1
|
Matalon S. The long road to Ithaca: a physiologist's journey. Am J Physiol Cell Physiol 2025; 328:C1526-C1534. [PMID: 39993005 DOI: 10.1152/ajpcell.00030.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/10/2025] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
It was an honor to be asked to deliver the Walter B. Cannon Lecture during the 2024 American Physiological Summit meeting. Dr. Cannon served as president of the American Physiological Society from 1914-1916. He coined the term "fight or flight" to describe an animal's response to threats and the concept of Homeostasis. He was the consummate physician-scientist, an outstanding mentor and teacher, a prolific writer, and a humanitarian. The title of my lecture is based on a poem entitled "Ithaca," written by the Greek poet C. P. Cavafy, who recounts the 10 yr travels of Ulysses, from Troy to his home, Ithaca. Odysseus had to overcome many obstacles to survive this long journey. Like Odysseus, I encountered myriad of professional and health problems. But, I also have experienced the thrill of contributing to scientific knowledge, the satisfaction of watching my mentees develop into independent scientists, the excitement of teaching respiration physiology to medical and professional students, and the pleasure of being of service to my discipline by serving as Editor of the American Journal of Physiology-Lung Cellular and Molecular Physiology and of Physiological Reviews. During my career, I have been interested in identifying the basic mechanisms by which oxidant gases and pathogens damage the blood gas barrier resulting in acute and chronic lung injury. In this brief review, I summarize the results of current studies implicating free heme as a major mediator of acute lung injury and our efforts to develop recombinant forms of human hemopexin, as a countermeasure.
Collapse
Affiliation(s)
- Sadis Matalon
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, Heersink School of Medicine UAB | The University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
2
|
Carlson HL, Ford DA. Human lung microvascular endothelial cell protein modification by 2-chlorohexadecanoic acid: RhoA mediates 2-chlorohexadecanoic acid-elicited endothelial activation. Redox Biol 2025; 82:103596. [PMID: 40117888 PMCID: PMC11979405 DOI: 10.1016/j.redox.2025.103596] [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: 02/25/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/23/2025] Open
Abstract
Chlorolipids are produced during the neutrophil respiratory burst as a result of myeloperoxidase (MPO)-generated hypochlorous acid (HOCl) targeting the vinyl ether bond of plasmalogen phospholipids. The initial products of this reaction are 2-chlorofatty aldehydes (2-ClFALDs), which are subsequently oxidized to 2-chlorofatty acids (2-ClFAs). 2-Chlorohexadecanoic acid (2-ClHA) is the 16-carbon 2-ClFA species, and previous studies have shown that increased levels of plasma 2-ClHA associate with acute respiratory distress syndrome (ARDS)-caused mortality in human sepsis. 2-ClHA causes endothelial barrier dysfunction and increases neutrophil and platelet adherence to the endothelium. In this study, click chemistry analogs of 2-ClHA and hexadecanoic acid (HA) were used to identify proteins covalently modified by 2-ClHA and HA in human lung microvascular endothelial cells (HLMVECs). Eleven proteins were specifically modified by 2-ClHA, and an additional one hundred and ninety-four proteins were modified by both 2-ClHA and HA. STRING analysis of 2-ClHA-modified proteins revealed a network of proteins with RhoA as a hub. RhoA is one of the proteins specifically modified by 2-ClHA and not HA. The RhoA inhibitors, Rhosin and C3, inhibited both 2-ClHA-elicited HLMVEC barrier dysfunction and angiopoietin-2 (Ang-2) release from HLMVEC. Further studies showed 2-ClHA activates HLMVEC RhoA activity. The specificity of the 2-ClHA-RhoA pathway for endothelial activation was further confirmed since HA did not cause HLMVEC barrier dysfunction, Ang-2 release and RhoA activation. Collectively, these studies have identified multiple proteins modified exclusively by 2-ClHA in HLMVECs, including RhoA. These proteomics studies led to the key finding that RhoA is an important mediator of 2-ClHA-caused endothelial barrier dysfunction.
Collapse
Affiliation(s)
- Haley L Carlson
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA
| | - David A Ford
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA.
| |
Collapse
|
3
|
Dubey S, Yu Z, Stephens EM, Lazrak A, Ahmad I, Aggarwal S, Andrabi S, Hossain MI, Jilling T, Fernandez SR, Bartels JL, Lapi SE, Mobley JA, Pastukh VM, Gillespie MN, Matalon S. Oxidative damage to lung mitochondrial DNA is a key contributor to the development of chemical lung injury. Redox Biol 2025; 82:103624. [PMID: 40209617 PMCID: PMC12013491 DOI: 10.1016/j.redox.2025.103624] [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: 02/18/2025] [Revised: 03/19/2025] [Accepted: 03/27/2025] [Indexed: 04/12/2025] Open
Abstract
Humans exposed to chlorine (Cl2) due to industrial accidents or acts of terrorism may develop lung injury culminating to Acute Respiratory Distress syndrome and death from respiratory failure. Early molecular targets of inhaled oxidant gases suitable for pharmacologic modulation have not been established. Because the mitochondrial genome is highly sensitive to oxidant stress, we tested the hypothesis that mice exposure to Cl2 gas causes oxidative damage to the mitochondrial DNA (mtDNA) that triggers the development of acute and chronic lung injury. Cl2 gas-exposed C57BL/6 mice and returned to room air, developed progressive loss of lung DNA glycosylase OGG1, followed by oxidative mtDNA damage. This resulted in activation of inflammatory pathways by circulating DNA, progressive increased airway resistance, alveolar injury and acute pulmonary edema due to loss of epithelial amiloride-sensitive sodium channels. Mice not succumbing acutely displayed a delayed syndrome of progressive increase in airway resistance and emphysematous-like changes in lung morphology. Global proteomics of lungs harvested 24 h post Cl2 exposure revealed alterations in over 1500 lung proteins, including 14 key mitochondrial proteins. Intranasal instillation of a recombinant protein targeting OGG1 to mitochondria (mitoOGG1) at 1 h post exposure decreased oxidized lung mtDNA, alterations to the lung and mitochondrial proteomes, severity of the acute and delayed lung injury and increased survival. These data show that injury to the mt-genome is a key contributor to the development of acute and chronic lung injury after Cl2 gas exposure and point to mtDNA oxidation as a target for pharmacologic intervention.
Collapse
Affiliation(s)
- Shubham Dubey
- Department of Anesthesiology and Perioperative Medicine, USA
| | - Zhihong Yu
- Department of Anesthesiology and Perioperative Medicine, USA
| | | | - Ahmed Lazrak
- Department of Anesthesiology and Perioperative Medicine, USA
| | - Israr Ahmad
- Department of Anesthesiology and Perioperative Medicine, USA
| | | | | | | | - Tamas Jilling
- Department of Pediatrics, Division of Neonatology, USA
| | - Solana R Fernandez
- Department of Radiology, The Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jennifer L Bartels
- Department of Radiology, The Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Suzanne E Lapi
- Department of Radiology, The Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - James A Mobley
- Department of Anesthesiology and Perioperative Medicine, USA
| | - Viktor M Pastukh
- Department of Pharmacology and Center for Lung Biology, University of South Alabama College of Medicine, Mobile, AL, 36688, USA
| | - Mark N Gillespie
- Department of Pharmacology and Center for Lung Biology, University of South Alabama College of Medicine, Mobile, AL, 36688, USA
| | - Sadis Matalon
- Department of Anesthesiology and Perioperative Medicine, USA.
