncRNAs, circRNAs, and miRNAs) were primarily associated with neuronal reflexes and synaptic signaling, including neurotransmitter transmission, ion signaling pathway conduction, neuronal projection, and synaptic vesicle circulation. They affected inflammatory factors and other metabolic pathways. This finding could be explored in future studies.
Collapse
Affiliation(s)
- Li-Na Wang
- Department of General Practice, Jinshan Hospital, Fudan University, Shanghai, China
| | - Yi-Ru Shao
- Center of Emergency and Critical Medicine, Jinshan Hospital, Fudan University, Shanghai, China
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, China
| | - Peng-Fei Wang
- Center of Emergency and Critical Medicine, Jinshan Hospital, Fudan University, Shanghai, China
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, China
| | - Jiang Lv
- Department of General Practice, Jinshan Hospital, Fudan University, Shanghai, China
| | - Dai-Kun He
- Department of General Practice, Jinshan Hospital, Fudan University, Shanghai, China
- Center of Emergency and Critical Medicine, Jinshan Hospital, Fudan University, Shanghai, China
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, China
| |
Collapse
|
2
|
Memet O, Cao C, Hu H, Dun Y, Bao X, Liu F, Zhang L, Zhou J, Shen J. Galectin-3 inhibition ameliorates alveolar epithelial cell pyroptosis in phosgene-induced acute lung injury. Int Immunopharmacol 2024; 132:111965. [PMID: 38583242 DOI: 10.1016/j.intimp.2024.111965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/10/2024] [Accepted: 03/26/2024] [Indexed: 04/09/2024]
Abstract
Phosgene is a type of poisonous gas that can cause acute lung injury (ALI) upon accidental exposure. Casualties still occur due to phosgene-induced acute lung injury (P-ALI) from accidents resulting from improper operations. The pathological mechanisms of P-ALI are still understudied. Thus, we performed scRNA-seq on cells isolated from all subpopulations of the BALF in P-ALI and found that Gal3 expression was significantly higher in the gas group than in the control group. Further analysis revealed a ligand-receptor correspondence between alveolar macrophages (AMs) and alveolar epithelial cells (AEC), with Gal3 playing a key role in this interaction. To confirm and elaborate on this discovery, we selected four time points during the previous week: sham (day 0), day 1, day 3, and day 7 in the P-ALI mouse model and found that Gal3 expression was significantly elevated in P-ALI, most abundantly expressed in AM cells. This was further confirmed with the use of a Gal3 inhibitor. The inhibition of Gal3 and elimination of AMs in mice both attenuated epithelial cell pyroptosis, as confirmed in in vitro experiments, and revealed the Gal3/caspase-8/GSDMD signaling pathway. These findings suggest that Galectin-3 inhibition can ameliorate AEC pyroptosis by inhibiting the Gal3/caspase-8/GSDMD signaling pathway, thus reducing alveolar damage in mice with P-ALI. This finding provides novel insights for improving treatment efficacy for P-ALI.
Collapse
Affiliation(s)
- Obulkasim Memet
- Center of Emergency and Critical Medicine in Jinshan Hospital of Fudan University, Shanghai 201508, China; Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai 201508, China; Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai 201508, China.
| | - Chao Cao
- Center of Emergency and Critical Medicine in Jinshan Hospital of Fudan University, Shanghai 201508, China; Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai 201508, China; Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai 201508, China
| | - Hanbing Hu
- Center of Emergency and Critical Medicine in Jinshan Hospital of Fudan University, Shanghai 201508, China; Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai 201508, China; Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai 201508, China
| | - Yu Dun
- Center of Emergency and Critical Medicine in Jinshan Hospital of Fudan University, Shanghai 201508, China; Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai 201508, China; Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai 201508, China
| | - Xuanrong Bao
- Center of Emergency and Critical Medicine in Jinshan Hospital of Fudan University, Shanghai 201508, China; Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai 201508, China; Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai 201508, China
| | - Fuli Liu
- Center of Emergency and Critical Medicine in Jinshan Hospital of Fudan University, Shanghai 201508, China; Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai 201508, China; Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai 201508, China
| | - Lin Zhang
- Center of Emergency and Critical Medicine in Jinshan Hospital of Fudan University, Shanghai 201508, China; Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai 201508, China; Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai 201508, China
| | - Jian Zhou
- Shanghai Key Laboratory of Lung Inflammation and Injury, Shanghai 200032, China; Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jie Shen
- Center of Emergency and Critical Medicine in Jinshan Hospital of Fudan University, Shanghai 201508, China; Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai 201508, China; Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai 201508, China.
| |
Collapse
|
3
|
Matalon S, Yu Z, Dubey S, Ahmad I, Stephens EM, Alishlash AS, Meyers A, Cossar D, Stewart D, Acosta EP, Kojima K, Jilling T, Mobley JA. Hemopexin reverses activation of lung eIF2α and decreases mitochondrial injury in chlorine-exposed mice. Am J Physiol Lung Cell Mol Physiol 2024; 326:L440-L457. [PMID: 38150547 PMCID: PMC11281818 DOI: 10.1152/ajplung.00273.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/15/2023] [Accepted: 12/18/2023] [Indexed: 12/29/2023] Open
Abstract
We assessed the mechanisms by which nonencapsulated heme, released in the plasma of mice after exposure to chlorine (Cl2) gas, resulted in the initiation and propagation of acute lung injury. We exposed adult male and female C57BL/6 mice to Cl2 (500 ppm for 30 min), returned them to room air, and injected them intramuscularly with either human hemopexin (hHPX; 5 µg/g BW in 50-µL saline) or vehicle at 1 h post-exposure. Upon return to room air, Cl2-exposed mice, injected with vehicle, developed respiratory acidosis, increased concentrations of plasma proteins in the alveolar space, lung mitochondrial DNA injury, increased levels of free plasma heme, and major alterations of their lung proteome. hHPX injection mice mitigated the onset and development of lung and mitochondrial injury and the increase of plasma heme, reversed the Cl2-induced changes in 83 of 237 proteins in the lung proteome at 24 h post-exposure, and improved survival at 15 days post-exposure. Systems biology analysis of the lung global proteomics data showed that hHPX reversed changes in a number of key pathways including elF2 signaling, verified by Western blotting measurements. Recombinant human hemopexin, generated in tobacco plants, injected at 1 h post-Cl2 exposure, was equally effective in reversing acute lung and mtDNA injury. The results of this study offer new insights as to the mechanisms by which exposure to Cl2 results in acute lung injury and the therapeutic effects of hemopexin.NEW & NOTEWORTHY Herein, we demonstrate that exposure of mice to chlorine gas causes significant changes in the lung proteome 24 h post-exposure. Systems biology analysis of the proteomic data is consistent with damage to mitochondria and activation of eIF2, the master regulator of transcription and protein translation. Post-exposure injection of hemopexin, which scavenges free heme, attenuated mtDNA injury, eIF2α phosphorylation, decreased lung injury, and increased survival.
