1
|
Wolf A, Tabasi M, Zacharek M, Martin G, Hershenson MB, Meyerhoff ME, Sajjan U. S-Nitrosoglutathione Reduces the Density of Staphylococcus aureus Biofilms Established on Human Airway Epithelial Cells. ACS OMEGA 2023; 8:846-856. [PMID: 36643497 PMCID: PMC9835527 DOI: 10.1021/acsomega.2c06212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/20/2022] [Indexed: 05/03/2023]
Abstract
Patients with chronic rhinosinusitis (CRS) often show persistent colonization by bacteria in the form of biofilms which are resistant to antibiotic treatment. One of the most commonly isolated bacteria in CRS is Staphylococcus aureus (S. aureus). Nitric oxide (NO) is a potent antimicrobial agent and disperses biofilms efficiently. We hypothesized that S-nitrosoglutathione (GSNO), an endogenous NO carrier/donor, synergizes with gentamicin to disperse and reduce the bacterial biofilm density. We prepared GSNO formulations which are stable up to 12 months at room temperature and show the maximum amount of NO release within 1 h. We examined the effects of this GSNO formulation on the S. aureus biofilm established on the apical surface of the mucociliary-differentiated airway epithelial cell cultures regenerated from airway basal (stem) cells from cystic fibrosis (CF) and CRS patients. We demonstrate that for CF cells, which are defective in producing NO, treatment with GSNO at 100 μM increased the NO levels on the apical surface and reduced the biofilm bacterial density by 2 log units without stimulating pro-inflammatory effects or inducing epithelial cell death. In combination with gentamicin, GSNO further enhanced the killing of biofilm bacteria. Compared to placebo, GSNO significantly increased the ciliary beat frequency (CBF) in both infected and uninfected CF cell cultures. The combination of GSNO and gentamicin also reduced the bacterial density of biofilms grown on sinonasal epithelial cells from CRS patients and improved the CBF. These findings demonstrate that GSNO in combination with gentamicin may effectively reduce the density of biofilm bacteria in CRS patients. GSNO treatment may also enhance the mucociliary clearance by improving the CBF.
Collapse
Affiliation(s)
- Alex Wolf
- NOTA
Laboratories LLC, Ann Arbor, Michigan 48109, United States
| | - Mohsen Tabasi
- Department
of Microbiology Immunology and Inflammation, Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Mark Zacharek
- Deparment
of Otolaryngology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Glenn Martin
- NOTA
Laboratories LLC, Ann Arbor, Michigan 48109, United States
| | - Marc B. Hershenson
- Department
of Pediatrics, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Mark E. Meyerhoff
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Umadevi Sajjan
- Department
of Microbiology Immunology and Inflammation, Temple University, Philadelphia, Pennsylvania 19140, United States
- Center
of
Inflammation and Lung Research, Lewis Katz Medical School, Temple University, Philadelphia, Pennsylvania 19140, United States
- . Phone: (215) 707-7139
| |
Collapse
|
2
|
Getsy PM, Young AP, Bates JN, Baby SM, Seckler JM, Grossfield A, Hsieh YH, Lewis THJ, Jenkins MW, Gaston B, Lewis SJ. S-nitroso-L-cysteine stereoselectively blunts the adverse effects of morphine on breathing and arterial blood gas chemistry while promoting analgesia. Biomed Pharmacother 2022; 153:113436. [PMID: 36076552 PMCID: PMC9464305 DOI: 10.1016/j.biopha.2022.113436] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/08/2022] [Accepted: 07/15/2022] [Indexed: 01/05/2023] Open
Affiliation(s)
- Paulina M Getsy
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - Alex P Young
- Department of Pediatrics, University of Virginia, Charlottesville, VA, USA
| | - James N Bates
- Department of Anesthesia, University of Iowa, Iowa City, IA, USA
| | - Santhosh M Baby
- Galleon Pharmaceuticals, Inc., 213 Witmer Road, Horsham, PA, USA.
| | - James M Seckler
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Alan Grossfield
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Yee-Hsee Hsieh
- Division of Pulmonary, Critical Care and Sleep Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Tristan H J Lewis
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - Michael W Jenkins
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA; Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Benjamin Gaston
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Stephen J Lewis
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA; Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA; Functional Electrical Stimulation Center, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
3
|
Watchorn D, Menzies-Gow A. Investigational approaches for unmet need in severe asthma. Expert Rev Respir Med 2022; 16:661-678. [PMID: 35786146 DOI: 10.1080/17476348.2022.2096593] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/28/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Molecular antibodies (mAb) targeting inflammatory mediators are effective in T2-high asthma. The recent approval of Tezepelumab presents a novel mAb therapeutic option for those with T2-low asthma. AREAS COVERED We discuss a number of clinical problems pertinent to severe asthma that are less responsive to current therapies, such as persistent airflow obstruction and airway hyperresponsiveness. We discuss selected investigational approaches, including a number of candidate therapies under investigation in two adaptive platform trials currently in progress, with particular reference to this unmet need, as well as their potential in phenotypes such as neutrophilic asthma and obese asthma, which may or may not overlap with a T2-high phenotype. EXPERT OPINION The application of discrete targeting approaches to T2-low molecular phenotypes, including those phenotypes in which inflammation may not arise within the airway, has yielded variable results to date. Endotypes associated with T2-low asthma are likely to be diverse but await validation. Investigational therapeutic approaches must, likewise, be diverse if the goal of remission is to become attainable for all those living with asthma.
Collapse
Affiliation(s)
- David Watchorn
- Lung Division, Royal Brompton & Harefield Hospitals,London,UK
| | | |
Collapse
|
4
|
Georas SN, Wright RJ, Ivanova A, Israel E, LaVange LM, Akuthota P, Carr TF, Denlinger LC, Fajt ML, Kumar R, O'Neal WK, Phipatanakul W, Szefler SJ, Aronica MA, Bacharier LB, Burbank AJ, Castro M, Crotty Alexander L, Bamdad J, Cardet JC, Comhair SAA, Covar RA, DiMango EA, Erwin K, Erzurum SC, Fahy JV, Gaffin JM, Gaston B, Gerald LB, Hoffman EA, Holguin F, Jackson DJ, James J, Jarjour NN, Kenyon NJ, Khatri S, Kirwan JP, Kraft M, Krishnan JA, Liu AH, Liu MC, Marquis MA, Martinez F, Mey J, Moore WC, Moy JN, Ortega VE, Peden DB, Pennington E, Peters MC, Ross K, Sanchez M, Smith LJ, Sorkness RL, Wechsler ME, Wenzel SE, White SR, Zein J, Zeki AA, Noel P. The Precision Interventions for Severe and/or Exacerbation-Prone (PrecISE) Asthma Network: An overview of Network organization, procedures, and interventions. J Allergy Clin Immunol 2022; 149:488-516.e9. [PMID: 34848210 PMCID: PMC8821377 DOI: 10.1016/j.jaci.2021.10.035] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/24/2021] [Accepted: 10/07/2021] [Indexed: 12/24/2022]
Abstract
Asthma is a heterogeneous disease, with multiple underlying inflammatory pathways and structural airway abnormalities that impact disease persistence and severity. Recent progress has been made in developing targeted asthma therapeutics, especially for subjects with eosinophilic asthma. However, there is an unmet need for new approaches to treat patients with severe and exacerbation-prone asthma, who contribute disproportionately to disease burden. Extensive deep phenotyping has revealed the heterogeneous nature of severe asthma and identified distinct disease subtypes. A current challenge in the field is to translate new and emerging knowledge about different pathobiologic mechanisms in asthma into patient-specific therapies, with the ultimate goal of modifying the natural history of disease. Here, we describe the Precision Interventions for Severe and/or Exacerbation-Prone Asthma (PrecISE) Network, a groundbreaking collaborative effort of asthma researchers and biostatisticians from around the United States. The PrecISE Network was designed to conduct phase II/proof-of-concept clinical trials of precision interventions in the population with severe asthma, and is supported by the National Heart, Lung, and Blood Institute of the National Institutes of Health. Using an innovative adaptive platform trial design, the PrecISE Network will evaluate up to 6 interventions simultaneously in biomarker-defined subgroups of subjects. We review the development and organizational structure of the PrecISE Network, and choice of interventions being studied. We hope that the PrecISE Network will enhance our understanding of asthma subtypes and accelerate the development of therapeutics for severe asthma.
Collapse
Affiliation(s)
- Steve N Georas
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Rochester Medical Center, Rochester, NY.
| | | | - Anastasia Ivanova
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| | - Elliot Israel
- Department of Medicine, Divisions of Pulmonary & Critical Care Medicine & Allergy & Immunology, Brigham & Women's Hospital, Harvard Medical School, Boston, Mass
| | - Lisa M LaVange
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| | - Praveen Akuthota
- Pulmonary Division, Department of Medicine, University of California-San Diego, La Jolla, Calif
| | - Tara F Carr
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Loren C Denlinger
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Merritt L Fajt
- University of Pittsburgh Asthma Institute, University of Pittsburgh, Pittsburgh, Pa
| | | | - Wanda K O'Neal
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina, Chapel Hill, NC
| | | | - Stanley J Szefler
- Children's Hospital Colorado, Aurora, Colo; University of Colorado School of Medicine, Aurora, Colo
| | - Mark A Aronica
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | | | - Allison J Burbank
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina, Chapel Hill, NC
| | - Mario Castro
- University of Kansas School of Medicine, Kansas City, Mo
| | - Laura Crotty Alexander
- Pulmonary Division, Department of Medicine, University of California-San Diego, La Jolla, Calif
| | - Julie Bamdad
- Division of Lung Diseases, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Md
| | | | | | | | | | - Kim Erwin
- Institute for Healthcare Delivery Design, University of Illinois at Chicago, Chicago, Ill
| | | | - John V Fahy
- University of California, San Francisco School of Medicine, San Francisco, Calif
| | | | - Benjamin Gaston
- Wells Center for Pediatric Research, Indiana University, Indianapolis, Ind
| | - Lynn B Gerald
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Eric A Hoffman
- Department of Radiology, University of Iowa, Iowa City, Iowa
| | | | - Daniel J Jackson
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - John James
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| | - Nizar N Jarjour
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Nicholas J Kenyon
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis School of Medicine, Davis, Calif
| | - Sumita Khatri
- Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - John P Kirwan
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, La
| | - Monica Kraft
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Jerry A Krishnan
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Ill
| | - Andrew H Liu
- Children's Hospital Colorado, Aurora, Colo; University of Colorado School of Medicine, Aurora, Colo
| | - Mark C Liu
- Pulmonary and Critical Care Medicine, Department of Medicine, the Johns Hopkins University, Baltimore, Md
| | - M Alison Marquis
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| | - Fernando Martinez
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Jacob Mey
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, La
| | - Wendy C Moore
- Wake Forest University School of Medicine, Winston-Salem, NC
| | - James N Moy
- Rush University Medical Center, Chicago, Ill
| | - Victor E Ortega
- Wake Forest University School of Medicine, Winston-Salem, NC
| | - David B Peden
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina, Chapel Hill, NC
| | | | - Michael C Peters
- University of California, San Francisco School of Medicine, San Francisco, Calif
| | - Kristie Ross
- The Cleveland Clinic, Cleveland, Ohio; UH Rainbow Babies and Children's Hospitals, Cleveland, Ohio
| | - Maria Sanchez
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| | | | - Ronald L Sorkness
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Michael E Wechsler
- Children's Hospital Colorado, Aurora, Colo; University of Colorado School of Medicine, Aurora, Colo
| | - Sally E Wenzel
- University of Pittsburgh Asthma Institute, University of Pittsburgh, Pittsburgh, Pa
| | - Steven R White
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Ill
| | - Joe Zein
- Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - Amir A Zeki
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis School of Medicine, Davis, Calif
| | - Patricia Noel
- Division of Lung Diseases, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Md
| |
Collapse
|
5
|
Liu M, Zaman R, Sawczak V, Periasamy A, Sun F, Zaman K. S-nitrosothiols signaling in cystic fibrosis airways. J Biosci 2021. [DOI: 10.1007/s12038-021-00223-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
6
|
Pophal M, Grimmett ZW, Chu C, Margevicius S, Raffay T, Ross K, Jafri A, Giddings O, Stamler JS, Gaston B, Reynolds JD. Airway Thiol-NO Adducts as Determinants of Exhaled NO. Antioxidants (Basel) 2021; 10:antiox10101527. [PMID: 34679661 PMCID: PMC8532745 DOI: 10.3390/antiox10101527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 12/02/2022] Open
Abstract
Thiol-NO adducts such as S-nitrosoglutathione (GSNO) are endogenous bronchodilators in human airways. Decreased airway S-nitrosothiol concentrations are associated with asthma. Nitric oxide (NO), a breakdown product of GSNO, is measured in exhaled breath as a biomarker in asthma; an elevated fraction of expired NO (FENO) is associated with asthmatic airway inflammation. We hypothesized that FENO could reflect airway S-nitrosothiol concentrations. To test this hypothesis, we first studied the relationship between mixed expired NO and airway S-nitrosothiols in patients endotracheally intubated for respiratory failure. The inverse (Lineweaver-Burke type) relationship suggested that expired NO could reflect the rate of pulmonary S-nitrosothiol breakdown. We thus studied NO evolution from the lungs of mice (GSNO reductase −/−) unable reductively to catabolize GSNO. More NO was produced from GSNO in the −/− compared to wild type lungs. Finally, we formally tested the hypothesis that airway GSNO increases FENO using an inhalational challenge model in normal human subjects. FENO increased in all subjects tested, with a median t1/2 of 32.0 min. Taken together, these data demonstrate that FENO reports, at least in part, GSNO breakdown in the lungs. Unlike GSNO, NO is not present in the lungs in physiologically relevant concentrations. However, FENO following a GSNO challenge could be a non-invasive test for airway GSNO catabolism.