| |
Collapse
|
4
|
Manikanta, NaveenKumar SK, Thushara RM, Hemshekhar M, Sumedini ML, Sunitha K, Kemparaju K, Girish KS. Counteraction of unconjugated bilirubin against heme-induced toxicity in platelets. Thromb Res 2024; 244:109199. [PMID: 39467509 DOI: 10.1016/j.thromres.2024.109199] [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: 05/06/2024] [Revised: 10/09/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024]
Abstract
Platelets are essential for normal hemostasis and thrombosis but become hyperactive in hemolytic disorders. Cell-free heme is known to be toxic to platelets and endothelial cells, playing a significant role in the progression of pathological complications in various hemolytic conditions. The abnormal activation of circulatory platelets results in micro/macrovascular thrombosis and clot formation in the lungs, worsening the disease. This work aimed to establish the potent bioactive molecule that can regulate the heme-induced toxicity in platelets. We found that unconjugated bilirubin (UCB), an endogenous antioxidant and a byproduct of heme degradation, exhibited a higher protective effect against hemin-induced platelet aggregation and activation. This protective effect could mainly be due to reducing ROS and lipid peroxidation-mediated ferroptosis in hemin-treated platelets. Further experiments suggested that by blocking the interaction between hemin and the CLEC-2 receptor, UCB regulates the downstream Syk phosphorylation, a key event in hemin-induced platelet toxicity. Thus, UCB is emerging as a natural regulatory molecule that mitigates hemin-induced platelet toxicity and holds promise as an adjunctive therapy for managing platelet-associated complications, particularly in hemolytic disorders.
Collapse
Affiliation(s)
- Manikanta
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysuru 570006, India
| | | | - Ram M Thushara
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysuru 570006, India
| | - Mahadevappa Hemshekhar
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysuru 570006, India
| | - Mysuru L Sumedini
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysuru 570006, India
| | - Kabburahalli Sunitha
- Department of Studies and Research in Biochemistry, Tumkur University, Tumakuru 572103, India
| | - Kempaiah Kemparaju
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysuru 570006, India.
| | - Kesturu S Girish
- Department of Studies and Research in Biochemistry, Tumkur University, Tumakuru 572103, India.
| |
Collapse
|
5
|
Dubey S, Yu Z, Stephens EM, Lazrak A, Ahmad I, Aggarwal S, Andrabi S, Hossain MI, Jilling T, Fernadez SR, Bartels JL, Lapi SE, Mobley J, Pastukh VM, Gillespie M, Matalon S. Oxidative Injury to Lung Mitochondrial DNA is a Key Contributor for the Development of Chemical Lung Injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624949. [PMID: 39651262 PMCID: PMC11623505 DOI: 10.1101/2024.11.22.624949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
The mechanisms and extent to which inhalation of oxidant gases damage the mitochondrial genome contributing to the development of acute and chronic lung injury have not been investigated. C57BL/6 mice exposed to chlorine (Cl 2 ) gas and returned to room air, developed progressive loss of lung DNA glycosylase OGG1, significant oxidative injury to mtDNA, decreased intact lung mitochondrial (mt) DNA, generation of inflammatory pathway by DAMPs causing airway and alveolar injury with significant mortality. Global proteomics identified over 1400 lung proteins with alteration of key mitochondrial proteins at 24 h post Cl 2 exposure. Intranasal instillation of a recombinant protein containing mitochondrial targeted OGG1 (mitoOGG1) post exposure, decreased oxidative injury to mtDNA, lung mitochondrial proteome, severity of the acute and chronic lung injury and increased survival. These data show that injury to the mt-genome is a key contributor to the development of acute and chronic lung injury.
Collapse
|
6
|
Shafi H, Lora AJ, Donow HM, Aggarwal S, Fu P, Wang T, Mansour HM. Advanced Spray-Dried Inhalable Microparticles/Nanoparticles of an Innovative Mitophagy Activator for Targeted Lung Delivery: Design, Comprehensive Characterization, Human Lung Cell Culture, and In Vitro Aerosol Dispersion Performance. ACS Pharmacol Transl Sci 2024; 7:3540-3558. [PMID: 39539257 PMCID: PMC11555509 DOI: 10.1021/acsptsci.4c00436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 11/16/2024]
Abstract
Urolithin A (UA) has demonstrated the ability to stimulate mitophagy and enhance mitochondrial and cellular health in skeletal muscles in humans after oral administration. It is hypothesized that targeted delivery of UA as inhaled dry powders to the lungs will enhance mitochondrial health through mitochondrial biogenesis. This study aimed to engineer inhalable excipient-free powders of UA as dry powder inhalers (DPIs) for targeted pulmonary delivery. The particles were designed by particle engineering from dilute organic solutions of UA using the state-of-the-art spray drying technology in a closed mode. Comprehensive physicochemical characterization and advanced microscopy techniques were conducted to examine phase behavior, molecular properties, and particle properties, which are necessary for the rational design of advanced pulmonary inhalation aerosols. Molecular fingerprinting was conducted by using attenuated total reflectance-Fourier transform infrared (ATR-FTIR) spectroscopy and Raman spectroscopy. Chemical imaging and mapping were conducted using confocal Raman microscopy (CRM) and IR microscopy. The advanced spray-dried (SD) excipient-free powders were successfully produced at different spraying pump feed rates and exhibited favorable molecular and particle properties. The excipient-free SD powders exhibited outstanding in vitro aerosol dispersion performance with an FDI-approved human DPI device (Neohaler) and correlated with the spray drying pump rate. In vitro, cell viability of various human pulmonary cells from different lung regions demonstrated biocompatibility and safety at different doses of UA. The transepithelial electrical resistance (TEER) assay shows that UA maintains cell membrane integrity and barrier tightness, indicating its potential for safe and effective localized drug delivery without long-term adverse effects. These results demonstrated that UA has favorable physicochemical and in vitro properties for inhalation and can be successfully engineered into excipient-free inhalable microparticles/nanoparticles as DPIs.
Collapse
Affiliation(s)
- Hasham Shafi
- Florida
International University Center for Translational Science, Port St. Lucie, Florida 34987, United States
| | - Andrea J. Lora
- Florida
International University Center for Translational Science, Port St. Lucie, Florida 34987, United States
| | - Haley M. Donow
- Florida
International University Center for Translational Science, Port St. Lucie, Florida 34987, United States
| | - Saurabh Aggarwal
- Dept.
of Cellular and Molecular Medicine, FIU
Herbert Wertheim College of Medicine, Miami, Florida 33199, United States
| | - Panfeng Fu
- Florida
International University Center for Translational Science, Port St. Lucie, Florida 34987, United States
- Dept.
of Environmental Health Sciences, FIU Robert
Stempel College of Public Health & Social Work, Miami, Florida 33199, United States
| | - Ting Wang
- Florida
International University Center for Translational Science, Port St. Lucie, Florida 34987, United States
- Dept.
of Cellular and Molecular Medicine, FIU
Herbert Wertheim College of Medicine, Miami, Florida 33199, United States
- Dept.
of Environmental Health Sciences, FIU Robert
Stempel College of Public Health & Social Work, Miami, Florida 33199, United States
| | - Heidi M. Mansour
- Florida
International University Center for Translational Science, Port St. Lucie, Florida 34987, United States
- Dept.
of Cellular and Molecular Medicine, FIU
Herbert Wertheim College of Medicine, Miami, Florida 33199, United States
- Dept.
of Environmental Health Sciences, FIU Robert
Stempel College of Public Health & Social Work, Miami, Florida 33199, United States
- Dept.
