1
|
Yang K, He T, Sun X, Dong W. Post-translational modifications and bronchopulmonary dysplasia. Front Pediatr 2025; 12:1426030. [PMID: 39830627 PMCID: PMC11738936 DOI: 10.3389/fped.2024.1426030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
Bronchopulmonary dysplasia is a prevalent respiratory disorder posing a significant threat to the quality of life in premature infants. Its pathogenesis is intricate, and therapeutic options are limited. Besides genetic coding, protein post-translational modification plays a pivotal role in regulating cellular function, contributing complexity and diversity to substrate proteins and influencing various cellular processes. Substantial evidence indicates that post-translational modifications of several substrate proteins are intricately related to the molecular mechanisms underlying bronchopulmonary dysplasia. These modifications facilitate the progression of bronchopulmonary dysplasia through a cascade of signal transduction events. This review outlines the relationships between substrate protein phosphorylation, acetylation, ubiquitination, SUMOylation, methylation, glycosylation, glycation, S-glutathionylation, S-nitrosylation and bronchopulmonary dysplasia. The aim is to provide novel insights into bronchopulmonary dysplasia's pathogenesis and potential therapeutic targets for clinical management.
Collapse
Affiliation(s)
| | | | | | - Wenbin Dong
- Department of Neonatology, Children’s Medical Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
2
|
Maeda H, Li X, Go H, Dennery PA, Yao H. miRNA Signatures in Bronchopulmonary Dysplasia: Implications for Biomarkers, Pathogenesis, and Therapeutic Options. FRONT BIOSCI-LANDMRK 2024; 29:271. [PMID: 39082345 PMCID: PMC11799892 DOI: 10.31083/j.fbl2907271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/21/2024] [Accepted: 05/27/2024] [Indexed: 01/18/2025]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease in premature infants characterized by alveolar dysplasia, vascular simplification and dysmorphic vascular development. Supplemental oxygen and mechanical ventilation commonly used as life-saving measures in premature infants may cause BPD. microRNAs (miRNAs), a class of small, non-coding RNAs, regulate target gene expression mainly through post-transcriptional repression. miRNAs play important roles in modulating oxidative stress, proliferation, apoptosis, senescence, inflammatory responses, and angiogenesis. These cellular processes play pivotal roles in the pathogenesis of BPD. Accumulating evidence demonstrates that miRNAs are dysregulated in the lung of premature infants with BPD, and in animal models of this disease, suggesting contributing roles of dysregulated miRNAs in the development of BPD. Therefore, miRNAs are considered promising biomarker candidates and therapeutic agents for this disease. In this review, we discuss how dysregulated miRNAs and their modulation alter cellular processes involved in BPD. We then focus on therapeutic approaches targeting miRNAs for BPD. This review provides an overview of miRNAs as biomarkers, and highlights potential pathogenic roles, and therapeutic strategies for BPD using miRNAs.