Collapse
Affiliation(s)
- Sadis Matalon
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Pulmonary Injury and Repair Center, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Zhihong Yu
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Pulmonary Injury and Repair Center, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Shubham Dubey
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Pulmonary Injury and Repair Center, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Israr Ahmad
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Pulmonary Injury and Repair Center, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Emily M Stephens
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Pulmonary Injury and Repair Center, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Ammar Saadoon Alishlash
- Division of Pediatric Pulmonary and Sleep Medicine, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | | | | | | | - Edward P Acosta
- Division of Clinical Pharmacology, Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Kyoko Kojima
- O'Neal Comprehensive Cancer Center, Mass Spectrometry and Proteomics Shared Facility, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Tamas Jilling
- Division of Neonatology, Department of Pediatrics, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - James A Mobley
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Pulmonary Injury and Repair Center, University of Alabama at Birmingham, Birmingham, Alabama, United States
- O'Neal Comprehensive Cancer Center, Mass Spectrometry and Proteomics Shared Facility, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
4
|
Marzec J, Nadadur S. Countermeasures against Pulmonary Threat Agents. J Pharmacol Exp Ther 2024; 388:560-567. [PMID: 37863486 PMCID: PMC10801713 DOI: 10.1124/jpet.123.001822] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 10/02/2023] [Accepted: 10/04/2023] [Indexed: 10/22/2023] Open
Abstract
Inhaled toxicants are used for diverse purposes, ranging from industrial applications such as agriculture, sanitation, and fumigation to crowd control and chemical warfare, and acute exposure can induce lasting respiratory complications. The intentional release of chemical warfare agents (CWAs) during World War I caused life-long damage for survivors, and CWA use is outlawed by international treaties. However, in the past two decades, chemical warfare use has surged in the Middle East and Eastern Europe, with a shift toward lung toxicants. The potential use of industrial and agricultural chemicals in rogue activities is a major concern as they are often stored and transported near populated areas, where intentional or accidental release can cause severe injuries and fatalities. Despite laws and regulatory agencies that regulate use, storage, transport, emissions, and disposal, inhalational exposures continue to cause lasting lung injury. Industrial irritants (e.g., ammonia) aggravate the upper respiratory tract, causing pneumonitis, bronchoconstriction, and dyspnea. Irritant gases (e.g., acrolein, chloropicrin) affect epithelial barrier integrity and cause tissue damage through reactive intermediates or by direct adduction of cysteine-rich proteins. Symptoms of CWAs (e.g., chlorine gas, phosgene, sulfur mustard) progress from airway obstruction and pulmonary edema to acute lung injury (ALI) and acute respiratory distress syndrome (ARDS), which results in respiratory depression days later. Emergency treatment is limited to supportive care using bronchodilators to control airway constriction and rescue with mechanical ventilation to improve gas exchange. Complications from acute exposure can promote obstructive lung disease and/or pulmonary fibrosis, which require long-term clinical care. SIGNIFICANCE STATEMENT: Inhaled chemical threats are of growing concern in both civilian and military settings, and there is an increased need to reduce acute lung injury and delayed clinical complications from exposures. This minireview highlights our current understanding of acute toxicity and pathophysiology of a select number of chemicals of concern. It discusses potential early-stage therapeutic development as well as challenges in developing countermeasures applicable for administration in mass casualty situations.
Collapse
Affiliation(s)
- Jacqui Marzec
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Srikanth Nadadur
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| |
Collapse
|
5
|
He G, Yu W, Li H, Liu J, Tu Y, Kong D, Long Z, Liu R, Peng J, Wang Z, Liu P, Hai C, Yan W, Li W. Alpha-1 antitrypsin protects against phosgene-induced acute lung injury by activating the ID1-dependent anti-inflammatory response. Eur J Pharmacol 2023; 957:176017. [PMID: 37673367 DOI: 10.1016/j.ejphar.2023.176017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/24/2023] [Accepted: 08/24/2023] [Indexed: 09/08/2023]
Abstract
Phosgene is widely used as an industrial chemical, and phosgene inhalation causes acute lung injury (ALI), which may further progress into pulmonary edema. Currently, an antidote for phosgene poisoning is not known. Alpha-1 antitrypsin (α1-AT) is a protease inhibitor used to treat patients with emphysema who are deficient in α1-AT. Recent studies have revealed that α1-AT has both anti-inflammatory and anti-SARS-CoV-2 effects. Herein, we aimed to investigate the role of α1-AT in phosgene-induced ALI. We observed a time-dependent increase in α1-AT expression and secretion in the lungs of rats exposed to phosgene. Notably, α1-AT was derived from neutrophils but not from macrophages or alveolar type II cells. Moreover, α1-AT knockdown aggravated phosgene- and lipopolysaccharide (LPS)-induced inflammation and cell death in human bronchial epithelial cells (BEAS-2B). Conversely, α1-AT administration suppressed the inflammatory response and prevented death in LPS- and phosgene-exposed BEAS-2B cells. Furthermore, α1-AT treatment increased the inhibitor of DNA binding 1 (ID1) gene expression, which suppressed NF-κB pathway activation, reduced inflammation, and inhibited cell death. These data demonstrate that neutrophil-derived α1-AT acts as a self-protective mechanism, which protects against phosgene-induced ALI by activating the ID1-dependent anti-inflammatory response. This study may provide novel strategies for the treatment of patients with phosgene-induced ALI.
Collapse
Affiliation(s)
- Gaihua He
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Weihua Yu
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Hongwei Li
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Jiangzheng Liu
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Yongmei Tu
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Deqin Kong
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Zi Long
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Rui Liu
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Jie Peng
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhao Wang
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Penghui Liu
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Chunxu Hai
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China.
| | - Wenjun Yan
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Rd, Xi'an, 710032, China.
| | - Wenli Li
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
6
|
Matalon S, Yu Z, Dubey S, Ahmad I, Stephens EM, Alishlash AS, Meyers A, Cossar D, Stewart D, Acosta EP, Kojima K, Jilling T, Mobley JA. Hemopexin Reverses Activation of Lung eIF2a and Decreases Mitochondrial Injury in Chlorine Exposed Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.17.553717. [PMID: 37645744 PMCID: PMC10462122 DOI: 10.1101/2023.08.17.553717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
We assessed the mechanisms by which non-encapsulated heme, released in the plasma of mice post exposure to chlorine (Cl 2 ) gas, resulted in the initiation and propagation of acute lung injury. We exposed adult C57BL/6 male and female to Cl 2 (500 ppm for 30 min) in environmental chambers and returned them to room air and injected them intramuscularly with a single dose of human hemopexin (hHPX; 5 µg/ g BW), the most efficient scavenger of heme, 30-60 min post exposure. Concentrations of hHPX in plasma of air and Cl 2 exposed mice were 9081±900 vs. 1879± 293 at 6 h and 2966±463 vs. 1555±250 at 50 h post injection (ng/ml; X±1 SEM=3; p<0.01). Cl 2 exposed mice developed progressive acute lung injury post exposure characterized by increased concentrations of plasma heme, marked inflammatory response, respiratory acidosis and increased concentrations of plasma proteins in the alveolar space. Injection of hHPX decreased the onset of acute lung injury at 24 h post exposure; mean survival, for the saline and hHPX groups were 40 vs. 80% (P<0.001) at 15 d post exposure. Non-supervised global proteomics analysis of mouse lungs at 24 h post exposure, revealed the upregulation of 92 and downregulation of 145 lung proteins. Injection of hHPX at one h post exposure moderated the Cl 2 induced changes in eighty-three of these 237 lung proteins. System biology analysis of the global proteomics data showed that hHPX reversed changes in mitochondrial dysfunction and elF2 and integrin signaling. Western blot analysis of lung tissue showed significant increase of phosphorylated elF2 at 24 h post exposure in vehicle treated mice but normal levels in those injected with hHPX. Similarly, RT-PCR analysis of lung tissue showed that hHPX reversed the onset of mtDNA lesions. A form of recombinant human hemopexin generated in tobacco plants was equally effective in reversing acute lung and mtDNA injury. The results of this study offer new insights as to the mechanisms by which exposure to Cl 2 results in acute lung injury and to the therapeutic effects of hemopexin.