Collapse
Affiliation(s)
- Megan Pophal
- Institute for Transformative Molecular Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (M.P.); (Z.W.G.); (C.C.); (J.S.S.); (J.D.R.)
| | - Zachary W. Grimmett
- Institute for Transformative Molecular Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (M.P.); (Z.W.G.); (C.C.); (J.S.S.); (J.D.R.)
| | - Clara Chu
- Institute for Transformative Molecular Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (M.P.); (Z.W.G.); (C.C.); (J.S.S.); (J.D.R.)
| | - Seunghee Margevicius
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH 44106, USA;
| | - Thomas Raffay
- Division of Pediatric Pulmonology, Department of Pediatrics, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; (T.R.); (K.R.); (A.J.); (O.G.)
| | - Kristie Ross
- Division of Pediatric Pulmonology, Department of Pediatrics, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; (T.R.); (K.R.); (A.J.); (O.G.)
| | - Anjum Jafri
- Division of Pediatric Pulmonology, Department of Pediatrics, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; (T.R.); (K.R.); (A.J.); (O.G.)
| | - Olivia Giddings
- Division of Pediatric Pulmonology, Department of Pediatrics, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; (T.R.); (K.R.); (A.J.); (O.G.)
| | - Jonathan S. Stamler
- Institute for Transformative Molecular Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (M.P.); (Z.W.G.); (C.C.); (J.S.S.); (J.D.R.)
- Division of Cardiology, Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Benjamin Gaston
- Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Correspondence: ; Tel.: +1-317-274-8899
| | - James D. Reynolds
- Institute for Transformative Molecular Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (M.P.); (Z.W.G.); (C.C.); (J.S.S.); (J.D.R.)
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
- Department of Anesthesiology & Perioperative Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| |
Collapse
|
7
|
Nutritional immunity: the impact of metals on lung immune cells and the airway microbiome during chronic respiratory disease. Respir Res 2021; 22:133. [PMID: 33926483 PMCID: PMC8082489 DOI: 10.1186/s12931-021-01722-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/15/2021] [Indexed: 12/15/2022] Open
Abstract
Nutritional immunity is the sequestration of bioavailable trace metals such as iron, zinc and copper by the host to limit pathogenicity by invading microorganisms. As one of the most conserved activities of the innate immune system, limiting the availability of free trace metals by cells of the immune system serves not only to conceal these vital nutrients from invading bacteria but also operates to tightly regulate host immune cell responses and function. In the setting of chronic lung disease, the regulation of trace metals by the host is often disrupted, leading to the altered availability of these nutrients to commensal and invading opportunistic pathogenic microbes. Similarly, alterations in the uptake, secretion, turnover and redox activity of these vitally important metals has significant repercussions for immune cell function including the response to and resolution of infection. This review will discuss the intricate role of nutritional immunity in host immune cells of the lung and how changes in this fundamental process as a result of chronic lung disease may alter the airway microbiome, disease progression and the response to infection.
Collapse
|
8
|
Chang-Chien J, Huang HY, Tsai HJ, Lo CJ, Lin WC, Tseng YL, Wang SL, Ho HY, Cheng ML, Yao TC. Metabolomic differences of exhaled breath condensate among children with and without asthma. Pediatr Allergy Immunol 2021; 32:264-272. [PMID: 32920883 DOI: 10.1111/pai.13368] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 08/24/2020] [Accepted: 08/26/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND There remains an unmet need in objective tests for diagnosing asthma in children. The objective of this study was to investigate the potential of metabolomic profiles of exhaled breath condensate (EBC) to discriminate stable asthma in Asian children in the community. METHODS One hundred and sixty-five Asian children (92 stable asthma and 73 non-asthmatic controls) participating in a population-based cohort were enrolled and divided into training and validation sets. Nuclear magnetic resonance-based metabolomic profiles of EBC samples were analyzed by using orthogonal partial least squares discriminant analysis. RESULTS EBC metabolomic signature (lactate, formate, butyrate, and isobutyrate) had an area under the receiver operator characteristic curve (AUC) of 0.826 in discriminating children with and without asthma in the training set, which significantly outperformed FeNO (AUC = 0.574; P < .001) and FEV1 /FVC % predicted (AUC = 0.569; P < .001). The AUC for EBC metabolomic signature was 0.745 in the validation set, which was slightly but not significantly lower than in the testing set (P = .282). We further extrapolated two potentially involved metabolic pathways, including pyruvate (P = 1.67 × 10-3 ; impact: 0.14) and methane (P = 1.89 × 10-3 ; impact: 0.15), as the most likely divergent metabolisms between children with and without asthma. CONCLUSION This study provided evidence supporting the role of EBC metabolomic signature to discriminate stable asthma in Asian children in the community, with a discriminative property outperforming conventional clinical tests such as FeNO or spirometry.
Collapse
Affiliation(s)
- Ju Chang-Chien
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Hsin-Yi Huang
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Hui-Ju Tsai
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Chi-Jen Lo
- Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Wan-Chen Lin
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yu-Lun Tseng
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Shih-Ling Wang
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Hung-Yao Ho
- Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan.,Clinical Metabolomics Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Department of Medical Biotechnology and Laboratory Science, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Mei-Ling Cheng
- Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan.,Clinical Metabolomics Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Department of Biomedical Sciences, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Tsung-Chieh Yao
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Taoyuan, Taiwan.,Community Medicine Research Center, Chang Gung Memorial Hospital at Keelung, Keelung, Taiwan
| |
Collapse
|
9
|
Fuschillo S, Palomba L, Capparelli R, Motta A, Maniscalco M. Nitric Oxide and Hydrogen Sulfide: A Nice Pair in the Respiratory System. Curr Med Chem 2020; 27:7136-7148. [DOI: 10.2174/0929867327666200310120550] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/25/2020] [Accepted: 02/05/2020] [Indexed: 01/15/2023]
Abstract
Nitric Oxide (NO) is internationally regarded as a signal molecule involved in several
functions in the respiratory tract under physiological and pathogenic conditions. Hydrogen Sulfide
(H2S) has also recently been recognized as a new gasotransmitter with a diverse range of functions
similar to those of NO.
Depending on their respective concentrations, both these molecules act synergistically or antagonistically
as signals or damage promoters. Nevertheless, available evidence shows that the complex
biological connections between NO and H2S involve multiple pathways and depend on the site of
action in the respiratory tract, as well as on experimental conditions. This review will provide an
update on these two gasotransmitters in physiological and pathological processes.
Collapse
Affiliation(s)
- Salvatore Fuschillo
- Istituti Clinici Scientifici Maugeri IRCCS, Pulmonary Rehabilitation Division of the Telese Terme Institute, 82037 Telese Terme (BN), Italy
| | - Letizia Palomba
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino (PU), Italy
| | - Rosanna Capparelli
- Department of Agriculture, University of Naples “Federico II”, 80055 Portici, (NA), Italy
| | - Andrea Motta
- Institute of Biomolecular Chemistry, National Research Council, 80078 Pozzuoli (NA), Italy
| | - Mauro Maniscalco
- Istituti Clinici Scientifici Maugeri IRCCS, Pulmonary Rehabilitation Division of the Telese Terme Institute, 82037 Telese Terme (BN), Italy
| |
Collapse
|
10
|
Einisman HJ, Gaston B, Wijers C, Smith LA, Lewis TH, Lewis SJ, Raffay TM. Tracheomalacia in bronchopulmonary dysplasia: Trachealis hyper-relaxant responses to S-nitrosoglutathione in a hyperoxic murine model. Pediatr Pulmonol 2019; 54:1989-1996. [PMID: 31486289 PMCID: PMC7329187 DOI: 10.1002/ppul.24513] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 08/26/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) with airway hyperreactivity is a long-term pulmonary complication of prematurity. The endogenous nonadrenergic, noncholinergic signaling molecule, S-nitrosoglutathione (GSNO) and its catabolism by GSNO reductase (GSNOR) modulate airway reactivity. Tracheomalacia is a major, underinvestigated complication of BPD. We studied trachealis, left main bronchus (LB), and intrapulmonary bronchiolar (IPB) relaxant responses to GSNO in a murine hyperoxic BPD model. METHODS Wild-type (WT) or GSNOR knockout (KO) newborn mice were raised in 60% (BPD) or 21% (control) oxygen during the first 3 weeks of life. After room air recovery, adult trachealis, LB, and IPB smooth muscle relaxant responses to GSNO (after methacholine preconstriction) were studied using wire myographs. Studies were repeated after GSNOR inhibitor (GSNORi) pretreatment and in KO mice. RESULTS GSNO relaxed all airway preparations. GSNO relaxed WT BPD trachealis substantially more than WT controls (P < .05). Pharmacologic or genetic ablation of GSNOR abolished the exaggerated BPD tracheal relaxation to GSNO and also augmented BPD IPB relaxation to GSNO. LB ring contractility was not significantly different between groups or conditions. Additionally, GSNORi treatment induced relaxation of WT IPBs but not trachealis or LB. CONCLUSION GSNO dramatically relaxed the trachealis in our BPD model, an effect paradoxically reversed by loss of GSNOR. Conversely, GSNOR inhibition augmented IBP relaxation. These data suggest that GSNOR inhibition could benefit both the BPD trachealis and distal airways, restoring relaxant responses to those of room air controls. Because therapeutic options are limited in this high-risk population, future studies of GSNOR inhibition are needed.
Collapse
Affiliation(s)
- Helly J Einisman
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio.,Division of Pediatric Pulmonology, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Benjamin Gaston
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio.,Division of Pediatric Pulmonology, UH Rainbow Babies and Children's Hospital, Cleveland, Ohio
| | - Christiaan Wijers
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Laura A Smith
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Tristan H Lewis
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Stephen J Lewis
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio.,Division of Pediatric Pulmonology, UH Rainbow Babies and Children's Hospital, Cleveland, Ohio
| | - Thomas M Raffay
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio.,Division of Neonatology, UH Rainbow Babies and Children's Hospital, Cleveland, Ohio
| |
Collapse
|
11
|
Que LG, Yang Z, Lugogo NL, Katial RK, Shoemaker SA, Troha JM, Rodman DM, Tighe RM, Kraft M. Effect of the S-nitrosoglutathione reductase inhibitor N6022 on bronchial hyperreactivity in asthma. IMMUNITY INFLAMMATION AND DISEASE 2018; 6:322-331. [PMID: 29642282 PMCID: PMC5946144 DOI: 10.1002/iid3.220] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 02/20/2018] [Accepted: 02/22/2018] [Indexed: 12/20/2022]
Abstract
RATIONALE Patients with asthma demonstrate depletion of the endogenous bronchodilator GSNO and upregulation of GSNOR. OBJECTIVES An exploratory proof of concept clinical study of N6022 in mild asthma to determine the potential bronchoprotective effects of GSNOR inhibition. Mechanistic studies aimed to provide translational evidence of effect. METHODS Fourteen mild asthma patients were treated with intravenous N6022 (5 mg) or placebo and observed for 7 days, with repeated assessments of the provocative dose of methacholine causing a 20% fall in FEV1 (methacholine PC20 FEV1), followed by a washout period and crossover treatment and observation. In vitro studies in isolated eosinophils investigated the effect of GSNO and N6022 on apoptosis. MEASUREMENTS AND MAIN RESULTS This was a negative trial as it failed to reach its primary endpoint, which was change from baseline in methacholine PC20 FEV1 at 24 h. However, our exploratory analysis demonstrated significantly more two dose-doubling increases in PC20 FEV1 for N6022 compared with placebo (21% vs 6%, P < 0.05) over the 7-day observation period. Furthermore, a significant treatment effect was observed in the change in PC20 FEV1 from baseline averaged over the 7-day observation period (mean change: +0.82 mg/ml [N6022] from 1.34 mg/ml [baseline] vs -0.18 mg/ml [placebo] from 1.16 mg/ml [baseline], P = 0.023). N6022 was well tolerated in mild asthmatics. In vitro studies demonstrated enhanced eosinophilic apoptosis with N6022. CONCLUSIONS In this early phase exploratory proof of concept trial in asthma, N6022 did not significantly alter methacholine PC20 FEV1 at 24 h, but did have a treatment effect at 7 days compared to baseline. Further investigation of the efficacy of S-nitrosoglutathione reductase inhibition in a patient population with eosinophilic asthma is warranted.