of Biomedical Engineering, FIU College of
Engineering & Computing, Miami, Florida 33199, United States
| |
Collapse
|
7
|
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
|
8
|
Achanta S, Gentile MA, Albert CJ, Schulte KA, Pantazides BG, Crow BS, Quiñones-González J, Perez JW, Ford DA, Patel RP, Blake TA, Gunn MD, Jordt SE. Recapitulation of human pathophysiology and identification of forensic biomarkers in a translational model of chlorine inhalation injury. Am J Physiol Lung Cell Mol Physiol 2024; 326:L482-L495. [PMID: 38318664 PMCID: PMC11281795 DOI: 10.1152/ajplung.00162.2023] [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: 05/18/2023] [Revised: 01/16/2024] [Accepted: 01/27/2024] [Indexed: 02/07/2024] Open
Abstract
Chlorine gas (Cl2) has been repeatedly used as a chemical weapon, first in World War I and most recently in Syria. Life-threatening Cl2 exposures frequently occur in domestic and occupational environments, and in transportation accidents. Modeling the human etiology of Cl2-induced acute lung injury (ALI), forensic biomarkers, and targeted countermeasures development have been hampered by inadequate large animal models. The objective of this study was to develop a translational model of Cl2-induced ALI in swine to understand toxico-pathophysiology and evaluate whether it is suitable for screening potential medical countermeasures and to identify biomarkers useful for forensic analysis. Specific pathogen-free Yorkshire swine (30-40 kg) of either sex were exposed to Cl2 (≤240 ppm for 1 h) or filtered air under anesthesia and controlled mechanical ventilation. Exposure to Cl2 resulted in severe hypoxia and hypoxemia, increased airway resistance and peak inspiratory pressure, and decreased dynamic lung compliance. Cl2 exposure resulted in increased total leucocyte and neutrophil counts in bronchoalveolar lavage fluid, vascular leakage, and pulmonary edema compared with the air-exposed group. The model recapitulated all three key histopathological features of human ALI, such as neutrophilic alveolitis, deposition of hyaline membranes, and formation of microthrombi. Free and lipid-bound 2-chlorofatty acids and chlorotyrosine-modified proteins (3-chloro-l-tyrosine and 3,5-dichloro-l-tyrosine) were detected in plasma and lung tissue after Cl2 exposure. In this study, we developed a translational swine model that recapitulates key features of human Cl2 inhalation injury and is suitable for testing medical countermeasures, and validated chlorinated fatty acids and protein adducts as biomarkers of Cl2 inhalation.NEW & NOTEWORTHY We established a swine model of chlorine gas-induced acute lung injury that exhibits several features of human acute lung injury and is suitable for screening potential medical countermeasures. We validated chlorinated fatty acids and protein adducts in plasma and lung samples as forensic biomarkers of chlorine inhalation.
Collapse
Affiliation(s)
- Satyanarayana Achanta
- Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Michael A Gentile
- Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Carolyn J Albert
- Department of Biochemistry and Molecular Biology, Saint Louis University, St. Louis, Missouri, United States
| | - Kevin A Schulte
- Department of Biochemistry and Molecular Biology, Saint Louis University, St. Louis, Missouri, United States
| | - Brooke G Pantazides
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, Georgia, United States
| | - Brian S Crow
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, Georgia, United States
| | - Jennifer Quiñones-González
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, Georgia, United States
| | - Jonas W Perez
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, Georgia, United States
| | - David A Ford
- Department of Biochemistry and Molecular Biology, Saint Louis University, St. Louis, Missouri, United States
| | - Rakesh P Patel
- Center for Free Radical Biology and Lung Injury and Repair Center, The University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Thomas A Blake
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, Georgia, United States
| | - Michael D Gunn
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States
| | - Sven E Jordt
- Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina, United States
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina, United States
- Integrated Toxicology & Environmental Health Program, Duke University, Durham, North Carolina, United States
| |
Collapse
|
9
|
Wang S, Ma J, Li X, Xian X, Tan G, Cai H, Yang B, Zhang A, Guo J, Gu G, Meng Z, Fu B. EGR-1 Contributes to Pulmonary Edema by Regulating the Epithelial Sodium Channel in Lipopolysaccharide-Induced Acute Lung Injury. Immunol Invest 2023; 52:925-939. [PMID: 37732637 DOI: 10.1080/08820139.2023.2256778] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Acute lung injury (ALI) is a common lung disease with increasing morbidity and mortality rates due to the lack of specific drugs. Impaired alveolar fluid clearance (AFC) is a primary pathological feature of ALI. Epithelial sodium channel (ENaC) is a primary determinant in regulating the transport of Na+ and the clearance of alveolar edema fluid. Therefore, ENaC is an important target for the development of drugs for ALI therapy. However, the role of ENaC in the progression of ALI remains unclear. Inhibition of early growth response factor (EGR-1) expression has been reported to induce a protective effect on ALI; therefore, we evaluated whether EGR-1 participates in the progression of ALI by regulating ENaC-α in alveolar epithelium. We investigated the potential mechanism of EGR-1-mediated regulation of ENaC in ALI. We investigated whether EGR-1 aggravates the pulmonary edema response in ALI by regulating ENaC. ALI mouse models were established by intrabronchial injection of lipopolysaccharides (LPS). Lentiviruses with EGR-1 knockdown were transfected into LPS-stimulated A549 cells. We found that EGR-1 expression was upregulated in the lung tissues of ALI mice and in LPS-induced A549 cells, and was negatively correlated with ENaC-α expression. Knockdown of EGR-1 increased ENaC-α expression and relieved cellular edema in ALI. Moreover, EGR-1 regulated ENaC-α expression at the transcriptional level, and correspondingly promoted pulmonary edema and aggravated ALI symptoms. In conclusion, our study demonstrated that EGR-1 could promote pulmonary edema by downregulating ENaC-α at the transcriptional level in ALI. Our study provides a new potential therapeutic strategy for treatment of ALI.