Collapse
|
7
|
Saha S, Sahoo P. Detection of exposed phosgene in household bleach: development of a selective and cost-effective sensing tool. ENVIRONMENTAL SCIENCE. PROCESSES & IMPACTS 2023; 25:1144-1149. [PMID: 37345355 DOI: 10.1039/d3em00171g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/23/2023]
Abstract
Sensing of gaseous environment pollutants and health hazards is in demand these days and in this regard, lethal phosgene has emerged as a leading entrant. In this contribution, we have successfully developed a facile chemodosimeter (ANO) based on an anthracene fluorophore and oxime recognition site with an interesting mechanism to sense lethal phosgene evolved from bleaching powder, a very popular disinfectant and sanitizer. The ANO probe is highly competent in recognizing deadly phosgene in solution and in the gaseous phase with a detection limit in the nanomolar range (1.52 nM). The sensing mechanism is confirmed by UV-vis, emission spectroscopy, mass spectrometry, and computational studies.
Collapse
Affiliation(s)
- Shrabani Saha
- Department of Chemistry, Visva-Bharati University, Santiniketan, 731235, India.
| | - Prithidipa Sahoo
- Department of Chemistry, Visva-Bharati University, Santiniketan, 731235, India.
| |
Collapse
|
8
|
Single-Cell RNA-Sequencing Reveals Epithelial Cell Signature of Multiple Subtypes in Chemically Induced Acute Lung Injury. Int J Mol Sci 2022; 24:ijms24010277. [PMID: 36613719 PMCID: PMC9820093 DOI: 10.3390/ijms24010277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 12/28/2022] Open
Abstract
Alveolar epithelial cells (AECs) play a role in chemically induced acute lung injury (CALI). However, the mechanisms that induce alveolar epithelial type 2 cells (AEC2s) to proliferate, exit the cell cycle, and transdifferentiate into alveolar epithelial type 1 cells (AEC1s) are unclear. Here, we investigated the epithelial cell types and states in a phosgene-induced CALI rat model. Single-cell RNA-sequencing of bronchoalveolar lavage fluid (BALF) samples from phosgene-induced CALI rat models (Gas) and normal controls (NC) was performed. From the NC and Gas BALF samples, 37,245 and 29,853 high-quality cells were extracted, respectively. All cell types and states were identified and divided into 23 clusters; three cell types were identified: macrophages, epithelial cells, and macrophage proliferating cells. From NC and Gas samples, 1315 and 1756 epithelial cells were extracted, respectively, and divided into 11 clusters. The number of AEC1s decreased considerably following phosgene inhalation. A unique SOX9-positive AEC2 cell type that expanded considerably in the CALI state was identified. This progenitor cell type may develop into alveolar cells, indicating its stem cell differentiation potential. We present a single-cell genome-scale transcription map that can help uncover disease-associated cytologic signatures for understanding biological changes and regeneration of lung tissues during CALI.
Collapse
|
9
|
Cao C, Zhang L, Shen J. Phosgene-Induced acute lung injury: Approaches for mechanism-based treatment strategies. Front Immunol 2022; 13:917395. [PMID: 35983054 PMCID: PMC9378823 DOI: 10.3389/fimmu.2022.917395] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Phosgene (COCl2) gas is a chemical intermediate of high-volume production with numerous industrial applications worldwide. Due to its high toxicity, accidental exposure to phosgene leads to various chemical injuries, primarily resulting in chemical-induced lung injury due to inhalation. Initially, the illness is mild and presents as coughing, chest tightness, and wheezing; however, within a few hours, symptoms progress to chronic respiratory depression, refractory pulmonary edema, dyspnea, and hypoxemia, which may contribute to acute respiratory distress syndrome or even death in severe cases. Despite rapid advances in medicine, effective treatments for phosgene-inhaled poisoning are lacking. Elucidating the pathophysiology and pathogenesis of acute inhalation toxicity caused by phosgene is necessary for the development of appropriate therapeutics. In this review, we discuss extant literature on relevant mechanisms and therapeutic strategies to highlight novel ideas for the treatment of phosgene-induced acute lung injury.
Collapse
Affiliation(s)
- Chao Cao
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, China
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai, China
- Training Center of Acute Poisoning Treatment Technology of Fudan University Shanghai Medical College, Shanghai, China
| | - Lin Zhang
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, China
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai, China
| | - Jie Shen
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, China
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai, China
- Training Center of Acute Poisoning Treatment Technology of Fudan University Shanghai Medical College, Shanghai, China
| |
Collapse
|
10
|
RGD-Hydrogel Improves the Therapeutic Effect of Bone Marrow-Derived Mesenchymal Stem Cells on Phosgene-Induced Acute Lung Injury in Rats. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2022; 2022:2743878. [PMID: 35619760 PMCID: PMC9129938 DOI: 10.1155/2022/2743878] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/11/2022] [Accepted: 04/23/2022] [Indexed: 11/17/2022]
Abstract
Mesenchymal stem cells (MSCs) have promising potential in the treatment of various diseases, such as the therapeutic effect of bone marrow-derived MSCs for phosgene-induced acute lung injury (P-ALI). However, MSC-related therapeutics are limited due to poor cell survival, requiring appropriate MSC delivery systems to maximise therapeutic capacity. Biomaterial RGD-hydrogel is a potential cell delivery vehicle as it can mimic the natural extracellular matrix and provide cell adhesion support. The application of RGD-hydrogel in the MSC treatment of respiratory diseases is scarce. This study reports that RGD-hydrogel has good biocompatibility and can increase the secretion of Angiopoietin-1, hepatocyte growth factor, epidermal growth factor, vascular endothelial cell growth factor, and interleukin-10 in vitro MSCs. The hydrogel-encapsulated MSCs could further alleviate P-ALI and show better cell survival in vivo. Overall, RGD-hydrogel could improve the MSC treatment of P-ALI by modulating cell survival and reparative activities. It is exciting to see more and more ways to unlock the therapeutic potential of MSCs.
Collapse
|
11
|
Shao Y, Jiang Z, He D, Shen J. NEDD4 attenuates phosgene-induced acute lung injury through the inhibition of Notch1 activation. J Cell Mol Med 2022; 26:2831-2840. [PMID: 35355403 PMCID: PMC9097839 DOI: 10.1111/jcmm.17296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 02/21/2022] [Accepted: 03/04/2022] [Indexed: 11/28/2022] Open
Abstract
Phosgene gas leakage can cause life-threatening acute lung injury (ALI), which is characterized by inflammation, increased vascular permeability, pulmonary oedema and oxidative stress. Although the downregulation of neuronal precursor cell-expressed developmentally downregulated 4 (NEDD4) is known to be associated with inflammation and oxidative damage, its functions in phosgene-induced ALI remain unclear. In this study, rats with phosgene-induced ALI were intravenously injected with NEDD4-overexpressing lentiviruses to determine the functions of NEDD4 in this inflammatory condition. NEDD4 expression was decreased in the lung parenchyma of phosgene-exposed control rats, whereas its expression level was high in the NEDD4-overexpressing rats. Phosgene exposure increased the wet-to-dry lung weight ratio, but NEDD4 abrogated this effect. NEDD4 overexpression attenuated phosgene-induced lung inflammation, lowering the high lung injury score (based on total protein, inflammatory cells and inflammatory factors in bronchoalveolar lavage fluid) and also reduced phosgene-induced oxidative stress and cell apoptosis. Finally, NEDD4 was found to interact with Notch1, enhancing its ubiquitination and thereby its degradation, thus attenuating the inflammatory responses to ALI. Therefore, we demonstrated that NEDD4 plays a protective role in alleviating phosgene-induced ALI, suggesting that enhancing the effect of NEDD4 may be a new approach for treating phosgene-induced ALI.