Collapse
Affiliation(s)
- Loretta G Que
- Departmentof Medicine, Duke University Health System, Durham, North Carolina, USA
| | - Zhonghui Yang
- Departmentof Medicine, Duke University Health System, Durham, North Carolina, USA
| | - Njira L Lugogo
- Departmentof Medicine, Duke University Health System, Durham, North Carolina, USA
| | - Rohit K Katial
- Division of Allergy & Clinical Immunology, National Jewish Health, Denver, Colorado, USA
| | | | | | | | - Robert M Tighe
- Departmentof Medicine, Duke University Health System, Durham, North Carolina, USA
| | - Monica Kraft
- Department of Medicine, College of Medicine, University of Arizona Health Sciences, Tucson, Arizona, USA
| |
Collapse
|
12
|
Seckler JM, Meyer NM, Burton ST, Bates JN, Gaston B, Lewis SJ. Detection of trace concentrations of S-nitrosothiols by means of a capacitive sensor. PLoS One 2017; 12:e0187149. [PMID: 29073241 PMCID: PMC5658150 DOI: 10.1371/journal.pone.0187149] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 10/14/2017] [Indexed: 01/25/2023] Open
Abstract
Small molecule S-nitrosothiols are a class of endogenous chemicals in the body, which have been implicated in a variety of biological functions. However, the labile nature of NO and the limits of current detection assays have made studying these molecules difficult. Here we present a method for detecting trace concentrations of S-nitrosothiols in biological fluids. Capacitive sensors when coupled to a semiconducting material represent a method for detecting trace quantities of a chemical in complex solutions. We have taken advantage of the semiconducting and chemical properties of polydopamine to construct a capacitive sensor and associated method of use, which specifically senses S-nitrosothiols in complex biological solutions.
Collapse
Affiliation(s)
- James M. Seckler
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Nikki M. Meyer
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Spencer T. Burton
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - James N. Bates
- Department of Anesthesia, University of Iowa, Iowa City, Iowa, United States of America
| | - Benjamin Gaston
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, United States of America
- Rainbow Babies and Children’s Hospital, Cleveland, Ohio, United States of America
| | - Stephen J. Lewis
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
13
|
Abstract
The history of nitric oxide (NO) in the respiratory field dates back to the beginning of the 1990s with the pioneering study by Lars Gustafsson et al describing the presence of endogenous NO in the exhaled breath of human beings. Soon after, independent studies showed that exhaled NO concentrations (FENO) is higher in asthmatics than in normal subjects. Not all asthmatics demonstrate a high FENO, reflecting the heterogeneity of asthma. High values of FENO are associated with over-expression of corticosteroid-sensitive iNOS isoform and allergic/eosinophilic inflammation. A major feature of elevated FENO in asthma is the prediction of inhaled corticosteroid (ICS) response, and FENO more than 50 ppb in adults is a strong indicator of likely ICS sensitivity. In addition, FENO values are elevated in asthma when asthma control deteriorates, identifying patients at risk of exacerbations, and, on the other hand, FENO reductions during ICS therapy precede improvement in respiratory symptoms and lung function, suggesting that FENO is a sensitive predictor of loss of asthma control. FENO also predicts the response to biological therapy (anti-IgE, -IL-5 and -IL-13 antibodies) in severe asthma but, interestingly, FENO values fall only after treatment with anti-IL-13 and -IL-4/IL-13 receptor antibodies. The use of FENO as a Type-2 inflammatory biomarker, in constellation with other Type-2 markers, could help to determine who might benefit from ICS and biological treatment. It remains to find out more precise cut-off values of FENO to identify potential ICS responders in specific phenotypes.
Collapse
|
14
|
Raffay TM, Dylag AM, Di Fiore JM, Smith LA, Einisman HJ, Li Y, Lakner MM, Khalil AM, MacFarlane PM, Martin RJ, Gaston B. S-Nitrosoglutathione Attenuates Airway Hyperresponsiveness in Murine Bronchopulmonary Dysplasia. Mol Pharmacol 2016; 90:418-26. [PMID: 27484068 PMCID: PMC5034690 DOI: 10.1124/mol.116.104125] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 07/28/2016] [Indexed: 12/20/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is characterized by lifelong obstructive lung disease and profound, refractory bronchospasm. It is observed among survivors of premature birth who have been treated with prolonged supplemental oxygen. Therapeutic options are limited. Using a neonatal mouse model of BPD, we show that hyperoxia increases activity and expression of a mediator of endogenous bronchoconstriction, S-nitrosoglutathione (GSNO) reductase. MicroRNA-342-3p, predicted in silico and shown in this study in vitro to suppress expression of GSNO reductase, was decreased in hyperoxia-exposed pups. Both pretreatment with aerosolized GSNO and inhibition of GSNO reductase attenuated airway hyperresponsiveness in vivo among juvenile and adult mice exposed to neonatal hyperoxia. Our data suggest that neonatal hyperoxia exposure causes detrimental effects on airway hyperreactivity through microRNA-342-3p-mediated upregulation of GSNO reductase expression. Furthermore, our data demonstrate that this adverse effect can be overcome by supplementing its substrate, GSNO, or by inhibiting the enzyme itself. Rates of BPD have not improved over the past two decades; nor have new therapies been developed. GSNO-based therapies are a novel treatment of the respiratory problems that patients with BPD experience.
Collapse
Affiliation(s)
- Thomas M Raffay
- Division of Neonatology (T.M.R., A.M.D., J.M.D.F., P.M.M., R.J.M.) and Division of Pediatric Pulmonology (L.A.S., H.J.E., Y.L., B.G.), Department of Pediatrics, Rainbow Babies and Children's Hospital, and Department of Pharmacology (M.M.L.) and Department of Genetics and Genome Sciences (A.M.K.), Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Andrew M Dylag
- Division of Neonatology (T.M.R., A.M.D., J.M.D.F., P.M.M., R.J.M.) and Division of Pediatric Pulmonology (L.A.S., H.J.E., Y.L., B.G.), Department of Pediatrics, Rainbow Babies and Children's Hospital, and Department of Pharmacology (M.M.L.) and Department of Genetics and Genome Sciences (A.M.K.), Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Juliann M Di Fiore
- Division of Neonatology (T.M.R., A.M.D., J.M.D.F., P.M.M., R.J.M.) and Division of Pediatric Pulmonology (L.A.S., H.J.E., Y.L., B.G.), Department of Pediatrics, Rainbow Babies and Children's Hospital, and Department of Pharmacology (M.M.L.) and Department of Genetics and Genome Sciences (A.M.K.), Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Laura A Smith
- Division of Neonatology (T.M.R., A.M.D., J.M.D.F., P.M.M., R.J.M.) and Division of Pediatric Pulmonology (L.A.S., H.J.E., Y.L., B.G.), Department of Pediatrics, Rainbow Babies and Children's Hospital, and Department of Pharmacology (M.M.L.) and Department of Genetics and Genome Sciences (A.M.K.), Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Helly J Einisman
- Division of Neonatology (T.M.R., A.M.D., J.M.D.F., P.M.M., R.J.M.) and Division of Pediatric Pulmonology (L.A.S., H.J.E., Y.L., B.G.), Department of Pediatrics, Rainbow Babies and Children's Hospital, and Department of Pharmacology (M.M.L.) and Department of Genetics and Genome Sciences (A.M.K.), Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Yuejin Li
- Division of Neonatology (T.M.R., A.M.D., J.M.D.F., P.M.M., R.J.M.) and Division of Pediatric Pulmonology (L.A.S., H.J.E., Y.L., B.G.), Department of Pediatrics, Rainbow Babies and Children's Hospital, and Department of Pharmacology (M.M.L.) and Department of Genetics and Genome Sciences (A.M.K.), Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Mitchell M Lakner
- Division of Neonatology (T.M.R., A.M.D., J.M.D.F., P.M.M., R.J.M.) and Division of Pediatric Pulmonology (L.A.S., H.J.E., Y.L., B.G.), Department of Pediatrics, Rainbow Babies and Children's Hospital, and Department of Pharmacology (M.M.L.) and Department of Genetics and Genome Sciences (A.M.K.), Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Ahmad M Khalil
- Division of Neonatology (T.M.R., A.M.D., J.M.D.F., P.M.M., R.J.M.) and Division of Pediatric Pulmonology (L.A.S., H.J.E., Y.L., B.G.), Department of Pediatrics, Rainbow Babies and Children's Hospital, and Department of Pharmacology (M.M.L.) and Department of Genetics and Genome Sciences (A.M.K.), Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Peter M MacFarlane
- Division of Neonatology (T.M.R., A.M.D., J.M.D.F., P.M.M., R.J.M.) and Division of Pediatric Pulmonology (L.A.S., H.J.E., Y.L., B.G.), Department of Pediatrics, Rainbow Babies and Children's Hospital, and Department of Pharmacology (M.M.L.) and Department of Genetics and Genome Sciences (A.M.K.), Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Richard J Martin
- Division of Neonatology (T.M.R., A.M.D., J.M.D.F., P.M.M., R.J.M.) and Division of Pediatric Pulmonology (L.A.S., H.J.E., Y.L., B.G.), Department of Pediatrics, Rainbow Babies and Children's Hospital, and Department of Pharmacology (M.M.L.) and Department of Genetics and Genome Sciences (A.M.K.), Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Benjamin Gaston
- Division of Neonatology (T.M.R., A.M.D., J.M.D.F., P.M.M., R.J.M.) and Division of Pediatric Pulmonology (L.A.S., H.J.E., Y.L., B.G.), Department of Pediatrics, Rainbow Babies and Children's Hospital, and Department of Pharmacology (M.M.L.) and Department of Genetics and Genome Sciences (A.M.K.), Case Western Reserve University School of Medicine, Cleveland, Ohio
| |
Collapse
|
15
|
Guo C, Atochina-Vasserman E, Abramova H, George B, Manoj V, Scott P, Gow A. Role of NOS2 in pulmonary injury and repair in response to bleomycin. Free Radic Biol Med 2016; 91:293-301. [PMID: 26526764 PMCID: PMC5059840 DOI: 10.1016/j.freeradbiomed.2015.10.417] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 12/17/2022]
Abstract
Nitric oxide (NO) is derived from multiple isoforms of the Nitric Oxide Synthases (NOSs) within the lung for a variety of functions; however, NOS2-derived nitrogen oxides seem to play an important role in inflammatory regulation. In this study, we investigate the role of NOS2 in pulmonary inflammation/fibrosis in response to intratracheal bleomycin instillation (ITB) and to determine if these effects are related to macrophage phenotype. Systemic NOS2 inhibition was achieved by administration of 1400W, a specific and potent NOS2 inhibitor, via osmotic pump starting six days prior to ITB. 1400W administration attenuated lung inflammation, decreased chemotactic activity of the broncheoalveolar lavage (BAL), and reduced BAL cell count and nitrogen oxide production. S-nitrosylated SP-D (SNO-SP-D), which has a pro-inflammatory function, was formed in response to ITB; but this formation, as well as structural disruption of SP-D, was inhibited by 1400W. mRNA levels of IL-1β, CCL2 and Ptgs2 were decreased by 1400W treatment. In contrast, expression of genes associated with alternate macrophage activation and fibrosis Fizz1, TGF-β and Ym-1 was not changed by 1400W. Similar to the effects of 1400W, NOS2-/- mice displayed an attenuated inflammatory response to ITB (day 3 and day 8 post-instillation). The DNA-binding activity of NF-κB was attenuated in NOS2-/- mice; in addition, expression of alternate activation genes (Fizz1, Ym-1, Gal3, Arg1) was increased. This shift towards an increase in alternate activation was confirmed by western blot for Fizz-1 and Gal-3 that show persistent up-regulation 15 days after ITB. In contrast arginase, which is increased in expression at 8 days post ITB in NOS2-/-, resolves by day 15. These data suggest that NOS2, while critical to the development of the acute inflammatory response to injury, is also necessary to control the late phase response to ITB.