Collapse
Affiliation(s)
- Hajime Maeda
- Department of Molecular Biology, Cellular Biology, and Biochemistry, Brown University, Providence, RI 02912, USA
- Department of Pediatrics, Fukushima Medical University School of Medicine, 960-1295 Fukushima, Japan
| | - Xiaoyun Li
- Department of Molecular Biology, Cellular Biology, and Biochemistry, Brown University, Providence, RI 02912, USA
- Providence Veterans Affairs Medical Center, Providence, RI 02908, USA
- Department of Medicine, Warren Alpert School of Medicine of Brown University, Providence, RI 02903, USA
- College of Pharmacy, Jinan University, 510632 Guangzhou, Guangdong, China
| | - Hayato Go
- Department of Pediatrics, Fukushima Medical University School of Medicine, 960-1295 Fukushima, Japan
| | - Phyllis A. Dennery
- Department of Molecular Biology, Cellular Biology, and Biochemistry, Brown University, Providence, RI 02912, USA
- Department of Pediatrics, Warren Alpert School of Medicine of Brown University, Providence, RI 02903, USA
| | - Hongwei Yao
- Department of Molecular Biology, Cellular Biology, and Biochemistry, Brown University, Providence, RI 02912, USA
- Providence Veterans Affairs Medical Center, Providence, RI 02908, USA
- Department of Medicine, Warren Alpert School of Medicine of Brown University, Providence, RI 02903, USA
| |
Collapse
|
3
|
Reçica R, Kryeziu I, Thaçi Q, Avtanski D, Mladenov M, Basholli-Salihu M, Sopi RB. Protective Effects of Resveratrol Against Airway Hyperreactivity, Oxidative Stress, and Lung Inflammation in a Rat Pup Model of Bronchopulmonary Dysplasia. Physiol Res 2024; 73:239-251. [PMID: 38710061 PMCID: PMC11081184 DOI: 10.33549/physiolres.935239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/10/2024] [Indexed: 12/20/2024] Open
Abstract
Oxygen therapy provides an important treatment for preterm and low-birth-weight neonates, however, it has been shown that prolonged exposure to high levels of oxygen (hyperoxia) is one of the factors contributing to the development of bronchopulmonary dysplasia (BPD) by inducing lung injury and airway hyperreactivity. There is no effective therapy against the adverse effects of hyperoxia. Therefore, this study was undertaken to test the hypothesis that natural phytoalexin resveratrol will overcome hyperoxia-induced airway hyperreactivity, oxidative stress, and lung inflammation. Newborn rats were exposed to hyperoxia (fraction of inspired oxygen - FiO2>95 % O2) or ambient air (AA) for seven days. Resveratrol was supplemented either in vivo (30 mg·kg-1·day-1) by intraperitoneal administration or in vitro to the tracheal preparations in an organ bath (100 mikroM). Contractile and relaxant responses were studied in tracheal smooth muscle (TSM) using the in vitro organ bath system. To explain the involvement of nitric oxide in the mechanisms of the protective effect of resveratrol against hyperoxia, a nitric oxide synthase inhibitor - Nomega-nitro-L-arginine methyl ester (L-NAME), was administered in some sets of experiments. The superoxide dismutase (SOD) and glutathione peroxidase (GPx) activities and the tumor necrosis factor-alpha (TNF-alpha) and interleukin-1beta (IL-1beta) levels in the lungs were determined. Resveratrol significantly reduced contraction and restored the impaired relaxation of hyperoxia-exposed TSM (p<0.001). L-NAME reduced the inhibitory effect of resveratrol on TSM contractility, as well as its promotion relaxant effect (p<0.01). Resveratrol preserved the SOD and GPx activities and decreased the expression of TNF-alpha and IL-1beta in hyperoxic animals. The findings of this study demonstrate the protective effect of resveratrol against hyperoxia-induced airway hyperreactivity and lung damage and suggest that resveratrol might serve as a therapy to prevent the adverse effects of neonatal hyperoxia. Keywords: Bronchopulmonary dysplasia, Hyperoxia, Airway hyperreactivity, Resveratrol, Pro-inflammatory cytokines.