Collapse
Affiliation(s)
- Song Wang
- Precision Biomedical Laboratory, Liaocheng People's Hospital, Liaocheng, China
| | - Jing Ma
- Office of Academic Research, Liaocheng People's Hospital, Liaocheng, China
| | - Xin Li
- Department of Clinical Laboratory, Zibo Central Hospital, Zibo, China
| | - Xinmiao Xian
- Precision Biomedical Laboratory, Liaocheng People's Hospital, Liaocheng, China
| | - Guikun Tan
- Pharmacy Department, Liaocheng Woman and Child Health Care Hospital, Liaocheng, China
| | - Hongwei Cai
- Department of Critical Care Medicine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Bingwu Yang
- Precision Biomedical Laboratory, Liaocheng People's Hospital, Liaocheng, China
| | - Anqi Zhang
- Central Laboratory, Liaocheng People's Hospital, Liaocheng, China
| | - Jianran Guo
- Precision Biomedical Laboratory, Liaocheng People's Hospital, Liaocheng, China
| | - Guohao Gu
- Precision Biomedical Laboratory, Liaocheng People's Hospital, Liaocheng, China
| | - Zhen Meng
- Biomedical Laboratory, Medical School of Liaocheng University, Liaocheng, China
| | - Bo Fu
- Precision Biomedical Laboratory, Liaocheng People's Hospital, Liaocheng, China
| |
Collapse
|
10
|
Shovlin CL, Vizcaychipi MP. Vascular inflammation and endothelial injury in SARS-CoV-2 infection: the overlooked regulatory cascades implicated by the ACE2 gene cluster. QJM 2023; 116:629-634. [PMID: 32777054 PMCID: PMC7454888 DOI: 10.1093/qjmed/hcaa241] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 12/14/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) has presented physicians with an unprecedented number of challenges and mortality. The basic question is why, in contrast to other 'respiratory' viruses, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection can result in such multi-systemic, life-threatening complications and a severe pulmonary vasculopathy. It is widely known that SARS-CoV-2 uses membrane-bound angiotensin-converting enzyme 2 (ACE2) as a receptor, resulting in internalization of the complex by the host cell. We discuss the evidence that failure to suppress coronaviral replication within 5 days results in sustained downregulation of ACE2 protein expression and that ACE2 is under negative-feedback regulation. We then expose openly available experimental repository data that demonstrate the gene for ACE2 lies in a novel cluster of inter-regulated genes on the X chromosome including PIR encoding pirin (quercetin 2,3-dioxygenase), and VEGFD encoding the predominantly lung-expressed vascular endothelial growth factor D. The five double-elite enhancer/promoters pairs that are known to be operational, and shared read-through lncRNA transcripts, imply that ongoing SARS-CoV-2 infection will reduce host defences to reactive oxygen species, directly generate superoxide O2·- and H2O2 (a ' ROS storm'), and impair pulmonary endothelial homeostasis. Published cellular responses to oxidative stress complete the loop to pathophysiology observed in severe COVID-19. Thus, for patients who fail to rapidly suppress viral replication, the newly appreciated ACE2 co-regulated gene cluster predicts delayed responses that would account for catastrophic deteriorations. We conclude that ACE2 homeostatic drives provide a unified understanding that should help optimize therapeutic approaches during the wait until safe, effective vaccines and antiviral therapies for SARS-CoV-2 are delivered.
Collapse
Affiliation(s)
- Claire L Shovlin
- NHLI Vascular Science, Imperial College London, UK
- Respiratory Medicine, Imperial College Healthcare NHS Trust, London UK
| | | |
Collapse
|
11
|
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
|
12
|
Freeman SL, Oliveira ASF, Gallio AE, Rosa A, Simitakou MK, Arthur CJ, Mulholland AJ, Cherepanov P, Raven EL. Heme binding to the SARS-CoV-2 spike glycoprotein. J Biol Chem 2023; 299:105014. [PMID: 37414149 PMCID: PMC10416065 DOI: 10.1016/j.jbc.2023.105014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/12/2023] [Accepted: 07/01/2023] [Indexed: 07/08/2023] Open
Abstract
The target for humoral immunity, SARS-CoV-2 spike glycoprotein, has become the focus of vaccine research and development. Previous work demonstrated that the N-terminal domain (NTD) of SARS-CoV-2 spike binds biliverdin-a product of heme catabolism-causing a strong allosteric effect on the activity of a subset of neutralizing antibodies. Herein, we show that the spike glycoprotein is also able to bind heme (KD = 0.5 ± 0.2 μM). Molecular modeling indicated that the heme group fits well within the same pocket on the SARS-CoV-2 spike NTD. Lined by aromatic and hydrophobic residues (W104, V126, I129, F192, F194, I203, and L226), the pocket provides a suitable environment to stabilize the hydrophobic heme. Mutagenesis of N121 has a substantive effect on heme binding (KD = 3000 ± 220 μM), confirming the pocket as a major heme binding location of the viral glycoprotein. Coupled oxidation experiments in the presence of ascorbate indicated that the SARS-CoV-2 glycoprotein can catalyze the slow conversion of heme to biliverdin. The heme trapping and oxidation activities of the spike may allow the virus to reduce levels of free heme during infection to facilitate evasion of the adaptive and innate immunity.
Collapse
Affiliation(s)
- Samuel L Freeman
- School of Chemistry, Cantock's Close, University of Bristol, Bristol, United Kingdom
| | - A Sofia F Oliveira
- School of Chemistry, Cantock's Close, University of Bristol, Bristol, United Kingdom
| | - Andrea E Gallio
- School of Chemistry, Cantock's Close, University of Bristol, Bristol, United Kingdom
| | - Annachiara Rosa
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Maria K Simitakou
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Christopher J Arthur
- School of Chemistry, Cantock's Close, University of Bristol, Bristol, United Kingdom
| | - Adrian J Mulholland
- School of Chemistry, Cantock's Close, University of Bristol, Bristol, United Kingdom
| | - Peter Cherepanov
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, London, United Kingdom; Department of Infectious Disease, St-Mary's Campus, Imperial College London, United Kingdom.
| | - Emma L Raven
- School of Chemistry, Cantock's Close, University of Bristol, Bristol, United Kingdom.
| |
Collapse
|
13
|
Schoettler ML, Dandoy CE, Harris A, Chan M, Tarquinio KM, Jodele S, Qayed M, Watkins B, Kamat P, Petrillo T, Obordo J, Higham CS, Dvorak CC, Westbrook A, Zinter MS, Williams KM. Diffuse alveolar hemorrhage after hematopoietic cell transplantation- response to treatments and risk factors for mortality. Front Oncol 2023; 13:1232621. [PMID: 37546403 PMCID: PMC10399223 DOI: 10.3389/fonc.2023.1232621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 06/26/2023] [Indexed: 08/08/2023] Open
Abstract
Diffuse alveolar hemorrhage (DAH) is a life-threatening complication of hematopoietic cellular therapy (HCT). This study aimed to evaluate the effect of DAH treatments on outcomes using data from consecutive HCT patients clinically diagnosed with DAH from 3 institutions between January 2018-August 2022. Endpoints included sustained complete response (sCR) defined as bleeding cessation without recurrent bleeding, and non-relapse mortality (NRM). Forty children developed DAH at a median of 56.5 days post-HCT (range 1-760). Thirty-five (88%) had at least one concurrent endothelial disorder, including transplant-associated thrombotic microangiopathy (n=30), sinusoidal obstructive syndrome (n=19), or acute graft versus host disease (n=10). Fifty percent had a concurrent pulmonary infection at the time of DAH. Common treatments included steroids (n=17, 25% sCR), inhaled tranexamic acid (INH TXA,n=26, 48% sCR), and inhaled recombinant activated factor VII (INH fVIIa, n=10, 73% sCR). NRM was 56% 100 days after first pulmonary bleed and 70% at 1 year. Steroid treatment was associated with increased risk of NRM (HR 2.25 95% CI 1.07-4.71, p=0.03), while treatment with INH TXA (HR 0.43, 95% CI 0.19- 0.96, p=0.04) and INH fVIIa (HR 0.22, 95% CI 0.07-0.62, p=0.005) were associated with decreased risk of NRM. Prospective studies are warranted to validate these findings.