Collapse
Affiliation(s)
- Changjiang Guo
- Department of Pharmacology & Toxicology, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| | - Elena Atochina-Vasserman
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Helen Abramova
- Department of Pharmacology & Toxicology, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| | - Blessy George
- Department of Pharmacology & Toxicology, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| | - Veleeparambil Manoj
- Department of Molecular Genetics, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, USA
| | - Pamela Scott
- Department of Pharmacology & Toxicology, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| | - Andrew Gow
- Department of Pharmacology & Toxicology, Rutgers, the State University of New Jersey, Piscataway, NJ, USA.
| |
Collapse
|
16
|
Zaman K, Sawczak V, Zaidi A, Butler M, Bennett D, Getsy P, Zeinomar M, Greenberg Z, Forbes M, Rehman S, Jyothikumar V, DeRonde K, Sattar A, Smith L, Corey D, Straub A, Sun F, Palmer L, Periasamy A, Randell S, Kelley TJ, Lewis SJ, Gaston B. Augmentation of CFTR maturation by S-nitrosoglutathione reductase. Am J Physiol Lung Cell Mol Physiol 2015; 310:L263-70. [PMID: 26637637 DOI: 10.1152/ajplung.00269.2014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 11/20/2015] [Indexed: 12/27/2022] Open
Abstract
S-nitrosoglutathione (GSNO) reductase regulates novel endogenous S-nitrosothiol signaling pathways, and mice deficient in GSNO reductase are protected from airways hyperreactivity. S-nitrosothiols are present in the airway, and patients with cystic fibrosis (CF) tend to have low S-nitrosothiol levels that may be attributed to upregulation of GSNO reductase activity. The present study demonstrates that 1) GSNO reductase activity is increased in the cystic fibrosis bronchial epithelial (CFBE41o(-)) cells expressing mutant F508del-cystic fibrosis transmembrane regulator (CFTR) compared with the wild-type CFBE41o(-) cells, 2) GSNO reductase expression level is increased in the primary human bronchial epithelial cells expressing mutant F508del-CFTR compared with the wild-type cells, 3) GSNO reductase colocalizes with cochaperone Hsp70/Hsp90 organizing protein (Hop; Stip1) in human airway epithelial cells, 4) GSNO reductase knockdown with siRNA increases the expression and maturation of CFTR and decreases Stip1 expression in human airway epithelial cells, 5) increased levels of GSNO reductase cause a decrease in maturation of CFTR, and 6) a GSNO reductase inhibitor effectively reverses the effects of GSNO reductase on CFTR maturation. These studies provide a novel approach to define the subcellular location of the interactions between Stip1 and GSNO reductase and the role of S-nitrosothiols in these interactions.
Collapse
Affiliation(s)
- Khalequz Zaman
- Pediatric Pulmonology Division, Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Victoria Sawczak
- Pediatric Pulmonology Division, Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Atiya Zaidi
- Pediatric Pulmonology Division, Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Maya Butler
- Pediatric Respiratory Medicine, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Deric Bennett
- Pediatric Respiratory Medicine, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Paulina Getsy
- Pediatric Pulmonology Division, Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Maryam Zeinomar
- Pediatric Pulmonology Division, Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Zivi Greenberg
- Pediatric Respiratory Medicine, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Michael Forbes
- Pediatric Respiratory Medicine, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Shagufta Rehman
- W. M. Keck Center for Cellular Imaging, Department of Biology, University of Virginia, Charlottesville, Virginiga
| | - Vinod Jyothikumar
- W. M. Keck Center for Cellular Imaging, Department of Biology, University of Virginia, Charlottesville, Virginiga
| | - Kim DeRonde
- Pediatric Respiratory Medicine, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Abdus Sattar
- Department of Epidemiology and Biostatistics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Laura Smith
- Pediatric Pulmonology Division, Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Deborah Corey
- Pediatric Pulmonology Division, Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Adam Straub
- Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Fei Sun
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| | - Lisa Palmer
- Pediatric Respiratory Medicine, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Ammasi Periasamy
- W. M. Keck Center for Cellular Imaging, Department of Biology, University of Virginia, Charlottesville, Virginiga
| | - Scott Randell
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Thomas J Kelley
- Pediatric Pulmonology Division, Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Stephen J Lewis
- Pediatric Pulmonology Division, Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Benjamin Gaston
- Pediatric Pulmonology Division, Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio; Pediatric Pulmonology Division, Rainbow Babies and Children's Hospital, Cleveland, Ohio
| |
Collapse
|
17
|
Abstract
The versatile chemistry of nitrogen is important to pulmonary physiology. Indeed, almost all redox forms of nitrogen are relevant to pulmonary physiology and to pathophysiology. Here we review the relevance to pulmonary biology of (a) elemental nitrogen; (b) reduced forms of nitrogen such as amines, ammonia, and hydroxylamine; and (c) oxidized forms of nitrogen such as the nitroxyl anion, the nitric oxide free radical, and S-nitrosothiols. Our focus is on oxidized nitrogen in the form of S-nitrosothiol bond-containing species, which are now appreciated to be important to every type of cell-signaling process in the lung. We also review potential clinical applications of nitrogen oxide biochemistry. These principles are being translated into clinical practice as diagnostic techniques and therapies for a range of pulmonary diseases including asthma, cystic fibrosis, adult respiratory distress syndrome, primary ciliary dyskinesia, and pulmonary hypertension.
Collapse
Affiliation(s)
- Nadzeya V Marozkina
- Department of Pediatrics, Rainbow Babies and Children's Hospital and Case Western Reserve University, Cleveland, Ohio 44106; ,
| | | |
Collapse
|
18
|
Marozkina NV, Wang XQ, Stsiapura V, Fitzpatrick A, Carraro S, Hawkins GA, Bleecker E, Meyers D, Jarjour N, Fain SB, Wenzel S, Busse W, Castro M, Panettieri RA, Moore W, Lewis SJ, Palmer LA, Altes T, de Lange EE, Erzurum S, Teague WG, Gaston B. Phenotype of asthmatics with increased airway S-nitrosoglutathione reductase activity. Eur Respir J 2015; 45:87-97. [PMID: 25359343 PMCID: PMC4283933 DOI: 10.1183/09031936.00042414] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
S-Nitrosoglutathione is an endogenous airway smooth muscle relaxant. Increased airway S-nitrosoglutathione breakdown occurs in some asthma patients. We asked whether patients with increased airway catabolism of this molecule had clinical features that distinguished them from other asthma patients. We measured S-nitrosoglutathione reductase expression and activity in bronchoscopy samples taken from 66 subjects in the Severe Asthma Research Program. We also analysed phenotype and genotype data taken from the program as a whole. Airway S-nitrosoglutathione reductase activity was increased in asthma patients (p=0.032). However, only a subpopulation was affected and this subpopulation was not defined by a "severe asthma" diagnosis. Subjects with increased activity were younger, had higher IgE and an earlier onset of symptoms. Consistent with a link between S-nitrosoglutathione biochemistry and atopy: 1) interleukin 13 increased S-nitrosoglutathione reductase expression and 2) subjects with an S-nitrosoglutathione reductase single nucleotide polymorphism previously associated with asthma had higher IgE than those without this single nucleotide polymorphism. Expression was higher in airway epithelium than in smooth muscle and was increased in regions of the asthmatic lung with decreased airflow. An early-onset, allergic phenotype characterises the asthma population with increased S-nitrosoglutathione reductase activity.
Collapse
Affiliation(s)
- Nadzeya V Marozkina
- Dept of Paediatrics, Rainbow Babies and Children's Hospital, Case Western Reserve University, Cleveland, OH, USA
| | - Xin-Qun Wang
- Dept of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - Vitali Stsiapura
- Dept of Chemistry, University of Virginia, Charlottesville, VA, USA
| | | | | | | | - Eugene Bleecker
- Dept of Medicine, Wake Forest University, Winston-Salem, NC, USA
| | - Deborah Meyers
- Dept of Medicine, Wake Forest University, Winston-Salem, NC, USA
| | - Nizar Jarjour
- Dept of Medicine, University of Wisconsin, Madison, WI, USA
| | - Sean B Fain
- Dept of Medical Physics, University of Wisconsin, Madison, WI, USA
| | | | - William Busse
- Dept of Medicine, University of Wisconsin, Madison, WI, USA
| | - Mario Castro
- Dept of Medicine, Washington University, St. Louis, MO, USA
| | - Reynold A Panettieri
- Pulmonary, Allergy and Critical Care Division, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Wendy Moore
- Dept of Medicine, Wake Forest University, Winston-Salem, NC, USA
| | - Stephen J Lewis
- Dept of Paediatrics, Rainbow Babies and Children's Hospital, Case Western Reserve University, Cleveland, OH, USA
| | - Lisa A Palmer
- Dept of Paediatrics, University of Virginia, Charlottesville, VA, USA
| | - Talissa Altes
- Dept of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | - Eduard E de Lange
- Dept of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | - Serpil Erzurum
- Dept of Pathobiology, Cleveland Clinic, Cleveland, OH, USA Dept of Pulmonary, Allergy, and Critical Care Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - W Gerald Teague
- Dept of Paediatrics, University of Virginia, Charlottesville, VA, USA
| | - Benjamin Gaston
- Dept of Paediatrics, Rainbow Babies and Children's Hospital, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
19
|
Mechanisms and targets of the modulatory action of S-nitrosoglutathione (GSNO) on inflammatory cytokines expression. Arch Biochem Biophys 2014; 562:80-91. [PMID: 25135357 DOI: 10.1016/j.abb.2014.08.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Revised: 08/04/2014] [Accepted: 08/05/2014] [Indexed: 02/07/2023]
Abstract
A number of experimental studies has documented that S-nitrosoglutathione (GSNO), the main endogenous low-molecular-weight S-nitrosothiol, can exert modulatory effects on inflammatory processes, thus supporting its potential employment in medicine for the treatment of important disease conditions. At molecular level, GSNO effects have been shown to modulate the activity of a series of transcription factors (notably NF-κB, AP-1, CREB and others) as well as other components of signal transduction chains (e.g. IKK-β, caspase 1, calpain and others), resulting in the modulation of several cytokines and chemokines expression (TNFα, IL-1β, IFN-γ, IL-4, IL-8, RANTES, MCP-1 and others). Results reported to date are however not univocal, and a single main mechanism of action for the observed anti-inflammatory effects of GSNO has not been identified. Conflicting observations can be explained by differences among the various cell types studies as to the relative abundance of enzymes in charge of GSNO metabolism (GSNO reductase, γ-glutamyltransferase, protein disulfide isomerase and others), as well as by variables associated with the individual experimental models employed. Altogether, anti-inflammatory properties of GSNO seem however to prevail, and exploration of the therapeutic potential of GSNO and analogues appears therefore warranted.
Collapse
|
20
|
Blonder JP, Mutka SC, Sun X, Qiu J, Green LH, Mehra NK, Boyanapalli R, Suniga M, Look K, Delany C, Richards JP, Looker D, Scoggin C, Rosenthal GJ. Pharmacologic inhibition of S-nitrosoglutathione reductase protects against experimental asthma in BALB/c mice through attenuation of both bronchoconstriction and inflammation. BMC Pulm Med 2014; 14:3. [PMID: 24405692 PMCID: PMC3893392 DOI: 10.1186/1471-2466-14-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Accepted: 01/03/2014] [Indexed: 12/16/2022] Open
Abstract
Background S-nitrosoglutathione (GSNO) serves as a reservoir for nitric oxide (NO) and thus is a key homeostatic regulator of airway smooth muscle tone and inflammation. Decreased levels of GSNO in the lungs of asthmatics have been attributed to increased GSNO catabolism via GSNO reductase (GSNOR) leading to loss of GSNO- and NO- mediated bronchodilatory and anti-inflammatory actions. GSNOR inhibition with the novel small molecule, N6022, was explored as a therapeutic approach in an experimental model of asthma. Methods Female BALB/c mice were sensitized and subsequently challenged with ovalbumin (OVA). Efficacy was determined by measuring both airway hyper-responsiveness (AHR) upon methacholine (MCh) challenge using whole body plethysmography and pulmonary eosinophilia by quantifying the numbers of these cells in the bronchoalveolar lavage fluid (BALF). Several other potential biomarkers of GSNOR inhibition were measured including levels of nitrite, cyclic guanosine monophosphate (cGMP), and inflammatory cytokines, as well as DNA binding activity of nuclear factor kappa B (NFκB). The dose response, onset of action, and duration of action of a single intravenous dose of N6022 given from 30 min to 48 h prior to MCh challenge were determined and compared to effects in mice not sensitized to OVA. The direct effect of N6022 on airway smooth muscle tone also was assessed in isolated rat tracheal rings. Results N6022 attenuated AHR (ED50 of 0.015 ± 0.002 mg/kg; Mean ± SEM) and eosinophilia. Effects were observed from 30 min to 48 h after treatment and were comparable to those achieved with three inhaled doses of ipratropium plus albuterol used as the positive control. N6022 increased BALF nitrite and plasma cGMP, while restoring BALF and plasma inflammatory markers toward baseline values. N6022 treatment also attenuated the OVA-induced increase in NFκB activation. In rat tracheal rings, N6022 decreased contractile responses to MCh. Conclusions The significant bronchodilatory and anti-inflammatory actions of N6022 in the airways are consistent with restoration of GSNO levels through GSNOR inhibition. GSNOR inhibition may offer a therapeutic approach for the treatment of asthma and other inflammatory lung diseases. N6022 is currently being evaluated in clinical trials for the treatment of inflammatory lung disease.