Collapse
Affiliation(s)
- Yiru Shao
- Center of Emergency & Intensive Care UnitJinshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health CommissionShanghaiChina
- Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical InjuryJinshan HospitalFudan UniversityShanghaiChina
| | - Zhifeng Jiang
- Center of Emergency & Intensive Care UnitJinshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health CommissionShanghaiChina
- Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical InjuryJinshan HospitalFudan UniversityShanghaiChina
| | - Daikun He
- Center of Emergency & Intensive Care UnitJinshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health CommissionShanghaiChina
- Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical InjuryJinshan HospitalFudan UniversityShanghaiChina
| | - Jie Shen
- Center of Emergency & Intensive Care UnitJinshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health CommissionShanghaiChina
- Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical InjuryJinshan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
12
|
Ajeh EA, Kayode OO, Omoregie IP. Comparative analysis of groundwater quality statuses and associated health risk indices of metals and total hydrocarbons at locations of tank farm in Delta State, Nigeria. Toxicol Rep 2022; 9:404-421. [PMID: 35299872 PMCID: PMC8920876 DOI: 10.1016/j.toxrep.2022.03.001] [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: 06/07/2021] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 11/28/2022] Open
Abstract
The study aimed at assessing the groundwater quality and the associated health implications of oil storage tank farms in Asaba, Oghara, Warri, and Koko towns, in Delta State, Nigeria. Fe, Cr, Cd, Ni, Pb, and V concentrations in the groundwater samples were determined using Atomic Absorption Spectrophotometry (AAS), while total hydrocarbons (THC) concentrations were determined using gas chromatography coupled with a flame ionization detector (GC-FID). The quality index of Warri groundwater was 66.38; being within the range of 51-75 was considered poor quality. The water quality indices (WQI) of Oghara, Koko, and Asaba were 163.79, 161.43, and 129.95 respectively, which were all > 100, hence amounting to very poor water quality status. Results indicated that children in Oghara who are orally exposed to chromium are at risk of cancer. Both adults and children orally exposed to THC in Oghara are also at risk of cancer. Furthermore, THC posed an oral route cancer risk to the children in Koko town. The study showed that chromium posed carcinogenic threats to children in Oghara, while THC posed carcinogenic threats to adults and children in Oghara and children alone in Koko. These risks are liable to be mediated through ingestion of the groundwater of Oghara and Koko by the susceptible groups.
Collapse
Key Words
- CDI, Chronic daily intake
- CRI, Cancer risk index
- Carcinogenic
- DACR, Dermal-associated cancer risk
- Groundwater
- HQ, Harzard quotient
- Health risk
- IACR, Ingestion-associated cancer risk
- LOD, Limit of detection
- LOQ, Limit of quantification
- ORCR, Oral route cancer risk
- Oil exploration
- PCA, Principal component analysis
- SF, Slope factor
- THC, Total hydrocarbons
- Water quality index
Collapse
Affiliation(s)
- Enuneku Alex Ajeh
- Department of Environmental Management and Toxicology, Faculty of Life Sciences, University of Benin, PMB 1154, Benin City, Nigeria
| | - Odeniyi Olalere Kayode
- Ecotoxicology and Environmental Forensics Laboratory, University of Benin, PMB 1154, Benin City, Nigeria
| | - Isibor Patrick Omoregie
- Department of Biological Sciences, College of Science and Technology, Covenant University, PMB 1023, Ota, Ogun State, Nigeria
| |
Collapse
|
13
|
Ahmad I, Molyvdas A, Jian MY, Zhou T, Traylor AM, Cui H, Liu G, Song W, Agarwal A, Jilling T, Aggarwal S, Matalon S. AICAR decreases acute lung injury by phosphorylating AMPK and upregulating heme oxygenase-1. Eur Respir J 2021; 58:2003694. [PMID: 34049949 PMCID: PMC9144003 DOI: 10.1183/13993003.03694-2020] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 05/11/2021] [Indexed: 11/05/2022]
Abstract
AIM We investigated the mechanisms by which N1-(β-d-ribofuranosyl)-5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), an activator of AMP-activated protein kinase (AMPK), decreases lung injury and mortality when administered to mice post exposure to bromine gas (Br2). METHODS We exposed male C57BL/6 mice and heme oxygenase-1 (HO-1)-deficient (HO-1-/-) and corresponding wild-type (WT) littermate mice to Br2 (600 ppm for 45 or 30 min, respectively) in environmental chambers and returned them to room air. AICAR was administered 6 h post exposure (10 mg·kg-1, intraperitoneal). We assessed survival, indices of lung injury, high mobility group box 1 (HMGB1) in the plasma, HO-1 levels in lung tissues and phosphorylation of AMPK and its upstream liver kinase B1 (LKB1). Rat alveolar type II epithelial (L2) cells and human club-like epithelial (H441) cells were also exposed to Br2 (100 ppm for 10 min). After 24 h we measured apoptosis and necrosis, AMPK and LKB1 phosphorylation, and HO-1 expression. RESULTS There was a marked downregulation of phosphorylated AMPK and LKB1 in lung tissues and in L2 and H441 cells post exposure. AICAR increased survival in C57BL/6 but not in HO-1-/- mice. In WT mice, AICAR decreased lung injury and restored phosphorylated AMPK and phosphorylated LKB1 to control levels and increased HO-1 levels in both lung tissues and cells exposed to Br2. Treatment of L2 and H441 cells with small interfering RNAs against nuclear factor erythroid 2-related factor 2 or HO-1 abrogated the protective effects of AICAR. CONCLUSIONS Our data indicate that the primary mechanism for the protective action of AICAR in toxic gas injury is the upregulation of lung HO-1 levels.
Collapse
Affiliation(s)
- Israr Ahmad
- Division of Molecular and Translational Biomedicine, Dept of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Pulmonary Injury and Repair, University of Alabama at Birmingham, Birmingham, AL, USA
- These authors contributed equally to this study
| | - Adam Molyvdas
- Division of Molecular and Translational Biomedicine, Dept of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Pulmonary Injury and Repair, University of Alabama at Birmingham, Birmingham, AL, USA
- These authors contributed equally to this study
| | - Ming-Yuan Jian
- Division of Molecular and Translational Biomedicine, Dept of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Pulmonary Injury and Repair, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ting Zhou
- Division of Molecular and Translational Biomedicine, Dept of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Pulmonary Injury and Repair, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Amie M Traylor
- Division of Nephrology, Dept of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Huachun Cui
- Division of Pulmonary, Allergy, and Critical Care Medicine, Dept of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gang Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Dept of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Weifeng Song
- Dept of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anupam Agarwal
- Division of Nephrology, Dept of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tamas Jilling
- Division of Neonatology, Dept of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Saurabh Aggarwal
- Division of Molecular and Translational Biomedicine, Dept of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Pulmonary Injury and Repair, University of Alabama at Birmingham, Birmingham, AL, USA
- These authors contributed equally as senior authors
| | - Sadis Matalon
- Division of Molecular and Translational Biomedicine, Dept of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Pulmonary Injury and Repair, University of Alabama at Birmingham, Birmingham, AL, USA
- These authors contributed equally as senior authors
| |
Collapse
|
14
|
Deng P, Li L, Liu D, Chen X, Jiang W. Adsorption and dissociation of COCl 2 on the rutile TiO 2(110) surfaces: a systematic first-principles study. Phys Chem Chem Phys 2021; 23:21218-21226. [PMID: 34542142 DOI: 10.1039/d1cp03062k] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The adsorption and dissociation of phosgene (COCl2) molecules on three kinds of rutile TiO2(110) surfaces (stoichiometric: TiO2-Sto; oxygen defective: TiO2-Ov; and substoichiometric: TiO1.875) were investigated based on density functional theory calculations. The nature of interactions between the COCl2 molecule and rutile TiO2(110) surfaces with different degrees of reduction was researched by the analysis of geometries, electron density difference, adsorption energies and density of states (DOS). Computational results show that COCl2 indicates instability and will dissociate directly without the presence of transition states on a substoichiometric TiO1.875(110) surface. The adsorption and dissociation behavior of COCl2 on the rutile surface is not only helpful in providing theoretical support for the clean and efficient degradation of COCl2, but also helpful in elucidating the role of COCl2 as an intermediate product in the carbochlorination of titanium ore.