Collapse
Affiliation(s)
- Michelle L. Schoettler
- Division of Blood and Marrow Transplantation, Children’s Healthcare of Atlanta, Aflac Blood and Cancer Disorders Center, Emory University, Atlanta, GA, United States
| | - Christopher E. Dandoy
- Cincinnati Children’s Medical Center, Division of Bone Marrow Transplantation and Immune Deficiency, University of Cincinnati School of Medicine, Cincinnati, OH, United States
| | - Anora Harris
- Division of Blood and Marrow Transplantation, Children’s Healthcare of Atlanta, Aflac Blood and Cancer Disorders Center, Emory University, Atlanta, GA, United States
| | - Marilynn Chan
- Pediatric Pulmonary Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Keiko M. Tarquinio
- Division of Critical Care Medicine, Department of Pediatrics, Children’s Healthcare of Atlanta, Emory University, Atlanta, GA, United States
| | - Sonata Jodele
- Cincinnati Children’s Medical Center, Division of Bone Marrow Transplantation and Immune Deficiency, University of Cincinnati School of Medicine, Cincinnati, OH, United States
| | - Muna Qayed
- Division of Blood and Marrow Transplantation, Children’s Healthcare of Atlanta, Aflac Blood and Cancer Disorders Center, Emory University, Atlanta, GA, United States
| | - Benjamin Watkins
- Division of Blood and Marrow Transplantation, Children’s Healthcare of Atlanta, Aflac Blood and Cancer Disorders Center, Emory University, Atlanta, GA, United States
| | - Pradip Kamat
- Division of Critical Care Medicine, Department of Pediatrics, Children’s Healthcare of Atlanta, Emory University, Atlanta, GA, United States
| | - Toni Petrillo
- Division of Critical Care Medicine, Department of Pediatrics, Children’s Healthcare of Atlanta, Emory University, Atlanta, GA, United States
| | - Jeremy Obordo
- Division of Blood and Marrow Transplantation, Children’s Healthcare of Atlanta, Aflac Blood and Cancer Disorders Center, Emory University, Atlanta, GA, United States
| | - Christine S. Higham
- Pediatric Allergy, Immunology, and Bone Marrow Transplant Division, University of California, San Francisco, San Francisco, CA, United States
| | - Christopher C. Dvorak
- Pediatric Allergy, Immunology, and Bone Marrow Transplant Division, University of California, San Francisco, San Francisco, CA, United States
| | - Adrianna Westbrook
- Department of Pediatrics, Pediatric Biostatistics Core, Emory University, Atlanta, GA, United States
| | - Matt S. Zinter
- Pediatric Allergy, Immunology, and Bone Marrow Transplant Division, University of California, San Francisco, San Francisco, CA, United States
- Pediatric Critical Care, University of California, San Francisco, San Francisco, CA, United States
| | - Kirsten M. Williams
- Division of Blood and Marrow Transplantation, Children’s Healthcare of Atlanta, Aflac Blood and Cancer Disorders Center, Emory University, Atlanta, GA, United States
| |
Collapse
|
14
|
Chatterjee T, Lewis TL, Arora I, Gryshyna AE, Underwood L, Masjoan Juncos JX, Aggarwal S. Sex-Based Disparities in Leukocyte Migration and Activation in Response to Inhalation Lung Injury: Role of SDF-1/CXCR4 Signaling. Cells 2023; 12:1719. [PMID: 37443753 PMCID: PMC10340292 DOI: 10.3390/cells12131719] [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: 05/25/2023] [Revised: 06/21/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
The aim of the study was to determine whether sex-related differences exist in immune response to inhalation lung injury. C57BL/6 mice were exposed to Cl2 gas (500 ppm for 15, 20, or 30 min). Results showed that male mice have higher rates of mortality and lung injury than females. The binding of the chemokine ligand C-X-C motif chemokine 12 (CXCL12), also called stromal-derived-factor-1 (SDF-1), to the C-X-C chemokine receptor type 4 (CXCR4) on lung cells promotes the migration of leukocytes from circulation to lungs. Therefore, the hypothesis was that elevated SDF-1/CXCR4 signaling mediates exaggerated immune response in males. Plasma, blood leukocytes, and lung cells were collected from mice post-Cl2 exposure. Plasma levels of SDF-1 and peripheral levels of CXCR4 in lung cells were higher in male vs. female mice post-Cl2 exposure. Myeloperoxidase (MPO) and elastase activity was significantly increased in leukocytes of male mice exposed to Cl2. Lung cells were then ex vivo treated with SDF-1 (100 ng/mL) in the presence or absence of the CXCR4 inhibitor, AMD3100 (100 nM). SDF-1 significantly increased migration, MPO, and elastase activity in cells obtained from male vs. female mice post-Cl2 exposure. AMD3100 attenuated these effects, suggesting that differential SDF-1/CXCR4 signaling may be responsible for sex-based disparities in the immune response to inhalation lung injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Saurabh Aggarwal
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35205-3703, USA; (T.C.); (T.L.L.); (I.A.); (A.E.G.); (L.U.); (J.X.M.J.)
| |
Collapse
|
15
|
Lazrak A, Song W, Yu Z, Zhang S, Nellore A, Hoopes CW, Woodworth BA, Matalon S. Low molecular weight hyaluronan inhibits lung epithelial ion channels by activating the calcium-sensing receptor. Matrix Biol 2023; 116:67-84. [PMID: 36758905 PMCID: PMC10012407 DOI: 10.1016/j.matbio.2023.02.002] [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: 10/20/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023]
Abstract
Herein, we tested the hypothesis that low molecular weight hyaluronan (LMW-HA) inhibits lung epithelial ions transport in-vivo, ex-vivo, and in-vitro by activating the calcium-sensing receptor (CaSR). Twenty-four hours post intranasal instillation of 50-150 µg/ml LMW-HA to C57BL/6 mice, there was a 75% inhibition of alveolar fluid clearance (AFC), a threefold increase in the epithelial lining fluid (ELF) depth, and a 20% increase in lung wet/dry (W/D) ratio. Incubation of human and mouse precision cut lung slices with 150 µg/ml LMW-HA reduced the activity and the open probability (Po) of epithelial sodium channel (ENaC) in alveolar epithelial type 2 (ATII) cells, and in mouse tracheal epithelial cells (MTEC) monolayers as early as 4 h. The Cl- current through cystic fibrosis transmembrane conductance regulator (CFTR) and the activity of Na,K-ATPase were both inhibited by more than 66% at 24 h. The inhibitory effects of LMW-HA on ion channels were reversed by 1 µM NPS-2143, or 150 µg/ml high molecular weight hyaluronan (HMW-HA). In HEK-293 cells expressing the calcium-sensitive Cl- channel TMEM16-A, CaSR was required for the activation of the Cl- current by LMW-HA. This is the first demonstration of lung ions and water transport inhibition by LMW-HA, and its mediation through the activation of CaSR.
Collapse
Affiliation(s)
- Ahmed Lazrak
- Department of Anesthesiology and Perioperative Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Division of Molecular and Translational Biomedicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Pulmonary Injury and Repair Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Gregory Fleming James Cystic Fibrosis Research Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA.