Collapse
Affiliation(s)
- Joan P Blonder
- N30 Pharmaceuticals, Inc, 3122 Sterling Circle, Suite 200, Boulder, CO 80301, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Kubáň P, Foret F. Exhaled breath condensate: Determination of non-volatile compounds and their potential for clinical diagnosis and monitoring. A review. Anal Chim Acta 2013; 805:1-18. [DOI: 10.1016/j.aca.2013.07.049] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 07/16/2013] [Accepted: 07/20/2013] [Indexed: 12/31/2022]
|
22
|
Increased susceptibility to Klebsiella pneumonia and mortality in GSNOR-deficient mice. Biochem Biophys Res Commun 2013; 442:122-6. [PMID: 24239886 DOI: 10.1016/j.bbrc.2013.11.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 11/06/2013] [Indexed: 12/16/2022]
Abstract
S-nitrosoglutathione reductase (GSNOR) is a key denitrosylase and critically important for protecting immune and other cells from nitrosative stress. Pharmacological inhibition of GSNOR is being actively pursued as a therapeutic approach to increase S-nitrosoglutathione levels for the treatment of asthma and cystic fibrosis. In the present study, we employed GSNOR-deficient (GSNOR(-/-)) mice to investigate whether inactivation of GSNOR may increase susceptibility to pulmonary infection by Klebsiella pneumoniae, a common cause of nosocomial pneumonia. We found that compared to wild-type mice, bacterial colony forming units 48 h after intranasal infection with K. pneumoniae were increased over 4-folds in lung and spleen and strikingly, over a 1000-folds in blood of GSNOR(-/-) mice. Lung injury was comparable between infected wild-type and GSNOR(-/-) mice, but inflammation and injury was significantly elevated in spleen of GSNOR(-/-) mice. Whereas all wild-type mice survived 48 h after infection, 10 of 23 GSNOR(-/-) mice died. Thus, GSNOR appears to play a crucial role in controlling pulmonary and systemic infection by K. pneumoniae. Our results suggest that patients treated in clinical trials with inhibitors of GSNOR should be carefully monitored for signs of infection.
Collapse
|
23
|
Zuo L, Koozechian MS, Chen LL. Characterization of reactive nitrogen species in allergic asthma. Ann Allergy Asthma Immunol 2013; 112:18-22. [PMID: 24331388 DOI: 10.1016/j.anai.2013.10.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 10/07/2013] [Accepted: 10/10/2013] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To investigate the molecular mechanism of reactive nitrogen species (RNS) in the pathogenesis of asthma and examine the use of fractional exhaled nitric oxide (FENO) measurements in close conjunction with standard clinical assessments of asthma. DATA SOURCES Through PubMed, Google Scholar, and Medline databases, a broad medical literature review was performed in the following areas of asthma pathobiology and management: allergic asthma, RNS, nitric oxide (NO), airway inflammation, and FENO. STUDY SELECTIONS Studies were selected based on the physiologic and pathophysiologic roles of RNS in relation to allergic asthma. Current evaluations on clinical applications of FENO in asthma treatment also were selected. RESULTS At the onset of an asthma attack, an enhanced production of NO strongly correlates with increase inducible NO synthase (NOS) activity, whereas endothelial NOS and neuronal NOS regulate primarily normal metabolic functions in the central and peripheral airways. During allergic inflammatory responses, NO and superoxide form peroxynitrite, which has deleterious effects in the respiratory tract. RNS directly accentuates airway inflammation and cytotoxicity through nitrosative stress. Moreover, the use of FENO to monitor eosinophilic-mediated airway inflammation is a potentially valuable assessment that supplements standard procedures to monitor the progression of asthma. CONCLUSION This review examines recent evidence implicating the molecular mechanisms of NO and NO-derived RNS in the pathobiology of asthma and suggests that monitoring FENO may markedly contribute to asthma diagnosis.
Collapse
Affiliation(s)
- Li Zuo
- Molecular Physiology and Rehabilitation Research Laboratory, Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio.
| | - Majid S Koozechian
- Exercise and Sport Nutrition Laboratory, Department of Health and Kinesiology, Texas A&M University, College Station, Texas
| | - Lauren L Chen
- Molecular Physiology and Rehabilitation Research Laboratory, Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| |
Collapse
|
24
|
S-nitrosoglutathione reductase inhibition regulates allergen-induced lung inflammation and airway hyperreactivity. PLoS One 2013; 8:e70351. [PMID: 23936192 PMCID: PMC3723687 DOI: 10.1371/journal.pone.0070351] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 06/17/2013] [Indexed: 11/19/2022] Open
Abstract
Allergic asthma is characterized by Th2 type inflammation, leading to airway hyperresponsivenes, mucus hypersecretion and tissue remodeling. S-Nitrosoglutathione reductase (GSNOR) is an alcohol dehydrogenase involved in the regulation of intracellular levels of S-nitrosothiols. GSNOR activity has been shown to be elevated in human asthmatic lungs, resulting in diminished S-nitrosothiols and thus contributing to increased airway hyperreactivity. Using a mouse model of allergic airway inflammation, we report that intranasal administration of a new selective inhibitor of GSNOR, SPL-334, caused a marked reduction in airway hyperreactivity, allergen-specific T cells and eosinophil accumulation, and mucus production in the lungs in response to allergen inhalation. Moreover, SPL-334 treatment resulted in a significant decrease in the production of the Th2 cytokines IL-5 and IL-13 and the level of the chemokine CCL11 (eotaxin-1) in the airways. Collectively, these observations reveal that GSNOR inhibitors are effective not only in reducing airway hyperresponsiveness but also in limiting lung inflammatory responses mediated by CD4(+) Th2 cells. These findings suggest that the inhibition of GSNOR may provide a novel therapeutic approach for the treatment of allergic airway inflammation.
Collapse
|
25
|
Misso NLA, Thompson PJ. Oxidative stress and antioxidant deficiencies in asthma: potential modification by diet. Redox Rep 2013; 10:247-55. [PMID: 16354413 DOI: 10.1179/135100005x70233] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The lungs of asthmatic patients are exposed to oxidative stress due to the generation of reactive oxygen and nitrogen species as a consequence of chronic airway inflammation. Increased concentrations of NO*, H2O2 and 8-isoprostane have been measured in exhaled breath and induced sputum of asthmatic patients. O2*-, NO*, and halides interact to form highly reactive species such as peroxynitrite and HOBr, which in turn cause nitration and bromination of protein tyrosine residues. Oxidative stress may also reduce glutathione levels and cause inactivation of antioxidant enzymes such as superoxide dismutase, with a consequent increase in apoptosis, shedding of airway epithelial cells and airway remodelling. The oxidant/antioxidant equilibrium in asthmatic patients may be further perturbed by low dietary intakes of the antioxidant vitamins C and E, selenium and flavonoids, with a consequent lowering of the concentrations of these and other non-dietary antioxidants such as bilirubin and albumin in plasma and airway epithelial lining fluid. Although supplementation with vitamins C and E appears to offer protection against the adverse effects of ozone, recent randomised, placebo-controlled trials of vitamin C or E supplements for patients with mild asthma have not shown significant benefits over standard therapy. However, genetic variation in glutathione S-transferase may influence the susceptibility of asthmatic individuals to oxidative stress and the extent to which they are likely to benefit from antioxidant supplementation. Long-term prospective trials are required to determine whether modification of dietary intake will benefit asthma patients and reduce the socio-economic burden of asthma in the community.
Collapse
Affiliation(s)
- Neil L A Misso
- Asthma & Allergy Research Institute (Inc) and Centre for Asthma, Allergy & Respiratory Research, The University of Western Australia, Perth, Australia.
| | | |
Collapse
|
26
|
Anti-inflammatory effect of arginase inhibitor and corticosteroid on airway allergic reactions in a Dermatophogoides farinae-induced NC/Nga mouse model. Inflammation 2013; 36:141-51. [PMID: 22915279 DOI: 10.1007/s10753-012-9529-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The present study was aimed to investigate the effect of an arginase inhibitor, N-hydroxy-nor-L-arginine (nor-NOHA) and a corticosteroid, prednisolone, in an intranasal mite-induced NC/Nga mouse model of asthma. The treatment with nor-NOHA and prednisolone inhibited the increase in airway hyperresponsiveness, the number of bronchoalveolar lavage fluid cells, protein expression of arginase I and arginase II, messenger RNA (mRNA) expression of nitric oxide synthase (NOS)2 and Th2 cytokines such as interleukin (IL)-4, IL-5, and IL-13, and the pathological inflammatory changes of the lung. NOx levels in the lung were not changed in mice treated with prednisolone and elevated in mice treated with nor-NOHA or prednisolone plus nor-NOHA despite suppressed NOS2 mRNA expression. The study concluded that anti-inflammatory effect by nor-NOHA might be dependent on NO supply from depleted NO by downregulated arginine availability of arginase and was not related with the anti-inflammatory mechanisms by prednisolone.
Collapse
|
27
|
Greenwald R, Johnson BA, Hoskins A, Dworski R. Exhaled breath condensate formate after inhaled allergen provocation in atopic asthmatics in vivo. J Asthma 2013; 50:619-22. [PMID: 23557458 DOI: 10.3109/02770903.2013.783065] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE The dual actions of S-nitrosoglutathione reductase comprise reduction of S-nitrosoglutathione, a potent endogenous airway smooth muscle relaxant that is depleted in asthmatics, and detoxification of formaldehyde to formate. Airway formate production is increased in children with asthma, suggesting increased activity of S-nitrosoglutathione reductase. We determined formate in exhaled breath condensate from adult atopic asthmatics with asthma exacerbation produced by inhaled allergen in vivo, METHODS Twenty-two adult atopic asthmatics underwent inhaled allergen challenge using specific allergen. Exhaled breath condensate was collected at baseline, 1 h after inhalation of the provocative dose of allergen, and then every 2 h for 8 h during the challenge. Formate was analyzed by ion chromatography, RESULTS Eleven asthmatics developed an isolated early airway response, and another 11 volunteers early response followed by late airway response (dual response). Formate concentrations doubled 1 h post-challenge in asthmatics with dual-airway response but essentially unchanged in patients with an isolated early reaction, CONCLUSIONS Dual-airway response to allergen in atopic asthmatics could be associated with increased activity of S-nitrosoglutathione reductase as suggested by greater concentrations of formate in exhaled breath condensate. Measurement of formate in exhaled breath condensate could serve as a noninvasive biomarker of S-nitrosoglutathione reductase activity in vivo. Our results need to be confirmed in a larger group of asthmatics.
Collapse
Affiliation(s)
- Roby Greenwald
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | | | | | | |
Collapse
|
28
|
Broniowska KA, Diers AR, Hogg N. S-nitrosoglutathione. Biochim Biophys Acta Gen Subj 2013; 1830:3173-81. [PMID: 23416062 DOI: 10.1016/j.bbagen.2013.02.004] [Citation(s) in RCA: 263] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Revised: 02/01/2013] [Accepted: 02/07/2013] [Indexed: 11/16/2022]
Abstract
BACKGROUND S-Nitrosoglutathione (GSNO) is the S-nitrosated derivative of glutathione and is thought to be a critical mediator of the down stream signaling effects of nitric oxide (NO). GSNO has also been implicated as a contributor to various disease states. SCOPE OF REVIEW This review focuses on the chemical nature of GSNO, its biological activities, the evidence that it is an endogenous mediator of NO action, and implications for therapeutic use. MAJOR CONCLUSIONS GSNO clearly exerts its cellular actions through both NO- and S-nitrosation-dependent mechanisms; however, the chemical and biological aspects of this compound should be placed in the context of S-nitrosation as a whole. GENERAL SIGNIFICANCE GSNO is a central intermediate in formation and degradation of cellular S-nitrosothiols with potential therapeutic applications; thus, it remains an important molecule of study. This article is part of a Special Issue entitled Cellular functions of glutathione.
Collapse
|
29
|
Modulation of Asthma Pathogenesis by Nitric Oxide Pathways and Therapeutic Opportunities. ACTA ACUST UNITED AC 2012; 9:e89-e94. [PMID: 23976894 DOI: 10.1016/j.ddmec.2012.10.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Asthma, a chronic airway inflammatory disease is typically associated with high levels of exhaled nitric oxide (NO). Over the past decades, extensive research has revealed that NO participates in a number of metabolic pathways that contribute to animal models of asthma and human asthma. In asthmatic airway, high levels of NO lead to greater formation of reactive nitrogen species (RNS), which modify proteins adversely affecting functional activities. In contrast, high levels of NO are associated with lower than normal levels of S-nitrosothiols, which serve a bronchodilator function in the airway. Detailed mechanistic studies have enabled the development of compounds that target NO metabolic pathways, and provide opportunities for novel asthma therapy. This review discusses the role of NO in asthma with the primary focus on therapeutic opportunities developed in recent years.