Collapse
Affiliation(s)
- Pan Deng
- National Engineering Laboratory of Vacuum Metallurgy, Kunming University of Science and Technology, Kunming 650093, Yunnan, P. R. China. .,State Key Laboratory of Complex Nonferrous Metal Resources Clear Utilization in Yunnan Province, Kunming 650093, Yunnan, P. R. China.,Faculty of Metallurgical and Energy Engineering, Kunming University of Science and Technology, Kunming 650093, P. R. China
| | - Liang Li
- National Engineering Laboratory of Vacuum Metallurgy, Kunming University of Science and Technology, Kunming 650093, Yunnan, P. R. China. .,State Key Laboratory of Complex Nonferrous Metal Resources Clear Utilization in Yunnan Province, Kunming 650093, Yunnan, P. R. China.,Faculty of Metallurgical and Energy Engineering, Kunming University of Science and Technology, Kunming 650093, P. R. China.,State Key Laboratory of Vanadium and Titanium Comprehensive Utilization, Pangang Group Research Institute Co. Ltd, Panzhihua, 617000, China
| | - Dachun Liu
- National Engineering Laboratory of Vacuum Metallurgy, Kunming University of Science and Technology, Kunming 650093, Yunnan, P. R. China. .,State Key Laboratory of Complex Nonferrous Metal Resources Clear Utilization in Yunnan Province, Kunming 650093, Yunnan, P. R. China.,Faculty of Metallurgical and Energy Engineering, Kunming University of Science and Technology, Kunming 650093, P. R. China
| | - Xiumin Chen
- National Engineering Laboratory of Vacuum Metallurgy, Kunming University of Science and Technology, Kunming 650093, Yunnan, P. R. China. .,State Key Laboratory of Complex Nonferrous Metal Resources Clear Utilization in Yunnan Province, Kunming 650093, Yunnan, P. R. China.,Faculty of Metallurgical and Energy Engineering, Kunming University of Science and Technology, Kunming 650093, P. R. China
| | - Wenlong Jiang
- National Engineering Laboratory of Vacuum Metallurgy, Kunming University of Science and Technology, Kunming 650093, Yunnan, P. R. China. .,State Key Laboratory of Complex Nonferrous Metal Resources Clear Utilization in Yunnan Province, Kunming 650093, Yunnan, P. R. China.,Faculty of Metallurgical and Energy Engineering, Kunming University of Science and Technology, Kunming 650093, P. R. China
| |
Collapse
|
15
|
Yu W, Wang L, Wang L, Li Y, Zhang N, Zheng K. Quinoline based colorimetric and “turn-off” fluorescent chemosensor for phosgene sensing in solution and vapor phase. Microchem J 2021. [DOI: 10.1016/j.microc.2021.106334] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
16
|
Wang T, Ashrafi A, Modareszadeh P, Deese AR, Chacon Castro MDC, Alemi PS, Zhang L. An Analysis of the Multifaceted Roles of Heme in the Pathogenesis of Cancer and Related Diseases. Cancers (Basel) 2021; 13:4142. [PMID: 34439295 PMCID: PMC8393563 DOI: 10.3390/cancers13164142] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/08/2021] [Accepted: 08/13/2021] [Indexed: 12/28/2022] Open
Abstract
Heme is an essential prosthetic group in proteins and enzymes involved in oxygen utilization and metabolism. Heme also plays versatile and fascinating roles in regulating fundamental biological processes, ranging from aerobic respiration to drug metabolism. Increasing experimental and epidemiological data have shown that altered heme homeostasis accelerates the development and progression of common diseases, including various cancers, diabetes, vascular diseases, and Alzheimer's disease. The effects of heme on the pathogenesis of these diseases may be mediated via its action on various cellular signaling and regulatory proteins, as well as its function in cellular bioenergetics, specifically, oxidative phosphorylation (OXPHOS). Elevated heme levels in cancer cells intensify OXPHOS, leading to higher ATP generation and fueling tumorigenic functions. In contrast, lowered heme levels in neurons may reduce OXPHOS, leading to defects in bioenergetics and causing neurological deficits. Further, heme has been shown to modulate the activities of diverse cellular proteins influencing disease pathogenesis. These include BTB and CNC homology 1 (BACH1), tumor suppressor P53 protein, progesterone receptor membrane component 1 protein (PGRMC1), cystathionine-β-synthase (CBS), soluble guanylate cyclase (sGC), and nitric oxide synthases (NOS). This review provides an in-depth analysis of heme function in influencing diverse molecular and cellular processes germane to disease pathogenesis and the modes by which heme modulates the activities of cellular proteins involved in the development of cancer and other common diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Li Zhang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA; (T.W.); (A.A.); (P.M.); (A.R.D.); (M.D.C.C.C.); (P.S.A.)
| |
Collapse
|
17
|
Alishlash AS, Sapkota M, Ahmad I, Maclin K, Ahmed NA, Molyvdas A, Doran S, Albert CJ, Aggarwal S, Ford DA, Ambalavanan N, Jilling T, Matalon S. Chlorine inhalation induces acute chest syndrome in humanized sickle cell mouse model and ameliorated by postexposure hemopexin. Redox Biol 2021; 44:102009. [PMID: 34044323 PMCID: PMC8167148 DOI: 10.1016/j.redox.2021.102009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/30/2021] [Accepted: 05/11/2021] [Indexed: 11/26/2022] Open
Abstract
Triggering factors of Acute Chest Syndrome (ACS) is a leading cause of death in patients with Sickle Cell Disease (SCD) and targeted therapies are limited. Chlorine (Cl2) inhalation happens frequently, but its role as a potential trigger of ACS has not been determined. In this study, we hypothesized that Cl2 exposure resembling that in the vicinity of industrial accidents induces acute hemolysis with acute lung injury, reminiscent of ACS in humanized SCD mice. When exposed to Cl2 (500 ppm for 30 min), 64% of SCD mice succumbed within 6 h while none of the control mice expressing normal human hemoglobin died (p<0.01). Surviving SCD mice had evidence of acute hemolysis, respiratory acidosis, acute lung injury, and high concentrations of chlorinated palmitic and stearic acids (p<0.05) in their plasmas and RBCs compared to controls. Treatment with a single intraperitoneal dose of human hemopexin 30 min after Cl2 inhalation reduced mortality to around 15% (p<0.01) with reduced hemolysis (decreased RBCs fragility (p<0.001) and returned plasma heme to normal levels (p<0.0001)), improved oxygenation (p<0.0001) and reduced acute lung injury scores (p<0.0001). RBCs from SCD mice had significant levels of carbonylation (which predisposes RBCs to hemolysis) 6 h post-Cl2 exposure which were absent in RBCs of mice treated with hemopexin. To understand the mechanisms leading to carbonylation, we incubated RBCs from SCD mice with chlorinated lipids and identified sickling and increased hemolysis compared to RBCs obtained from control mice and treated similarly. Our study indicates that Cl2 inhalation induces ACS in SCD mice via induction of acute hemolysis, and that post exposure administration of hemopexin reduces mortality and lung injury. Our data suggest that SCD patients are vulnerable in Cl2 exposure incidents and that hemopexin is a potential therapeutic agent.