| | - Weifeng Song
- Department of Anesthesiology and Perioperative Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Division of Molecular and Translational Biomedicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA
| | - Zhihong Yu
- Department of Anesthesiology and Perioperative Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Division of Molecular and Translational Biomedicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Pulmonary Injury and Repair Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA
| | - Shaoyan Zhang
- Department of Otolaryngology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Gregory Fleming James Cystic Fibrosis Research Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA
| | - Anoma Nellore
- Department of Medicine, Division of Infectious Diseases, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA
| | - Charles W Hoopes
- Division of Cardiothoracic Surgery, Heersink School of Medicine, University of Alabama at Birmingham, AL 35295, USA
| | - Bradford A Woodworth
- Department of Otolaryngology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Gregory Fleming James Cystic Fibrosis Research Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA
| | - Sadis Matalon
- Department of Anesthesiology and Perioperative Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Division of Molecular and Translational Biomedicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Pulmonary Injury and Repair Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA
| |
Collapse
|
16
|
Sun YF, Ma L, Li JH, Yang Y, Gong XH, Cai C. Involvement of miR-495 overexpression in the pathogenesis of bronchopulmonary dysplasia in preterm infants via the targeting of NEDD4L-ENaC pathway. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:4. [PMID: 36760249 PMCID: PMC9906200 DOI: 10.21037/atm-22-3293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 11/07/2022] [Indexed: 12/24/2022]
Abstract
Background Bronchopulmonary dysplasia (BPD) is a severe pulmonary complication causing morbidity and mortality in preterm infants. A key histopathological feature of BPD is late lung growth retardation, in which the process of alveolarization is hindered and the mechanism of which is unclear. Emerging evidence indicates that microRNAs (miRNAs) promote the development of BPD via the inhibition of their target genes. MiR-495 has been reported to be involved in various lung diseases. However, the physiological function of miR-495 in BPD has not yet been fully understood. Methods Differentially expressed miRNAs in peripheral blood of patients with BPD were compared with those of normal controls. A dual-luciferase reporter assay was performed to identify the target genes of miR-495. A BPD neonatal rat model was established by injecting lipopolysaccharide (LPS) in the amniotic sac of pregnant rats. The morphology of the lungs was observed using hematoxylin and eosin (HE) staining. The expression of miR-495, neural precursor cell expressed developmentally down-regulated 4-like (NEDD4L), and epithelial Na+ channel (ENaC) was tested using quantitative reverse transcription-polymerase chain reaction (qRT-PCR), Western blot analysis, and immunofluorescent (IF) staining. Results The expression of miR-495 was significantly increased in the peripheral blood samples of premature infants with BPD and verified using qRT-PCR. NEDD4L was proven to be the target gene of miR-495. Additionally, miR-495 expression was also increased in the lungs of rat pups with BPD at postnatal day (P) 3 compared with the control group. qRT-PCR and Western blot results showed that NEDD4L expression was decreased while ENaC expression was increased at the transcriptional and translational levels. IF staining results showed that NEDD4L level was decreased while ENaC level was increased in the LPS-induced BPD rat model, which was consistent with abnormal changes in alveolar structure. Conclusions The aberrant overexpression of miR-495 may contribute to the development of BPD by targeting NEDD4L-ENaC pathway, implying an imbalance in lung fluid clearance.
Collapse
Affiliation(s)
- Yi-Fan Sun
- Department of Neonatology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li Ma
- Department of Neonatology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jian-Hui Li
- Department of Neonatology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuan Yang
- Department of Neonatology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Hui Gong
- Department of Neonatology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Cheng Cai
- Department of Neonatology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
17
|
Jia Q, Yang Y, Chen X, Yao S, Hu Z. Emerging roles of mechanosensitive ion channels in acute lung injury/acute respiratory distress syndrome. Respir Res 2022; 23:366. [PMID: 36539808 PMCID: PMC9764320 DOI: 10.1186/s12931-022-02303-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is a devastating respiratory disorder with high rates of mortality and morbidity, but the detailed underlying mechanisms of ALI/ARDS remain largely unknown. Mechanosensitive ion channels (MSCs), including epithelial sodium channel (ENaC), Piezo channels, transient receptor potential channels (TRPs), and two-pore domain potassium ion (K2P) channels, are highly expressed in lung tissues, and the activity of these MSCs can be modulated by mechanical forces (e.g., mechanical ventilation) and other stimuli (e.g., LPS, hyperoxia). Dysfunction of MSCs has been found in various types of ALI/ARDS, and MSCs play a key role in regulating alveolar fluid clearance, alveolar epithelial/endothelial barrier function, the inflammatory response and surfactant secretion in ALI/ARDS lungs. Targeting MSCs exerts therapeutic effects in the treatment of ALI/ARDS. In this review, we summarize the structure and functions of several well-recognized MSCs, the role of MSCs in the pathogenesis of ALI/ARDS and recent advances in the pharmacological and molecular modulation of MSCs in the treatment of ALI/ARDS. According to the current literature, targeting MSCs might be a very promising therapeutic approach against ALI/ARDS.
Collapse
Affiliation(s)
- Qi Jia
- grid.33199.310000 0004 0368 7223Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiyi Yang
- grid.33199.310000 0004 0368 7223Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangdong Chen
- grid.33199.310000 0004 0368 7223Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shanglong Yao
- grid.33199.310000 0004 0368 7223Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiqiang Hu
- grid.33199.310000 0004 0368 7223Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
18
|
Liu Y, Zhou S, Xiang D, Ju L, Shen D, Wang X, Wang Y. Friend or Foe? The Roles of Antioxidants in Acute Lung Injury. Antioxidants (Basel) 2021; 10:1956. [PMID: 34943059 PMCID: PMC8750496 DOI: 10.3390/antiox10121956] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 12/18/2022] Open
Abstract
Acute lung injury (ALI) is an acute hypoxic respiratory insufficiency caused by various intra- and extra-pulmonary injury factors. The oxidative stress caused by excessive reactive oxygen species (ROS) produced in the lungs plays an important role in the pathogenesis of ALI. ROS is a "double-edged sword", which is widely involved in signal transduction and the life process of cells at a physiological concentration. However, excessive ROS can cause mitochondrial oxidative stress, leading to the occurrence of various diseases. It is well-known that antioxidants can alleviate ALI by scavenging ROS. Nevertheless, more and more studies found that antioxidants have no significant effect on severe organ injury, and may even aggravate organ injury and reduce the survival rate of patients. Our study introduces the application of antioxidants in ALI, and explore the mechanisms of antioxidants failure in various diseases including it.
Collapse
Affiliation(s)
- Yang Liu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (Y.L.); (D.S.)
| | - Shujun Zhou
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan 430071, China; (S.Z.); (D.X.)
| | - Du Xiang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan 430071, China; (S.Z.); (D.X.)
| | - Lingao Ju
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan 430071, China;
- Human Genetics Resource Preservation Center of Hubei Province, Wuhan 430071, China
| | - Dexin Shen
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (Y.L.); (D.S.)
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (Y.L.); (D.S.)
| | - Yanfeng Wang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan 430071, China; (S.Z.); (D.X.)
| |
Collapse
|
19
|
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: 30] [Impact Index Per Article: 7.5] [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
|
20
|
Wang C, Li YH, Yang ZT, Cheng NT, Tang HX, Xu M. The function and mechanism of microRNA-92a-3p in lipopolysaccharide-induced acute lung injury. Immunopharmacol Immunotoxicol 2021; 44:47-57. [PMID: 34783628 DOI: 10.1080/08923973.2021.2001497] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVES Sepsis-associated acute lung injury (ALI) is a clinically severe respiratory disorder and remains the leading cause of multiple organ failure and mortality. Herein, we used lipopolysaccharide (LPS) to generate sepsis-induced ALI and try to explore the role and mechanism of microRNA-92a-3p (miR-92a-3p) in this process. METHODS Mice were intravenously injected with miR-92a-3p agomir, antagomir and negative controls for 3 consecutive days and then were intratracheally instillated by LPS (5 mg/kg) for 12 h. To knock down the endogenous A-kinase anchoring protein 1 (AKAP1), mice were intratracheally injected with recombinant adenovirus carrying the short hairpin RNA targeting AKAP1 (shAkap1) at 1 week before LPS administration. RESULTS miR-92a-3p level was significantly upregulated in the lungs by LPS injection. miR-92a-3p antagomir reduced LPS-induced intrapulmonary inflammation and oxidative stress, thereby preventing pulmonary injury and dysfunction. In contrast, miR-92a-3p agomir aggravated LPS-induced intrapulmonary inflammation, oxidative stress, pulmonary injury and dysfunction. Moreover, we reported that AKAP1 upregulation was required for the beneficial effects of miR-92a-3p antagomir, and that AKAP1 knockdown completely abolished the anti-inflammatory and antioxidant capacities of miR-92a-3p antagomir. CONCLUSION Our data identify that miR-92a-3p modulates LPS-induced intrapulmonary inflammation, oxidative stress and ALI via AKAP1 in mice.