Collapse
|
30
|
Isaac J, Tarapore P, Zhang X, Lam YW, Ho SM. Site-specific S-nitrosylation of integrin α6 increases the extent of prostate cancer cell migration by enhancing integrin β1 association and weakening adherence to laminin-1. Biochemistry 2012; 51:9689-97. [PMID: 23106339 DOI: 10.1021/bi3012324] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The increased mortality in prostate cancer is usually the result of metastatic progression of the disease from the organ-confined location. Among the major events in this progression cascade are enhanced cell migration and loss of adhesion. Moreover, elevated levels of nitric oxide (NO) and inducible nitric oxide synthase (iNOS) found within the tumor microenvironment are hallmarks of progression of this cancer. To understand the role of nitrosative stress in prostate cancer progression, we investigated the effects of NO and iNOS on prostate cancer cell migration and adhesion. Our results indicate that ectopic expression of iNOS in prostate cancer cells increased the extent of cell migration, which could be blocked by selective ITGα6 blocking antibody or iNOS inhibitors. Furthermore, iNOS was found to cause S-nitrosylation of ITGα6 at Cys86 in prostate cancer cells. By comparing the activities of wild-type ITGα6 and a Cys86 mutant, we showed that treatment of prostate cancer cells with NO increased the level of ITGα6 heterodimerization with ITGβ1 but not with ITGβ4. Finally, S-nitrosylation of ITGα6 weakened its binding to laminin-β1 and weakened the adhesion of prostate cancer cells to laminin-1. In conclusion, S-nitrosylation of ITGα6 increased the extent of prostate cancer cell migration, which could be a potential mechanism of NO- and iNOS-induced enhancement of prostate cancer metastasis.
Collapse
Affiliation(s)
- Jared Isaac
- Cancer and Cell Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | | | | | | | | |
Collapse
|
31
|
Madrasi K, Joshi MS, Gadkari T, Kavallieratos K, Tsoukias NM. Glutathiyl radical as an intermediate in glutathione nitrosation. Free Radic Biol Med 2012; 53:1968-76. [PMID: 22951977 PMCID: PMC3494776 DOI: 10.1016/j.freeradbiomed.2012.08.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2012] [Revised: 06/26/2012] [Accepted: 08/09/2012] [Indexed: 11/30/2022]
Abstract
Nitrosation of thiols is thought to be mediated by dinitrogen trioxide (N(2)O(3)) or by nitrogen dioxide radical (()NO(2)). A kinetic study of glutathione (GSH) nitrosation by NO donors in aerated buffered solutions was undertaken. S-nitrosoglutathione (GSNO) formation was assessed spectrophotometrically and by chemiluminescence. The results suggest an increase in the rate of GSNO formation with an increase in GSH with a half-maximum constant EC(50) that depends on NO concentration. Our observed increase in EC(50) with NO concentration suggests a significant contribution of ()NO(2)-mediated nitrosation with the glutathiyl radical as an intermediate in the production of GSNO.
Collapse
Affiliation(s)
- Kumpal Madrasi
- Department of Biomedical Engineering, Florida International University, Miami, FL 33174
| | - Mahesh S. Joshi
- Department of Biomedical Engineering, Florida International University, Miami, FL 33174
- Correspondence to: Mahesh S. Joshi, Ph.D. Department of Biomedical Engineering, 10555 W. Flagler Street, Florida International University, Miami, FL 33174. Tel: 305-348-7292. Fax: 305-348-6954.
| | - Tushar Gadkari
- Department of Biomedical Engineering, Florida International University, Miami, FL 33174
| | | | - Nikolaos M. Tsoukias
- Department of Biomedical Engineering, Florida International University, Miami, FL 33174
| |
Collapse
|
32
|
Marshall HE, Gow A. Regulation of cellular processes by S-nitrosylation. Preface. Biochim Biophys Acta Gen Subj 2012; 1820:673-4. [PMID: 22554496 DOI: 10.1016/j.bbagen.2012.04.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Harvey E Marshall
- Division of Pulmonary, Allergy and Critical Care Medicine Duke University Medical Center Durham, North Carolina, USA.
| | | |
Collapse
|
33
|
Raffay TM, Martin RJ, Reynolds JD. Can nitric oxide-based therapy prevent bronchopulmonary dysplasia? Clin Perinatol 2012; 39:613-38. [PMID: 22954273 PMCID: PMC3437658 DOI: 10.1016/j.clp.2012.06.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
A growing understanding of endogenous nitric oxide (NO) biology is helping to explain how and when exogenous NO may confer benefit or harm; this knowledge is also helping to identify new better-targeted NO-based therapies. In this review, results of the bronchopulmonary dysplasia clinical trials that used inhaled NO in the preterm population are placed in context, the biologic basis for novel NO therapeutics is considered, and possible future directions for NO-focused clinical and basic research in developmental lung disease are identified.
Collapse
Affiliation(s)
- Thomas M. Raffay
- Division of Neonatology, Department of Pediatrics Rainbow Babies & Children’s Hospital, Case Medical Center/University Hospitals, Cleveland, Ohio
| | - Richard J. Martin
- Division of Neonatology, Department of Pediatrics Rainbow Babies & Children’s Hospital, Case Medical Center/University Hospitals, Cleveland, Ohio
| | - James D. Reynolds
- Department of Anesthesia and Perioperative Medicine, Case Medical Center/University Hospitals, Cleveland, Ohio
,Institute for Transformative Molecular Medicine, Case Medical Center/University Hospitals, Cleveland, Ohio
| |
Collapse
|
34
|
Abstract
Diagnosis and treatment of asthma are currently based on assessment of patient symptoms and physiologic tests of airway reactivity. Research over the past decade has identified an array of biochemical and cellular biomarkers, which reflect the heterogeneous and multiple mechanistic pathways that may lead to asthma. These mechanistic biomarkers offer hope for optimal design of therapies targeting the specific pathways that lead to inflammation. This article provides an overview of blood, urine, and airway biomarkers; summarizes the pathologic pathways that they signify; and begins to describe the utility of biomarkers in the future care of patients with asthma.
Collapse
Affiliation(s)
- Serpil C. Erzurum
- Professor and Chair, Department of Pathobiology, Lerner Research Institute, and the Respiratory Institute, Cleveland Clinic, Cleveland Clinic, Cleveland, USA
| | - Benjamin M. Gaston
- Professor, Department of Pediatric Pulmonary Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
35
|
Roomans GM. Pharmacological Approaches to Correcting the Ion Transport Defect in Cystic Fibrosis. ACTA ACUST UNITED AC 2012; 2:413-31. [PMID: 14719993 DOI: 10.1007/bf03256668] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Cystic fibrosis (CF) is a lethal genetic disease caused by a mutation in a membrane protein, the cystic fibrosis transmembrane conductance regulator (CFTR), which mainly (but not exclusively) functions as a chloride channel. The main clinical symptoms are chronic obstructive lung disease, which is responsible for most of the morbidity and mortality associated with CF, and pancreatic insufficiency. About 1000 mutations of the gene coding for CFTR are currently known; the most common of these, present in the great majority of the patients (Delta508) results in the deletion of a phenylalanine at position 508. In this mutation, the aberrant CFTR is not transported to the membrane but degraded in the ubiquitin-proteasome pathway. The aim of this review is to give an overview of the pharmacologic strategies currently used in attempts to overcome the ion transport defect in CF. One strategy to develop pharmacologic treatment for CF is to inhibit the breakdown of DeltaF508-CFTR by interfering with the chaperones involved in the folding of CFTR. At least in in vitro systems, this can be accomplished by sodium phenylbutyrate, or S-nitrosoglutathione (GSNO), and also by genistein or benzo[c]quinolizinium compounds. It is also possible to stimulate CFTR or its mutated forms, when present in the plasma membrane, using xanthines, genistein, and various other compounds, such as benzamidizoles and benzoxazoles, benzo[c]quinolizinium compounds or phenantrolines. Experimental results are not always unambiguous, and adverse effects have been incompletely tested. Some clinical tests have been done on sodium phenyl butyrate, GSNO and genistein, mostly in respect to other diseases, and the results demonstrate that these drugs are reasonably well tolerated. Their efficiency in the treatment of CF has not yet been demonstrated, however. An alternative strategy is to compensate for the defective chloride transport by CFTR by stimulation of other chloride channels. This can be done via purinergic receptors. A phase I study using a stable uridine triphosphate analog has recently been completed. A second alternative strategy is to attempt to maintain hydration of the airway mucus by inhibiting Na(+) uptake by the epithelial Na(+) channel using amiloride or stable analogs of amiloride. Clinical tests so far have been inconclusive. A number of other suggestions are currently being explored. The minority of patients with CF who have a stop mutation may benefit from treatment with gentamicin. The difficulties in finding a pharmacologic treatment for CF may be due to the fact that CFTR has additional functions besides chloride transport, and interfering with CFTR biosynthesis or activation implies interference with central cellular processes, which may have undesirable adverse effects.
Collapse
Affiliation(s)
- Godfried M Roomans
- Department of Medical Cell Biology, University of Uppsala, Uppsala, Sweden.
| |
Collapse
|
36
|
Fitzpatrick AM, Jones DP, Brown LAS. Glutathione redox control of asthma: from molecular mechanisms to therapeutic opportunities. Antioxid Redox Signal 2012; 17:375-408. [PMID: 22304503 PMCID: PMC3353819 DOI: 10.1089/ars.2011.4198] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 01/22/2012] [Accepted: 01/22/2012] [Indexed: 12/11/2022]
Abstract
Asthma is a chronic inflammatory disorder of the airways associated with airway hyper-responsiveness and airflow limitation in response to specific triggers. Whereas inflammation is important for tissue regeneration and wound healing, the profound and sustained inflammatory response associated with asthma may result in airway remodeling that involves smooth muscle hypertrophy, epithelial goblet-cell hyperplasia, and permanent deposition of airway extracellular matrix proteins. Although the specific mechanisms responsible for asthma are still being unraveled, free radicals such as reactive oxygen species and reactive nitrogen species are important mediators of airway tissue damage that are increased in subjects with asthma. There is also a growing body of literature implicating disturbances in oxidation/reduction (redox) reactions and impaired antioxidant defenses as a risk factor for asthma development and asthma severity. Ultimately, these redox-related perturbations result in a vicious cycle of airway inflammation and injury that is not always amenable to current asthma therapy, particularly in cases of severe asthma. This review will discuss disruptions of redox signaling and control in asthma with a focus on the thiol, glutathione, and reduced (thiol) form (GSH). First, GSH synthesis, GSH distribution, and GSH function and homeostasis are discussed. We then review the literature related to GSH redox balance in health and asthma, with an emphasis on human studies. Finally, therapeutic opportunities to restore the GSH redox balance in subjects with asthma are discussed.
Collapse
Affiliation(s)
- Anne M Fitzpatrick
- Department of Pediatrics, Emory University, Atlanta, Georgia 30322, USA.
| | | | | |
Collapse
|
37
|
Kuipers I, Bracke KR, Brusselle GG, Wouters EFM, Reynaert NL. Smoke decreases reversible oxidations S-glutathionylation and S-nitrosylation in mice. Free Radic Res 2012; 46:164-73. [PMID: 22145974 DOI: 10.3109/10715762.2011.647011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Cigarette smoke causes irreversible oxidations in lungs, but its impact on reversible and physiologically relevant redox-dependent protein modifications remains to be investigated. Here the effect of cigarette smoke exposure in mice was investigated on the covalent binding of glutathione to protein thiols, known as S-glutathionylation (PSSG), which can be reversed by glutaredoxins (Grx). Also, protein S-nitrosylation (PSNO) which is the modification of protein thiols by NO and which is reversed by the enzyme alcohol dehydrogenase (ADH) 5 was examined. Both PSSG and PSNO levels in lung tissue were markedly decreased after 4 weeks of cigarette smoke exposure. This coincided with attenuated protein free thiol levels and increased protein carbonylation. The expression of NOX4, DHE sensitive oxidant production and iNOS levels were induced by smoke, whereas Grx1 mRNA expression and activity were attenuated. Free GSH levels, protein expression and activity of ADH5 were unaffected by smoke. Taken together, smoke exposure decreases reversible cysteine oxidations PSSG and PSNO and enhances protein carbonylation. These alterations are not associated with differences in some of the regulatory enzymes, but are likely the result of oxidative stress.