Collapse
Affiliation(s)
| | - Muna Sapkota
- Department of Pediatrics, School of Medicine, University of Alabama at Birmingham, AL, USA
| | - Israr Ahmad
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, AL, USA
| | - Kelsey Maclin
- Department of Environmental Health Sciences, School of Public Health, University of Alabama at Birmingham, AL, USA
| | - Noor A Ahmed
- Department of Clinical and Diagnostic Sciences, School of Health Professions, University of Alabama at Birmingham, AL, USA
| | - Adam Molyvdas
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, AL, USA
| | - Stephen Doran
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, AL, USA
| | - Carolyn J Albert
- Saint Louis University Department of Biochemistry and Molecular Biology, USA
| | - Saurabh Aggarwal
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, AL, USA
| | - David A Ford
- Saint Louis University Department of Biochemistry and Molecular Biology, USA
| | | | - Tamas Jilling
- Department of Pediatrics, School of Medicine, University of Alabama at Birmingham, AL, USA; Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, AL, USA
| | - Sadis Matalon
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, AL, USA
| |
Collapse
|
18
|
Hobson ST, Richieri RA, Parseghian MH. Phosgene: toxicology, animal models, and medical countermeasures. Toxicol Mech Methods 2021; 31:293-307. [PMID: 33588685 DOI: 10.1080/15376516.2021.1885544] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Phosgene is a gas crucial to industrial chemical processes with widespread production (∼1 million tons/year in the USA, 8.5 million tons/year worldwide). Phosgene's high toxicity and physical properties resulted in its use as a chemical warfare agent during the First World War with a designation of CG ('Choky Gas'). The industrial availability of phosgene makes it a compound of concern as a weapon of mass destruction by terrorist organizations. The hydrophobicity of phosgene exacerbates its toxicity often resulting in a delayed toxidrome as the upper airways are moderately irritated; by the time symptoms appear, significant damage has occurred. As the standard of care for phosgene intoxication is supportive therapy, a pressing need for effective therapeutics and treatment regimens exists. Proposed toxicity mechanisms for phosgene based on human and animal exposures are discussed. Whereas intermediary components in the phosgene intoxication pathways are under continued discussion, generation of reactive oxygen species and oxidative stress is a common factor. As animal models are required for the study of phosgene and for FDA approval via the Animal Rule; the status of existing models and their adherence to Haber's Rule is discussed. Finally, we review the continued search for efficacious therapeutics for phosgene intoxication; and present a rapid post-exposure response that places exogenous human heat shock protein 72, in the form of a cell-penetrating fusion protein (Fv-HSP72), into lung tissues to combat apoptosis resulting from oxidative stress. Despite significant progress, additional work is required to advance effective therapeutics for acute phosgene exposure.
Collapse
Affiliation(s)
- Stephen T Hobson
- Department of Biology and Chemistry, Liberty University, Lynchburg, VA, USA.,Rubicon Biotechnology, Irvine, CA, USA
| | | | | |
Collapse
|
19
|
He DK, Xu N, Shao YR, Shen J. NLRP3 gene silencing ameliorates phosgene-induced acute lung injury in rats by inhibiting NLRP3 inflammasome and proinflammatory factors, but not anti-inflammatory factors. J Toxicol Sci 2020; 45:625-637. [PMID: 33012731 DOI: 10.2131/jts.45.625] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
NOD-like receptor protein 3 (NLRP3) is involved in acute lung injury (ALI), but its exact role in phosgene-induced ALI is not clearly understood. The aim of the study is to explore the potential therapeutic effect of NLRP3 inflammasome modulation in the management of phosgene-induced ALI. ALI was induced in rats by phosgene exposure at 8.33 g/m3 for 5 min, 30 hr before intravenous injection of adenovirus-NLRP3 shRNA (Ad/NLRP3-shRNA). The histological changes in the lung were evaluated. Bronchoalveolar lavage fluid (BALF) neutrophils were counted (smear), and protein content was measured using the BCA assay. The wet/dry ratio of lung tissue (W/D) was measured. TUNEL staining for DNA damage was used to indirectly assess pyroptosis. NLRP3 inflammasome was assessed by immunohistochemistry, RT-PCR, western blotting. Cytokines were measured by ELISA. Histological analyses revealed reduced severity in phosgene-induced ALI with Ad/NLRP3-shRNA pretreatment. TUNEL staining indicated decreased pyroptosis in Psg-Ad/NLRP3-shRNA rats. Decreased mRNA and protein levels of NLRP3 and caspase-1 (all P < 0.05), but not ASC (P > 0.05), were found in Psg-Ad/NLRP3-shRNA rats. Immunohistochemistry revealed that Ad/NLRP3-shRNA pretreatment inhibited NLRP3 inflammasome activation. Reduced level of pro-inflammatory interleukin (IL)-1β, IL-18, IL-33, and tumor necrosis factor (TNF)-α (all P < 0.05), but not of anti-inflammatory IL-4 and IL-10 (all P > 0.05), were found in serum and BALF from Ad/NLRP3-shRNA rats. NLRP3 gene silencing exerts beneficial effects on phosgene-induced lung injury by inhibiting NLRP3 inflammasome activation and pro-inflammatory factors, but not anti-inflammatory factors. Disruption of NLRP3 inflammasome activation might be used as a therapeutic modality for the treatment of phosgene-induced ALI.
Collapse
Affiliation(s)
- Dai-Kun He
- Center of Emergency & Intensive Care Unit, Medical Center of Chemical Injury and Medical Research Centre for Chemical Injury, Emergency and Critical Care, Jinshan Hospital, Fudan University, China
| | - Ning Xu
- Center of Emergency & Intensive Care Unit, Medical Center of Chemical Injury and Medical Research Centre for Chemical Injury, Emergency and Critical Care, Jinshan Hospital, Fudan University, China
| | - Yi-Ru Shao
- Center of Emergency & Intensive Care Unit, Medical Center of Chemical Injury and Medical Research Centre for Chemical Injury, Emergency and Critical Care, Jinshan Hospital, Fudan University, China
| | - Jie Shen
- Center of Emergency & Intensive Care Unit, Medical Center of Chemical Injury and Medical Research Centre for Chemical Injury, Emergency and Critical Care, Jinshan Hospital, Fudan University, China
| |
Collapse
|
20
|
Radbel J, Laskin DL, Laskin JD, Kipen HM. Disease-modifying treatment of chemical threat agent-induced acute lung injury. Ann N Y Acad Sci 2020; 1480:14-29. [PMID: 32726497 DOI: 10.1111/nyas.14438] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 06/10/2020] [Accepted: 06/21/2020] [Indexed: 02/04/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is a highly morbid lung pathology induced by exposure to chemical warfare agents, including vesicants, phosgene, chlorine, and ricin. In this review, we describe the pathology associated with the development of ARDS in humans and experimental models of acute lung injury following animal exposure to these high-priority threat agents. Potential future approaches to disease-modifying treatment used in preclinical animal studies, including antioxidants, anti-inflammatories, biologics, and mesenchymal stem cells, are also described. As respiratory pathologies, including ARDS, are the major cause of morbidity and mortality following exposure to chemical threat agents, understanding mechanisms of disease pathogenesis is key to the development of efficacious therapeutics beyond the primary intervention principle, which remains mechanical ventilation.