Collapse
Affiliation(s)
- Cong Wang
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yang-Hao Li
- Department of Thoracic Surgery, Huangmei People's Hospital, Huanggang, China
| | - Ze-Tian Yang
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ni-Tao Cheng
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - He-Xiao Tang
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ming Xu
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
21
|
Hu F, Dong X, Li W, Lv J, Lin F, Song G, Hou G, Li R. miR‑351‑5p aggravates lipopolysaccharide‑induced acute lung injury via inhibiting AMPK. Mol Med Rep 2021; 24:689. [PMID: 34328196 PMCID: PMC8365417 DOI: 10.3892/mmr.2021.12330] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 04/22/2021] [Indexed: 11/05/2022] Open
Abstract
Inflammation and oxidative stress have indispensable roles in the development of acute lung injury (ALI). MicroRNA (miRNA/miR)‑351‑5p was initially identified as a myogenesis‑associated miRNA; however, its role in lipopolysaccharide (LPS)‑induced ALI remains unclear. The aim of the present study was to investigate the role and potential mechanisms of miR‑351‑5p in ALI. ALI was induced through a single intratracheal injection of LPS for 12 h, and miR‑351‑5p agomir, antagomir or their corresponding negative controls were injected into the tail vein before LPS stimulation. Compound C, 2',5'‑dideoxyadenosine and H89 were used to inhibit AMP‑activated protein kinase (AMPK), adenylate cyclase and protein kinase A (PKA), respectively. miR‑351‑5p levels in the lungs were significantly increased in response to LPS injection. miR‑351‑5p antagomir alleviated, while miR‑351‑5p agomir aggravated LPS‑induced oxidative stress and inflammation in the lungs. The present results also demonstrated that miR‑351‑5p antagomir attenuated LPS‑induced ALI via activating AMPK, and that the cAMP/PKA axis was required for the activation of AMPK by the miR‑351‑5p antagomir. In conclusion, the present study indicated that miR‑351‑5p aggravated LPS‑induced ALI via inhibiting AMPK, suggesting that targeting miR‑351‑5p may help to develop efficient therapeutic approaches for treating ALI.
Collapse
Affiliation(s)
- Fen Hu
- Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Jiangxia District, Wuhan, Hubei 430200, P.R. China
| | - Xianfeng Dong
- Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Jiangxia District, Wuhan, Hubei 430200, P.R. China
| | - Weixin Li
- Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Jiangxia District, Wuhan, Hubei 430200, P.R. China
| | - Jianfa Lv
- Department of Thoracic Surgery, Hanchuan People's Hospital, Xiaogan, Hubei 431600, P.R. China
| | - Feng Lin
- Department of Thoracic Surgery, Macheng People's Hospital, Huanggang, Hubei 438300, P.R. China
| | - Gan Song
- Department of Thoracic Surgery, Macheng People's Hospital, Huanggang, Hubei 438300, P.R. China
| | - Guoqiang Hou
- Department of Thoracic Surgery, Yangxin People's Hospital, Huangshi, Hubei 435200, P.R. China
| | - Ruiyun Li
- Department of Pulmonary and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
22
|
Addis DR, Aggarwal S, Lazrak A, Jilling T, Matalon S. Halogen-Induced Chemical Injury to the Mammalian Cardiopulmonary Systems. Physiology (Bethesda) 2021; 36:272-291. [PMID: 34431415 DOI: 10.1152/physiol.00004.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The halogens chlorine (Cl2) and bromine (Br2) are highly reactive oxidizing elements with widespread industrial applications and a history of development and use as chemical weapons. When inhaled, depending on the dose and duration of exposure, they cause acute and chronic injury to both the lungs and systemic organs that may result in the development of chronic changes (such as fibrosis) and death from cardiopulmonary failure. A number of conditions, such as viral infections, coexposure to other toxic gases, and pregnancy increase susceptibility to halogens significantly. Herein we review their danger to public health, their mechanisms of action, and the development of pharmacological agents that when administered post-exposure decrease morbidity and mortality.
Collapse
Affiliation(s)
- Dylan R Addis
- Department of Anesthesiology and Perioperative Medicine, Division of Cardiothoracic Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama.,Comprehensive Cardiovascular Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Saurabh Aggarwal
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, University of Alabama at Birmingham, Birmingham, Alabama.,Pulmonary Injury and Repair Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ahmed Lazrak
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, University of Alabama at Birmingham, Birmingham, Alabama.,Pulmonary Injury and Repair Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Tamas Jilling
- Pulmonary Injury and Repair Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama.,Department of Pediatrics, Division of Neonatology, Children's Hospital, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sadis Matalon
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, University of Alabama at Birmingham, Birmingham, Alabama.,Pulmonary Injury and Repair Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
23
|
Rapozzi V, Juarranz A, Habib A, Ihan A, Strgar R. Is haem the real target of COVID-19? Photodiagnosis Photodyn Ther 2021; 35:102381. [PMID: 34119708 PMCID: PMC8192263 DOI: 10.1016/j.pdpdt.2021.102381] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/25/2021] [Accepted: 06/01/2021] [Indexed: 02/08/2023]
Abstract
Although a vaccination campaign has been launched in many countries, the COVID-19 pandemic is not under control. The main concern is the emergence of new variants of SARS-CoV-2; therefore, it is important to find approaches to prevent or reduce the virulence and pathogenicity of the virus. Currently, the mechanism of action of SARS-CoV-2 is not fully understood. Considering the clinical effects that occur during the disease, attacking the human respiratory and hematopoietic systems, and the changes in biochemical parameters (including decreases in haemoglobin [Hb] levels and increases in serum ferritin), it is clear that iron metabolism is involved. SARS-CoV-2 induces haemolysis and interacts with Hb molecules via ACE2, CD147, CD26, and other receptors located on erythrocytes and/or blood cell precursors that produce dysfunctional Hb. A molecular docking study has reported a potential link between the virus and the beta chain of haemoglobin and attack on haem. Considering that haem is involved in miRNA processing by binding to the DGCR8-DROSHA complex, we hypothesised that the virus may check this mechanism and thwart the antiviral response.
Collapse
Affiliation(s)
| | - Angeles Juarranz
- Department of Biology, University Autonoma of Madrid, Madrid 28049, Spain
| | - Ahsan Habib
- Department of Chemistry, University of Dhaka, Dhaka 1000, Bangladesh
| | - Alojz Ihan
- Institute for Microbiology and Immunology, Medical Faculty of Ljubljana, Slovenia
| | - Rebeka Strgar
- Institution of Applicative Biophotonics, Technological Park Ljubljana, Slovenia
| |
Collapse
|
24
|
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: 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: 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
|
25
|
Wang C, Chen Y, Cheng NT, Yang ZT, Tang HX, Xu M. MicroRNA-762 Modulates Lipopolysaccharide-induced Acute Lung Injury via SIRT7. Immunol Invest 2021; 51:1407-1422. [PMID: 34251977 DOI: 10.1080/08820139.2021.1951753] [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] [Indexed: 12/29/2022]
Abstract
Background: Inflammation and oxidative stress contribute to the pathogenesis of lipopolysaccharide (LPS)-induced acute lung injury (ALI). MicroRNA-762 (miR-762) has been implicated in the progression of inflammation and oxidative stress; however, its role in ALI remains unclear. In this study, we aim to investigate the role and underlying mechanisms of miR-762 in LPS-induced ALI. Methods: Mice were intravenously injected with miR-762 antagomir, agomir or the negative controls for 3 consecutive days and then received a single intratracheal instillation of LPS (5 mg/kg) for 12 h to establish ALI model. Adenoviral vectors were used to knock down the endogenous SIRT7 expression. Results: An increased miR-762 expression was detected in LPS-treated lungs. miR-762 antagomir significantly reduced inflammation, oxidative stress and ALI in mice, while the mice with miR-762 agomir treatment exhibited a deleterious phenotype. Besides, we found that SIRT7 upregulation was essential for the pulmonoprotective effects of miR-762 antagomir, and that SIRT7 silence completely abolished the anti-inflammatory and anti-oxidant capacities of miR-762 antagomir. Conclusion: miR-762 is implicated in the pathogenesis of LPS-induced ALI via modulating inflammation and oxidative stress, which depends on its regulation of SIRT7 expression. It might be a valuable therapeutic target for the treatment of ALI.