Collapse
Affiliation(s)
- Ine Kuipers
- Department of Respiratory Medicine, Nutrim School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | | | | | | | | |
Collapse
|
38
|
Ghosh S, Erzurum SC. Nitric oxide metabolism in asthma pathophysiology. BIOCHIMICA ET BIOPHYSICA ACTA 2011; 1810:1008-16. [PMID: 21718755 PMCID: PMC3200501 DOI: 10.1016/j.bbagen.2011.06.009] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Revised: 05/24/2011] [Accepted: 06/15/2011] [Indexed: 12/22/2022]
Abstract
BACKGROUND Asthma, a chronic inflammatory disease is typically characterized by bronchoconstriction and airway hyper-reactivity. SCOPE OF REVIEW A wealth of studies applying chemistry, molecular and cell biology to animal model systems and human asthma over the last decade has revealed that asthma is associated with increased synthesis of the gaseous molecule nitric oxide (NO). MAJOR CONCLUSION The high NO levels in the oxidative environment of the asthmatic airway lead to greater formation of reactive nitrogen species (RNS) and subsequent oxidation and nitration of proteins, which adversely affect protein functions that are biologically relevant to chronic inflammation. In contrast to the high levels of NO and nitrated products, there are lower levels of beneficial S-nitrosothiols (RSNO), which mediate bronchodilation, due to greater enzymatic catabolism of RSNO in the asthmatic airways. GENERAL SIGNIFICANCE This review discusses the rapidly accruing data linking metabolic products of NO as critical determinants in the chronic inflammation and airway reactivity of asthma. This article is part of a Special Issue entitled Biochemistry of Asthma.
Collapse
Affiliation(s)
- Sudakshina Ghosh
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| | | |
Collapse
|
39
|
Matsumoto A, Gow AJ. Membrane transfer of S-nitrosothiols. Nitric Oxide 2011; 25:102-7. [PMID: 21377531 PMCID: PMC3130086 DOI: 10.1016/j.niox.2011.02.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Revised: 02/12/2011] [Accepted: 02/24/2011] [Indexed: 02/07/2023]
Abstract
The distinctive function of nitric oxide (NO) in biology is to transmit cellular signals through membranes and regulate cellular functions in adjacent cells. NO conveys signals as a second messenger from a cell where NO is generated to contiguous cells in two ways; one is as gaseous molecule by free diffusion resulting in an activation of soluble guanylate cyclase (NO/cGMP pathway), and another form is by binding with a molecule such as cysteine or protein thiol through S-nitrosylation (SNO pathway). Both pathways transmit much of the biological influence of NO from cell where other messenger molecules but NO are confined, through the plasma membrane to the adjacent cells. Since SNO pathway cannot utilize free-diffusion mechanism to get through the membrane as the molecular size is significantly larger than NO molecule, it utilizes amino acid transporter to convey signals as a form of S-nitrosylated cysteine (CysNO). Although S-nitrosylated glutathione (GSNO) is the molecule which act as a determinant of the total S-nitrosothiol level in cell, transnitrosylation reaction from GSNO to CysNO is an initial requirement to pass through signal through the membrane. Thus, multiplexed combination of these steps and the regulatory factors involved in this system conform and modify the outcome from stimulus-response coupling via the SNO pathway.
Collapse
Affiliation(s)
- Akio Matsumoto
- Department of Pharmacology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana Chuo-ku Chiba, 260-8670, Japan,
| | - Andrew J. Gow
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, 160 Frelinghuysen Road, Piscataway, NJ 08854, U.S.A.,
| |
Collapse
|
40
|
Foster MW, Yang Z, Potts EN, Michael Foster W, Que LG. S-nitrosoglutathione supplementation to ovalbumin-sensitized and -challenged mice ameliorates methacholine-induced bronchoconstriction. Am J Physiol Lung Cell Mol Physiol 2011; 301:L739-44. [PMID: 21784966 PMCID: PMC3213990 DOI: 10.1152/ajplung.00134.2011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
S-nitrosoglutathione (GSNO) is an endogenous bronchodilator present in micromolar concentrations in airway lining fluid. Airway GSNO levels decrease in severe respiratory failure and asthma, which is attributable to increased metabolism by GSNO reductase (GSNOR). Indeed, we have found that GSNOR expression and activity correlate inversely with lung S-nitrosothiol (SNO) content and airway hyperresponsiveness (AHR) to methacholine (MCh) challenge in humans with asthmatic phenotypes (Que LG, Yang Z, Stamler JS, Lugogo NL, Kraft M. Am J Respir Crit Care Med 180: 226-231, 2009). Accordingly, we hypothesized that local aerosol delivery of GSNO could ameliorate AHR and inflammation in the ovalbumin-sensitized and -challenged (OVA) mouse model of allergic asthma. Anesthetized, paralyzed, and tracheotomized 6-wk-old male control and OVA C57BL/6 mice were administered a single 15-s treatment of 0-100 mM GSNO. Five minutes later, airway resistance to MCh was measured and SNOs were quantified in bronchoalveolar lavage (BAL). Duration of protection was evaluated following nose-only exposure to 10 mM GSNO for 10 min followed by measurements of airway resistance, inflammatory cells, and cytokines and chemokines at up to 4 h later. Acute delivery of GSNO aerosol protected OVA mice from MCh-induced AHR, with no benefit seen above 20 mM GSNO. The antibronchoconstrictive effects of GSNO aerosol delivered via nose cone were sustained for at least 4 h. However, administration of GSNO did not alter total BAL cell counts or cell differentials and had modest effects on cytokine and chemokine levels. In conclusion, in the OVA mouse model of allergic asthma, aerosolized GSNO has rapid and sustained antibronchoconstrictive effects but does not substantially alter airway inflammation.
Collapse
Affiliation(s)
- Matthew W Foster
- Division of Pulmonary, Allergy and Critical Care Medicine, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | |
Collapse
|
41
|
Kelleher ZT, Potts EN, Brahmajothi MV, Foster MW, Auten RL, Foster WM, Marshall HE. NOS2 regulation of LPS-induced airway inflammation via S-nitrosylation of NF-{kappa}B p65. Am J Physiol Lung Cell Mol Physiol 2011; 301:L327-33. [PMID: 21724860 DOI: 10.1152/ajplung.00463.2010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Inducible nitric oxide synthase (NOS2) expression is increased in the airway epithelium in acute inflammatory disorders although the physiological impact remains unclear. We have previously shown that NOS2 inhibits NF-κB (p50-p65) activation in respiratory epithelial cells by inducing S-nitrosylation of the p65 monomer (SNO-p65). In addition, we have demonstrated that mouse lung SNO-p65 levels are acutely depleted in a lipopolysaccharide (LPS) model of lung injury and that augmenting SNO-p65 levels before LPS treatment results in decreased airway epithelial NF-κB activation, airway inflammation, and lung injury. We now show that aerosolized LPS induces NOS2 expression in the respiratory epithelium concomitant with an increase in lung SNO-p65 levels and a decrease in airway NF-κB activity. Genetic deletion of NOS2 results in an absence of SNO-p65 formation, persistent NF-κB activity in the respiratory epithelium, and prolonged airway inflammation. These results indicate that a primary function of LPS-induced NOS2 expression in the respiratory epithelium is to modulate the inflammatory response through deactivation of NF-κB via S-nitrosylation of p65, thereby counteracting the initial stimulus-coupled denitrosylation.
Collapse
Affiliation(s)
- Zachary T Kelleher
- Division of Pulmonary Allergy, and Critical Care Medicine, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Marozkina NV, Gaston B. S-Nitrosylation signaling regulates cellular protein interactions. Biochim Biophys Acta Gen Subj 2011; 1820:722-9. [PMID: 21745537 DOI: 10.1016/j.bbagen.2011.06.017] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 06/13/2011] [Accepted: 06/16/2011] [Indexed: 10/18/2022]
Abstract
BACKGROUND S-Nitrosothiols are made by nitric oxide synthases and other metalloproteins. Unlike nitric oxide, S-nitrosothiols are involved in localized, covalent signaling reactions in specific cellular compartments. These reactions are enzymatically regulated. SCOPE S-Nitrosylation affects interactions involved in virtually every aspect of normal cell biology. This article is part of a Special Issue entitled Regulation of Cellular Processes by S-nitrosylation. MAJOR CONCLUSIONS AND SIGNIFICANCE S-Nitrosylation is a regulated signaling reaction.
Collapse
Affiliation(s)
- Nadzeya V Marozkina
- University of Virginia School of Medicine, Division of Pediatric Respiratory Medicine, PO Box 800386, Charlottesville, VA 22908, USA.
| | | |
Collapse
|
43
|
Gaston B. The biochemistry of asthma. Biochim Biophys Acta Gen Subj 2011; 1810:1017-24. [PMID: 21718756 DOI: 10.1016/j.bbagen.2011.06.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 05/18/2011] [Accepted: 06/15/2011] [Indexed: 01/27/2023]
Abstract
BACKGROUND Asthma is not one disease. Different patients have biochemically distinct phenotypes. SCOPE OF REVIEW Biomarker analysis was developed to identify inflammation in the asthmatic airway. It has led to a renewed interest in biochemical abnormalities in the asthmatic airway. The biochemical determinants of asthma heterogeneity are many. Examples include decreased activity of superoxide dismutases; increased activity of eosinophil peroxidase, S-nitrosoglutathione reductase, and arginases; decreased airway pH; and increased levels of asymmetric dimethyl arginine. MAJOR CONCLUSIONS New discoveries suggest that biomarkers such as exhaled nitric oxide reflect complex airway biochemistry. This biochemistry can be informative and therapeutically relevant. GENERAL SIGNIFICANCE Improved understanding of airway biochemistry will lead to new tests to identify biochemically unique subpopulations of patients with asthma. It will also likely lead to new, targeted treatments for these specific asthma subpopulations. This article is part of a Special Issue entitled Biochemistry of Asthma.
Collapse
Affiliation(s)
- Benjamin Gaston
- Universit of Virginia School of Medicine, Pediatric Respiratory Medicine, Charlottes, VA 22908-0386, USA.
| |
Collapse
|
44
|
Nitrative stress in inflammatory lung diseases. Nitric Oxide 2011; 25:138-44. [PMID: 21440655 DOI: 10.1016/j.niox.2011.03.079] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Revised: 03/09/2011] [Accepted: 03/23/2011] [Indexed: 12/14/2022]
Abstract
Since the discovery of nitric oxide (NO), an intracellular signal transmitter, the role of NO has been investigated in various organs. In the respiratory system, NO derived from the constitutive type of NO synthase (cNOS, NOS1, NOS3) induces bronchodilation and pulmonary vasodilatation to maintain homeostasis. In contrast, the roles of excessive NO derived from the inducible type of NOS (iNOS, NOS2) in airway and lung inflammation in inflammatory lung diseases including bronchial asthma and chronic obstructive pulmonary disease (COPD) are controversial. In these inflammatory lung diseases, excessive nitrosative stress has also been observed. In asthma, some reports have shown that nitrosative stress causes airway inflammation, airway hyperresponsiveness, and airway remodeling, which are the features of asthma, whereas others have demonstrated the anti-inflammatory role of NO derived from NOS2. In the case of refractory asthma, more nitrosative stress has been reported to be observed in such airways compared with that in well-controlled asthmatics. In COPD, reactive nitrogen species (RNS), which are NO and NO-related molecules including nitrogen dioxide and peroxynitrite, cause lung inflammation, oxidative stress, activation of matrix metalloproteinase, and inactivation of antiprotease, which are involved in the pathophysiology of the disease. In the present paper, we review the physiological and pathophysiological effects of NO and NO-related molecules in the respiratory system and in inflammatory lung diseases.
Collapse
|
45
|
Rolla G, Malinovschi A, Badiu I, Heffler E, Petrarulo M, Bucca C, Brussino L. The increase in exhaled NO following allergen challenge is not associated with airway acidification. Eur J Clin Invest 2011; 41:411-6. [PMID: 21114492 DOI: 10.1111/j.1365-2362.2010.02423.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Exhaled nitric oxide (NO), commonly accepted marker of airways inflammation, may be generated both by specific enzymes, NO synthases, as well as by nonenzymatic reduction in its metabolites. During asthma exacerbations, owing to lower airways pH, it has been reported that nitrite reduction may contribute to the increase in exhaled NO. Allergen exposure, an important cause of asthma exacerbations, is also known to increase exhaled NO. DESIGN To investigate whether cat allergen exposure of cat-sensitized asthmatics leads to airway acidification, which could explain the expected increase in exhaled NO. Twelve nonsmoking, cat-sensitized patients (nine women) aged 33·5 (22-54) years with mild intermittent asthma performed a cat allergen challenge. Exhaled NO at 50-200 mL s(-1), nasal NO, exhaled breath condensate (EBC) pH, nitrite and nitrate were measured before, 8 and 24 h after allergen challenge. RESULTS A significant increase in FE(NO 50) was observed 24 h after allergen challenge compared to baseline: 110 ppb (34, 143) vs. 60 ppb (19, 122), P = 0·006. This was mainly explained by an increase in bronchial NO flux (P = 0·02), while no changes in EBC pH were observed (P = 0·35). CONCLUSIONS Allergen exposure is not associated with airways acidification, implying that the observed increase in exhaled NO is probably because of enzymatic NO production.