Collapse
Affiliation(s)
- Jared Radbel
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Debra L Laskin
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey
| | - Jeffrey D Laskin
- Department of Environmental and Occupational Health, School of Public Health, Rutgers University, Piscataway, New Jersey
| | - Howard M Kipen
- Department of Environmental and Occupational Health, School of Public Health, Rutgers University, Piscataway, New Jersey
| |
Collapse
|
21
|
Addis DR, Molyvdas A, Ambalavanan N, Matalon S, Jilling T. Halogen exposure injury in the developing lung. Ann N Y Acad Sci 2020; 1480:30-43. [PMID: 32738176 DOI: 10.1111/nyas.14445] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/19/2020] [Accepted: 07/30/2020] [Indexed: 12/27/2022]
Abstract
Owing to a high-volume industrial usage of the halogens chlorine (Cl2 ) and bromine (Br2 ), they are stored and transported in abundance, creating a risk for accidental or malicious release to human populations. Despite extensive efforts to understand the mechanisms of toxicity upon halogen exposure and to develop specific treatments that could be used to treat exposed individuals or large populations, until recently, there has been little to no effort to determine whether there are specific features and or the mechanisms of halogen exposure injury in newborns or children. We established a model of neonatal halogen exposure and published our initial findings. In this review, we aim to contrast and compare the findings in neonatal mice exposed to Br2 with the findings published on adult mice exposed to Br2 and the neonatal murine models of bronchopulmonary dysplasia. Despite remarkable similarities across these models in overall alveolar architecture, there are distinct functional and apparent mechanistic differences that are characteristic of each model. Understanding the mechanistic and functional features that are characteristic of the injury process in neonatal mice exposed to halogens will allow us to develop countermeasures that are appropriate for, and effective in, this unique population.
Collapse
Affiliation(s)
- Dylan R Addis
- Department of Anesthesiology and Perioperative Medicine, the University of Alabama at Birmingham School of Medicine, Birmingham, Alabama.,Division of Molecular and Translational Biomedicine, Pulmonary Injury and Repair Center, the University of Alabama at Birmingham School of Medicine, Birmingham, Alabama.,UAB Comprehensive Cardiovascular Center, the University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | - Adam Molyvdas
- Department of Anesthesiology and Perioperative Medicine, the University of Alabama at Birmingham School of Medicine, Birmingham, Alabama.,Division of Molecular and Translational Biomedicine, Pulmonary Injury and Repair Center, the University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | - Namasivayam Ambalavanan
- Division of Neonatology, Department of Pediatrics, the University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | - Sadis Matalon
- Department of Anesthesiology and Perioperative Medicine, the University of Alabama at Birmingham School of Medicine, Birmingham, Alabama.,Division of Molecular and Translational Biomedicine, Pulmonary Injury and Repair Center, the University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | - Tamas Jilling
- Division of Neonatology, Department of Pediatrics, the University of Alabama at Birmingham School of Medicine, Birmingham, Alabama.,Department of Pediatrics, the University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| |
Collapse
|
22
|
Qu Y, Zhang L, He D, Xu N, Tang Y, Shao Y, Shen J. Protective role of mesenchymal stem cells transfected with miRNA-378a-5p in phosgene inhalation lung injury. Biochem Biophys Res Commun 2020; 530:189-195. [PMID: 32828284 DOI: 10.1016/j.bbrc.2020.06.112] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 06/23/2020] [Indexed: 02/08/2023]
Abstract
Phosgene-induced lung injury is an important type of acute lung injury (ALI). Currently, no effective clinical treatment has been developed yet. Our previous study revealed that expressions of 6 miRNAs were significantly increased in phosgene-induced lung injury. The screened miRNA with the most significant effect on hepatocyte growth factor (HGF) expression by mesenchymal stem cells (MSCs) was transfected into MSCs. This study aimed to investigate whether the transfected MSCs had better therapeutic effects than MSCs alone. MSCs were co-cultured with miRNA mimics for 24h and 48h. HGF expression in culture supernatant was detected by ELISA. HGF expression in MSCs was detected by Western blot after being co-cultured with the selected miRNA inhibitor. The transfected MSCs were given to rats suffering from phosgene-induced lung injury. Expressions of TNF-α, IL-6, IL-1β and IL-10, were assayed by ELISA. SP-C mRNA level was tested by RT-PCR. VE-CAD expression was tested by Western blot. We found that miRNA-378a-5p most increased HGF expression among the six miRNAs. After transfection of MSCs with miRNA-378a-5p inhibitor, HGF expression was decreased. Compared with untreated MSCs, MSCs transfected with miRNA-378a-5p exhibited more significant decreases in lung injury score, white blood cell count and protein content while restoring respiratory indexes. Meanwhile, expressions of TNF-α, IL-6, IL-1β were decreased while those of IL-10, SP-C and VE-cadherin were increased. In conclusion, MSCs transfected with miRNA-378a-5p were more effective in treating phosgene-induced lung injury by repairing the secretion of alveolar epithelial cells and improving the permeability of vascular endothelial cells compared with MSCs alone.
Collapse
Affiliation(s)
- Yubei Qu
- Department of Intensive Care Unit, Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Research Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Center of Radiation Injury, Jinshan Hospital, Fudan University, Shanghai, China
| | - Lin Zhang
- Department of Intensive Care Unit, Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Research Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Center of Radiation Injury, Jinshan Hospital, Fudan University, Shanghai, China
| | - Daikun He
- Department of Intensive Care Unit, Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Research Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Center of Radiation Injury, Jinshan Hospital, Fudan University, Shanghai, China
| | - Ning Xu
- Department of Intensive Care Unit, Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Research Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Center of Radiation Injury, Jinshan Hospital, Fudan University, Shanghai, China
| | - Yuedong Tang
- Department of Intensive Care Unit, Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Research Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Center of Radiation Injury, Jinshan Hospital, Fudan University, Shanghai, China
| | - Yiru Shao
- Department of Intensive Care Unit, Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Research Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Center of Radiation Injury, Jinshan Hospital, Fudan University, Shanghai, China
| | - Jie Shen
- Department of Intensive Care Unit, Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Research Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Center of Radiation Injury, Jinshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
23
|
Reactive species generated by heme impair alveolar epithelial sodium channel function in acute respiratory distress syndrome. Redox Biol 2020; 36:101592. [PMID: 32506040 PMCID: PMC7276446 DOI: 10.1016/j.redox.2020.101592] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/08/2020] [Accepted: 05/20/2020] [Indexed: 12/19/2022] Open
Abstract
We previously reported that the highly reactive cell-free heme (CFH) is increased in the plasma of patients with chronic lung injury and causes pulmonary edema in animal model of acute respiratory distress syndrome (ARDS) post inhalation of halogen gas. However, the mechanisms by which CFH causes pulmonary edema are unclear. Herein we report for the first time that CFH and chlorinated lipids (formed by the interaction of halogen gas, Cl2, with plasmalogens) are increased in the plasma of patients exposed to Cl2 gas. Ex vivo incubation of red blood cells (RBC) with halogenated lipids caused oxidative damage to RBC cytoskeletal protein spectrin, resulting in hemolysis and release of CFH. Patch clamp and short circuit current measurements revealed that CFH inhibited the activity of amiloride-sensitive epithelial Na+ channel (ENaC) and cation sodium (Na+) channels in mouse alveolar cells and trans-epithelial Na+ transport across human airway cells with EC50 of 125 nM and 500 nM, respectively. Molecular modeling identified 22 putative heme-docking sites on ENaC (energy of binding range: 86-1563 kJ/mol) with at least 2 sites within its narrow transmembrane pore, potentially capable of blocking Na+ transport across the channel. A single intramuscular injection of the heme-scavenging protein, hemopexin (4 μg/kg body weight), one hour post halogen gas exposure, decreased plasma CFH and improved lung ENaC activity in mice. In conclusion, results suggested that CFH mediated inhibition of ENaC activity may be responsible for pulmonary edema post inhalation injury.