Collapse
Affiliation(s)
- Cong Wang
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yun Chen
- Department of Thoracic Surgery, Xishui People's Hospital Affiliated to Hubei University of Science and Technology, Huanggang, Hubei, China
| | - Ni-Tao Cheng
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ze-Tian Yang
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - He-Xiao Tang
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ming Xu
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
26
|
Cho DY, Zhang S, Lazrak A, Skinner D, Thompson HM, Grayson J, Guroji P, Aggarwal S, Bebok Z, Rowe SM, Matalon S, Sorscher EJ, Woodworth BA. LPS decreases CFTR open probability and mucociliary transport through generation of reactive oxygen species. Redox Biol 2021; 43:101998. [PMID: 33971543 PMCID: PMC8129928 DOI: 10.1016/j.redox.2021.101998] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 12/26/2022] Open
Abstract
Lipopolysaccharide (LPS) serves as the interface between gram-negative bacteria (GNB) and the innate immune response in respiratory epithelial cells (REC). Herein, we describe a novel biological role of LPS that permits GNB to persist in the respiratory tract through inducing CFTR and mucociliary dysfunction. LPS reduced cystic fibrosis transmembrane conductance regulater (CFTR)-mediated short-circuit current in mammalian REC in Ussing chambers and nearly abrogated CFTR single channel activity (defined as forskolin-activated Cl- currents) in patch clamp studies, effects of which were blocked with toll-like receptor (TLR)-4 inhibitor. Unitary conductance and single-channel amplitude of CFTR were unaffected, but open probability and number of active channels were markedly decreased. LPS increased cytoplasmic and mitochondrial reactive oxygen species resulting in CFTR carbonylation. All effects of exposure were eliminated when reduced glutathione was added in the medium along with LPS. Functional microanatomy parameters, including mucociliary transport, in human sinonasal epithelial cells in vitro were also decreased, but restored with co-incubation with glutathione or TLR-4 inhibitor. In vivo measurements, following application of LPS in the nasal cavities showed significant decreases in transepithelial Cl- secretion as measured by nasal potential difference (NPD) – an effect that was nullified with glutathione and TLR-4 inhibitor. These data provide definitive evidence that LPS-generated reactive intermediates downregulate CFTR function in vitro and in vivo which results in cystic fibrosis-type disease. Findings have implications for therapeutic approaches intent on stimulating Cl- secretion and/or reducing oxidative stress to decrease the sequelae of GNB airway colonization and infection.
Collapse
Affiliation(s)
- Do Yeon Cho
- Department of Otolaryngology Head & Neck Surgery, University of Alabama at Birmingham, Birmingham, AL, USA; Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, USA; Division of Otolaryngology, Department of Surgery, Veterans Affairs, Birmingham, AL, USA
| | - Shaoyan Zhang
- Department of Otolaryngology Head & Neck Surgery, University of Alabama at Birmingham, Birmingham, AL, USA; Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ahmed Lazrak
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Daniel Skinner
- Department of Otolaryngology Head & Neck Surgery, University of Alabama at Birmingham, Birmingham, AL, USA; Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Harrison M Thompson
- Department of Otolaryngology Head & Neck Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jessica Grayson
- Department of Otolaryngology Head & Neck Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Purushotham Guroji
- Department of Cell Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Saurabh Aggarwal
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Zsuzsanna Bebok
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Cell Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Steven M Rowe
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Cell Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sadis Matalon
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Eric J Sorscher
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Bradford A Woodworth
- Department of Otolaryngology Head & Neck Surgery, University of Alabama at Birmingham, Birmingham, AL, USA; Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
27
|
Hopp MT, Domingo-Fernández D, Gadiya Y, Detzel MS, Graf R, Schmalohr BF, Kodamullil AT, Imhof D, Hofmann-Apitius M. Linking COVID-19 and Heme-Driven Pathophysiologies: A Combined Computational-Experimental Approach. Biomolecules 2021; 11:biom11050644. [PMID: 33925394 PMCID: PMC8147026 DOI: 10.3390/biom11050644] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 01/08/2023] Open
Abstract
The SARS-CoV-2 outbreak was declared a worldwide pandemic in 2020. Infection triggers the respiratory tract disease COVID-19, which is accompanied by serious changes in clinical biomarkers such as hemoglobin and interleukins. The same parameters are altered during hemolysis, which is characterized by an increase in labile heme. We present two computational–experimental approaches aimed at analyzing a potential link between heme-related and COVID-19 pathophysiologies. Herein, we performed a detailed analysis of the common pathways induced by heme and SARS-CoV-2 by superimposition of knowledge graphs covering heme biology and COVID-19 pathophysiology. Focus was laid on inflammatory pathways and distinct biomarkers as the linking elements. In a second approach, four COVID-19-related proteins, the host cell proteins ACE2 and TMPRSS2 as well as the viral proteins 7a and S protein were computationally analyzed as potential heme-binding proteins with an experimental validation. The results contribute to the understanding of the progression of COVID-19 infections in patients with different clinical backgrounds and may allow for a more individual diagnosis and therapy in the future.
Collapse
Affiliation(s)
- Marie-Thérèse Hopp
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany; (M.-T.H.); (M.S.D.); (R.G.); (B.F.S.)
| | - Daniel Domingo-Fernández
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, D-53757 Sankt Augustin, Germany; (D.D.-F.); (Y.G.); (A.T.K.)
- Enveda Biosciences, Inc., San Francisco, CA 94080, USA
| | - Yojana Gadiya
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, D-53757 Sankt Augustin, Germany; (D.D.-F.); (Y.G.); (A.T.K.)
| | - Milena S. Detzel
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany; (M.-T.H.); (M.S.D.); (R.G.); (B.F.S.)
| | - Regina Graf
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany; (M.-T.H.); (M.S.D.); (R.G.); (B.F.S.)
| | - Benjamin F. Schmalohr
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany; (M.-T.H.); (M.S.D.); (R.G.); (B.F.S.)
| | - Alpha T. Kodamullil
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, D-53757 Sankt Augustin, Germany; (D.D.-F.); (Y.G.); (A.T.K.)
- Causality Biomodels, Kinfra Hi-Tech Park, Kalamassery, Cochin, Kerala 683503, India
| | - Diana Imhof
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany; (M.-T.H.); (M.S.D.); (R.G.); (B.F.S.)
- Correspondence: (D.I.); (M.H.-A.)
| | - Martin Hofmann-Apitius
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, D-53757 Sankt Augustin, Germany; (D.D.-F.); (Y.G.); (A.T.K.)
- Correspondence: (D.I.); (M.H.-A.)
| |
Collapse
|