Collapse
Affiliation(s)
- Giovanni Rolla
- Allergy and Clinical Immunology, University of Torino, AO Mauriziano Umberto I, Torino, Italy.
| | | | | | | | | | | | | |
Collapse
|
46
|
Lee BJ, Jeung YJ, Lee JY, Choi DC. Increased S-nitrosothiol levels in nonasthmatic eosinophilic bronchitis compared with cough variant asthma. Int Arch Allergy Immunol 2011; 156:99-103. [PMID: 21447965 DOI: 10.1159/000321919] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Accepted: 10/04/2010] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Nonasthmatic eosinophilic bronchitis (NAEB) and cough variant asthma (CVA) are common causes of chronic cough. Both are characterized by eosinophilic inflammation in the airways. However, airway hyperresponsiveness, which is a characteristic feature of CVA, is not observed in NAEB. We hypothesized that endogenous bronchodilator S-nitrosothiol (SNO) levels are different between patients with NAEB and CVA. METHODS SNO concentrations in sputum supernatant were measured using a commercially available kit in 20 NAEB and 21 CVA patients. RESULTS The mean sputum eosinophil counts and exhaled nitric oxide values were similar in patients with NAEB (12.4 ± 2.3%, 80.6 ± 8.1 ppb) and CVA (15.3 ± 3.7%, 97.7 ± 9.2 ppb). By contrast, SNO levels in the airway lining fluid of NAEB patients were substantially higher than those of CVA patients (87.1 ± 9.8 vs. 46.8 ± 4.8 μM; p < 0.05). CONCLUSIONS SNOs may be an important factor in determining the development of airway hyperresponsiveness in the presence of eosinophilic inflammation.
Collapse
Affiliation(s)
- Byung-Jae Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | | | | |
Collapse
|
47
|
Liu L, Teague WG, Erzurum S, Fitzpatrick A, Mantri S, Dweik RA, Bleecker ER, Meyers D, Busse WW, Calhoun WJ, Castro M, Chung KF, Curran-Everett D, Israel E, Jarjour WN, Moore W, Peters SP, Wenzel S, Hunt JF, Gaston B, National Heart, Lung, and Blood Institute Severe Asthma Research Program (SARP). Determinants of exhaled breath condensate pH in a large population with asthma. Chest 2011; 139:328-336. [PMID: 20966042 PMCID: PMC3032364 DOI: 10.1378/chest.10-0163] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Accepted: 08/09/2010] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Exhaled breath condensate (EBC) pH is 2 log orders below normal during acute asthma exacerbations and returns to normal with antiinflammatory therapy. However, the determinants of EBC pH, particularly in stable asthma, are poorly understood. We hypothesized that patients with severe asthma would have low EBC pH and that there would be an asthma subpopulation of patients with characteristically low values. METHODS We studied the association of EBC pH with clinical characteristics in 572 stable subjects enrolled in the Severe Asthma Research Program. These included 250 subjects with severe asthma, 291 with nonsevere asthma, and 31 healthy control subjects. RESULTS Overall, EBC in this population of stable, treated study subjects was not lower in severe asthma (8.02; interquartile range [IQR], 7.61-8.41) or nonsevere asthma (7.90; IQR, 7.52-8.20) than in control subjects (7.9; IQR, 7.40-8.20). However, in subjects with asthma the data clustered below and above pH 6.5. Subjects in the subpopulation with pH < 6.5 had lower fraction of exhaled NO (FeNO) values (FeNO = 22.6 ± 18.1 parts per billion) than those with pH ≥ 6.5 (39.9 ± 40.2 parts per billion; P < .0001). By multiple linear regression, low EBC pH was associated with high BMI, high BAL neutrophil counts, low prebronchodilator FEV(1) ratio, high allergy symptoms, race other than white, and gastroesophageal reflux symptoms. CONCLUSION Asthma is a complex syndrome. Subjects who are not experiencing an exacerbation but have low EBC pH appear to be a unique subpopulation.
Collapse
Affiliation(s)
- Lei Liu
- Department of Public Health Sciences at the University of Virginia, Charlottesville, VA
| | | | - Serpil Erzurum
- Department of Pathobiology, the Cleveland Clinic, Cleveland, OH; Department of Pulmonary, Allergy, and Critical Care Medicine, the Cleveland Clinic, Cleveland, OH
| | | | | | - Raed A Dweik
- Department of Pathobiology, the Cleveland Clinic, Cleveland, OH; Department of Pulmonary, Allergy, and Critical Care Medicine, the Cleveland Clinic, Cleveland, OH
| | | | - Deborah Meyers
- Department of Medicine, Wake Forest University, Winston-Salem, NC
| | - William W Busse
- Department of Medicine, University of Wisconsin, Madison, WI
| | | | - Mario Castro
- Department of Medicine, Washington University, St. Louis, MO
| | | | | | | | - W Nizar Jarjour
- Department of Medicine, University of Wisconsin, Madison, WI
| | - Wendy Moore
- Department of Medicine, Wake Forest University, Winston-Salem, NC
| | - Stephen P Peters
- Department of Medicine, Wake Forest University, Winston-Salem, NC
| | | | - John F Hunt
- Department of Pediatrics, Charlottesville, VA
| | | | | |
Collapse
|
48
|
Olson N, Kasahara DI, Hristova M, Bernstein R, Janssen-Heininger Y, van der Vliet A. Modulation of NF-κB and hypoxia-inducible factor--1 by S-nitrosoglutathione does not alter allergic airway inflammation in mice. Am J Respir Cell Mol Biol 2010; 44:813-23. [PMID: 20693401 DOI: 10.1165/rcmb.2010-0035oc] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Induction of nitric oxide synthase (NOS)-2 and production of nitric oxide (NO) are common features of allergic airway disease. Conditions of severe asthma are associated with deficiency of airway S-nitrosothiols, a biological product of NO that can suppress inflammation by S-nitrosylation of the proinflammatory transcription factor, NF-κB. Therefore, restoration of airway S-nitrosothiols might have therapeutic benefit, and this was tested in a mouse model of ovalbumin (OVA)-induced allergic inflammation. Naive or OVA-sensitized animals were administered S-nitrosoglutathione (GSNO; 50 μl, 10 mM) intratracheally before OVA challenge and analyzed 48 hours later. GSNO administration enhanced lung tissue S-nitrosothiol levels and reduced NF-κB activity in OVA-challenged animals compared with control animals, but did not lead to significant changes in total bronchoalveolar lavage cell counts, differentials, or mucus metaplasia markers. Administration of GSNO also altered the activation of hypoxia-inducible factor (HIF)-1, leading to HIF-1 activation in naive mice, but suppressed HIF-1 activation in OVA-challenged mice. We assessed the contribution of endogenous NOS2 in regulating NF-κB and/or HIF-1 activation and allergic airway inflammation using NOS2(-/-) mice. Although OVA-induced NF-κB activation was slightly increased in NOS2(-/-) mice, associated with small increases in bronchoalveolar lavage neutrophils, other markers of allergic inflammation and HIF-1 activation were similar in NOS2(-/-) and wild-type mice. Collectively, our studies indicate that instillation of GSNO can suppress NF-κB activation during allergic airway inflammation, but does not significantly affect overall markers of inflammation or mucus metaplasia, thus potentially limiting its therapeutic potential due to effects on additional signaling pathways, such as HIF-1.
Collapse
Affiliation(s)
- Nels Olson
- Department of Pathology, College of Medicine, University of Vermont, Burlington, Vermont 05405, USA
| | | | | | | | | | | |
Collapse
|
49
|
Greenwald R, Fitzpatrick AM, Gaston B, Marozkina NV, Erzurum S, Teague WG. Breath formate is a marker of airway S-nitrosothiol depletion in severe asthma. PLoS One 2010; 5:e11919. [PMID: 20689836 PMCID: PMC2912922 DOI: 10.1371/journal.pone.0011919] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Accepted: 07/07/2010] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Children with severe asthma have poor symptom control and elevated markers of airway oxidative and nitrosative stress. Paradoxically, they have decreased airway levels of S-nitrosothiols (SNOs), a class of endogenous airway smooth muscle relaxants. This deficiency results from increased activity of an enzyme that both reduces SNOs to ammonia and oxidizes formaldehyde to formic acid, a volatile carboxylic acid that is more easily detected in exhaled breath condensate (EBC) than SNOs. We therefore hypothesize that depletion of airway SNOs is related to asthma pathology, and breath formate concentration may be a proxy measure of SNO catabolism. METHODS AND FINDINGS We collected EBC samples from children and adolescents, including 38 with severe asthma, 46 with mild-to-moderate asthma and 16 healthy adolescent controls, and the concentration of ionic constituents was quantified using ion chromatography. The concentrations of EBC components with volatile conjugates were log-normally distributed. Formate was the principal ion that displayed a significant difference between asthma status classifications. The mean EBC formate concentration was 40% higher in samples collected from all asthmatics than from healthy controls (mean = 5.7 microM, mean+/-standard deviation = 3.1-10.3 microM vs. 4.0, 2.8-5.8 microM, p = 0.05). EBC formate was higher in severe asthmatics than in mild-to-moderate asthmatics (6.8, 3.7-12.3 microM vs. 4.9, 2.8-8.7 microM, p = 0.012). In addition, formate concentration was negatively correlated with methacholine PC(20) (r = -0.39, p = 0.002, asthmatics only), and positively correlated with the NO-derived ion nitrite (r = 0.46, p<0.0001) as well as with total serum IgE (r = 0.28, p = 0.016, asthmatics only). Furthermore, formate was not significantly correlated with other volatile organic acids nor with inhaled corticosteroid dose. CONCLUSIONS We conclude that EBC formate concentration is significantly higher in the breath of children with asthma than in those without asthma. In addition, amongst asthmatics, formate is elevated in the breath of those with severe asthma compared to those with mild-to-moderate asthma. We suggest that this difference is related to asthma pathology and may be a product of increased catabolism of endogenous S-nitrosothiols.
Collapse
Affiliation(s)
- Roby Greenwald
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, United States of America
| | - Anne M. Fitzpatrick
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Benjamin Gaston
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Nadzeya V. Marozkina
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Serpil Erzurum
- Department of Medicine and Pathobiology, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - W. Gerald Teague
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
50
|
Marozkina NV, Yemen S, Borowitz M, Liu L, Plapp M, Sun F, Islam R, Erdmann-Gilmore P, Townsend RR, Lichti CF, Mantri S, Clapp PW, Randell SH, Gaston B, Zaman K. Hsp 70/Hsp 90 organizing protein as a nitrosylation target in cystic fibrosis therapy. Proc Natl Acad Sci U S A 2010; 107:11393-8. [PMID: 20534503 PMCID: PMC2895117 DOI: 10.1073/pnas.0909128107] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The endogenous signaling molecule S-nitrosoglutathione (GSNO) and other S-nitrosylating agents can cause full maturation of the abnormal gene product DeltaF508 cystic fibrosis (CF) transmembrane conductance regulator (CFTR). However, the molecular mechanism of action is not known. Here we show that Hsp70/Hsp90 organizing protein (Hop) is a critical target of GSNO, and its S-nitrosylation results in DeltaF508 CFTR maturation and cell surface expression. S-nitrosylation by GSNO inhibited the association of Hop with CFTR in the endoplasmic reticulum. This effect was necessary and sufficient to mediate GSNO-induced cell-surface expression of DeltaF508 CFTR. Hop knockdown using siRNA recapitulated the effect of GSNO on DeltaF508 CFTR maturation and expression. Moreover, GSNO acted additively with decreased temperature, which promoted mutant CFTR maturation through a Hop-independent mechanism. We conclude that GSNO corrects DeltaF508 CFTR trafficking by inhibiting Hop expression, and that combination therapies--using differing mechanisms of action--may have additive benefits in treating CF.
Collapse
Affiliation(s)
| | - Sean Yemen
- Department of Pediatric Respiratory Medicine and
| | | | - Lei Liu
- Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA 22908
| | | | - Fei Sun
- Department of Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Rafique Islam
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843
| | | | - R. Reid Townsend
- Washington University Proteomics Center, St. Louis, MO 63108-2259; and
| | - Cheryl F. Lichti
- Washington University Proteomics Center, St. Louis, MO 63108-2259; and
| | - Sneha Mantri
- Department of Pediatric Respiratory Medicine and
| | - Phillip W. Clapp
- Department of Medicine, University of North Carolina, Chapel Hill, NC 27599
| | - Scott H. Randell
- Department of Medicine, University of North Carolina, Chapel Hill, NC 27599
| | | | | |
Collapse
|