Collapse
|
24
|
DeLey Cox VE, Hartog MA, Pueblo E, Racine M, Jennings L, Tressler J, Tuet WY, Stone S, Pierce SA, Thompson L, Dukes A, Hoard-Fruchey H, Wong B, McCranor BJ. Methylene blue and monosodium glutamate improve neurologic signs after fluoroacetate poisoning. Ann N Y Acad Sci 2020; 1479:196-209. [PMID: 32285953 DOI: 10.1111/nyas.14347] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 12/17/2022]
Abstract
Fluoroacetate (FA) is a tasteless, odorless, water-soluble metabolic poison with severe toxicological effects. Characterized in the mid-1900s, it has been used as a rodenticide but is comparably lethal to all mammals. Many countries have restricted its use, and modern-day accidental human exposures are rare, but recently, concerns have been raised about its application as a chemical weapon with no known antidote. A combined treatment of methylene blue (MB), an antioxidant, and monosodium glutamate (MSG), a precursor of the citric acid cycle substrate alpha-ketoglutarate, has been recommended as an effective countermeasure; however, no peer-reviewed articles documenting the efficacy of this therapy have been published. Using a rodent model, we assessed the effects of MB and MSG on the neurologic, cardiac, and pulmonary systems. Transcriptomic analysis was used to elucidate inflammatory pathway activation and guide bioassays, which revealed the advantages and disadvantages of these candidate countermeasures. Results show that MB and MSG can reduce neurologic signs observed in rats exposed to sodium FA and improve some effects of intoxication. However, while this strategy resolved some signs of intoxication, ultimately it was unable to significantly reduce lethality.
Collapse
Affiliation(s)
- Vanessa E DeLey Cox
- Pharmaceutical Sciences Department, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Matthew A Hartog
- Pharmaceutical Sciences Department, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Erin Pueblo
- Pharmaceutical Sciences Department, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Michelle Racine
- Pharmaceutical Sciences Department, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Laura Jennings
- Pharmaceutical Sciences Department, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Justin Tressler
- Pharmaceutical Sciences Department, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Wing Y Tuet
- Pharmaceutical Sciences Department, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Samuel Stone
- Pharmaceutical Sciences Department, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Samuel A Pierce
- Pharmaceutical Sciences Department, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Lily Thompson
- Pharmaceutical Sciences Department, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Aliyah Dukes
- Pharmaceutical Sciences Department, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Heidi Hoard-Fruchey
- Pharmaceutical Sciences Department, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Benjamin Wong
- Pharmaceutical Sciences Department, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Bryan J McCranor
- Pharmaceutical Sciences Department, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| |
Collapse
|
25
|
Addis DR, Lambert JA, Ren C, Doran S, Aggarwal S, Jilling T, Matalon S. Vascular Endothelial Growth Factor-121 Administration Mitigates Halogen Inhalation-Induced Pulmonary Injury and Fetal Growth Restriction in Pregnant Mice. J Am Heart Assoc 2020; 9:e013238. [PMID: 32009528 PMCID: PMC7033856 DOI: 10.1161/jaha.119.013238] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 01/07/2020] [Indexed: 12/16/2022]
Abstract
Background Circulating levels of sFLT-1 (soluble fms-like tyrosine kinase 1), the extracellular domain of vascular endothelial growth factor (VEGF) receptor 1, and its ratio to levels of placental growth factor are markers of the occurrence and severity of preeclampsia. Methods and Results C57BL/6 pregnant mice on embryonic day 14.5 (E14.5), male, and non-pregnant female mice were exposed to air or to Br2 at 600 ppm for 30 minutes and were treated with vehicle or with VEGF-121 (100 μg/kg, subcutaneously) daily, starting 48 hours post-exposure. Plasma, bronchoalveolar lavage fluid, lungs, fetuses, and placentas were collected 120 hours post-exposure. In Br2-exposed pregnant mice, there was a time-dependent and significant increase in plasma levels of sFLT-1 which correlated with increases in mouse lung wet/dry weights and bronchoalveolar lavage fluid protein content. Supplementation of exogenous VEGF-121 improved survival and weight gain, reduced lung wet/dry weights, decreased bronchoalveolar lavage fluid protein levels, enhanced placental development, and improved fetal growth in pregnant mice exposed to Br2. Exogenous VEGF-121 administration had no effect in non-pregnant mice. Conclusions These results implicate inhibition of VEGF signaling driven by sFLT-1 overexpression as a mechanism of pregnancy-specific injury leading to lung edema, maternal mortality, and fetal growth restriction after bromine gas exposure.
Collapse
Affiliation(s)
- Dylan R. Addis
- Division of Cardiothoracic AnesthesiologyDepartment of Anesthesiology and Perioperative MedicineUniversity of Alabama at BirminghamBirminghamALUSA
- Division of Molecular and Translational BiomedicineDepartment of Anesthesiology and Perioperative MedicineUniversity of Alabama at BirminghamBirminghamALUSA
- UAB Comprehensive Cardiovascular CenterUniversity of Alabama at BirminghamBirminghamALUSA
- University of Alabama School of MedicineBirminghamALUSA
| | - James A. Lambert
- Division of Molecular and Translational BiomedicineDepartment of Anesthesiology and Perioperative MedicineUniversity of Alabama at BirminghamBirminghamALUSA
- Graduate Biomedical SciencesBiochemistry, Structural and Stem Cell Biology ThemeUniversity of Alabama at BirminghamBirminghamALUSA
- University of Alabama School of MedicineBirminghamALUSA
| | - Changchun Ren
- Department of PediatricsDivision of NeonatologyUniversity of Alabama at BirminghamBirminghamALUSA
- University of Alabama School of MedicineBirminghamALUSA
| | - Stephen Doran
- Division of Molecular and Translational BiomedicineDepartment of Anesthesiology and Perioperative MedicineUniversity of Alabama at BirminghamBirminghamALUSA
- University of Alabama School of MedicineBirminghamALUSA
| | - Saurabh Aggarwal
- Division of Molecular and Translational BiomedicineDepartment of Anesthesiology and Perioperative MedicineUniversity of Alabama at BirminghamBirminghamALUSA
- University of Alabama School of MedicineBirminghamALUSA
| | - Tamas Jilling
- Department of PediatricsDivision of NeonatologyUniversity of Alabama at BirminghamBirminghamALUSA
- University of Alabama School of MedicineBirminghamALUSA
| | - Sadis Matalon
- Division of Molecular and Translational BiomedicineDepartment of Anesthesiology and Perioperative MedicineUniversity of Alabama at BirminghamBirminghamALUSA
- UAB Comprehensive Cardiovascular CenterUniversity of Alabama at BirminghamBirminghamALUSA
- University of Alabama School of MedicineBirminghamALUSA
| |
Collapse
|