Collapse
Affiliation(s)
- R Reçica
- Faculty of Medicine, University of Prishtina, Prishtina, Kosovo
| | | | | | | | | | | | | |
Collapse
|
4
|
Kryeziu I, Reçica S, Thaçi Q, Kurshumliu F, Hadzi-Petrushev N, Basholli-Salihu M, Mladenov M, Sopi RB. Quercetin supplementation attenuates airway hyperreactivity and restores airway relaxation in rat pups exposed to hyperoxia. Exp Biol Med (Maywood) 2023; 248:1492-1499. [PMID: 37837396 PMCID: PMC10666724 DOI: 10.1177/15353702231199468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/20/2023] [Indexed: 10/16/2023] Open
Abstract
Hyperoxia exposure of immature lungs contributes to lung injury and airway hyperreactivity. Up to now, treatments of airway hyperreactivity induced by hyperoxia exposure have been ineffective. The aim of this study was to investigate the effects of quercetin on hyperoxia-induced airway hyperreactivity, impaired relaxation, and lung inflammation. Newborn rats were exposed to hyperoxia (FiO2 > 95%) or ambient air (AA) for seven days. Subgroups were injected with quercetin (10 mg·kg-1·day-1). After exposures, tracheal cylinders were prepared for in vitro wire myography. Contraction to methacholine was measured in the presence or absence of organ bath quercetin and/or Nω-nitro-L-arginine methyl ester (L-NAME). Relaxation responses were evoked in preconstricted tissues using electrical field stimulation (EFS). Lung tumor necrosis factor-alpha (TNF-α) and interleukin-1β (IL-1β) levels were measured by enzyme-linked immunosorbent assay (ELISA). A P < 0.05 was considered statistically significant. Contractile responses of tracheal smooth muscle (TSM) of hyperoxic animals were significantly increased compared with AA animals (P < 0.001). Treatment with quercetin significantly reduced contraction in hyperoxic groups compared with hyperoxic control (P < 0.01), but did not have any effect in AA groups. In hyperoxic animals, relaxation of TSM was significantly reduced compared with AA animals (P < 0.001), while supplementation of quercetin restored the lost relaxation in hyperoxic groups. Incubation of preparations in L-NAME significantly reduced the quercetin effects on both contraction and relaxation (P < 0.01). Treatment of hyperoxic animals with quercetin significantly decreased the expression of TNF-α and IL-1β compared with hyperoxic controls (P < 0.001 and P < 0.01, respectively).The findings of this study demonstrate the protective effect of quercetin on airway hyperreactivity and suggest that quercetin might serve as a novel therapy to prevent and treat neonatal hyperoxia-induced airway hyperreactivity and inflammation.
Collapse
Affiliation(s)
- Islam Kryeziu
- Faculty of Medicine, University of Prishtina, 10 000 Prishtina, Kosovo
| | - Shkëlzen Reçica
- Faculty of Medicine, University of Prishtina, 10 000 Prishtina, Kosovo
| | - Qëndrim Thaçi
- Faculty of Medicine, University of Prishtina, 10 000 Prishtina, Kosovo
| | - Fisnik Kurshumliu
- Faculty of Medicine, University of Prishtina, 10 000 Prishtina, Kosovo
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University in Skopje, 1000 Skopje, North Macedonia
| | | | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University in Skopje, 1000 Skopje, North Macedonia
| | - Ramadan B Sopi
- Faculty of Medicine, University of Prishtina, 10 000 Prishtina, Kosovo
| |
Collapse
|
5
|
McGraw MD, Yee M, Kim SY, Dylag AM, Lawrence BP, O'Reilly MA. Diacetyl inhalation impairs airway epithelial repair in mice infected with influenza A virus. Am J Physiol Lung Cell Mol Physiol 2022; 323:L578-L592. [PMID: 36068185 PMCID: PMC9639765 DOI: 10.1152/ajplung.00124.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/12/2022] [Accepted: 08/29/2022] [Indexed: 01/11/2023] Open
Abstract
Bronchiolitis obliterans (BO) is a debilitating disease of the small airways that can develop following exposure to toxic chemicals as well as respiratory tract infections. BO development is strongly associated with diacetyl (DA) inhalation exposures at occupationally relevant concentrations or severe influenza A viral (IAV) infections. However, it remains unclear whether lower dose exposures or more mild IAV infections can result in similar pathology. In the current work, we combined these two common environmental exposures, DA and IAV, to test whether shorter DA exposures followed by sublethal IAV infection would result in similar airways disease. Adult mice exposed to DA vapors 1 h/day for 5 consecutive days followed by infection with the airway-tropic IAV H3N2 (HKx31) resulted in increased mortality, increased bronchoalveolar lavage (BAL) neutrophil percentage, mixed obstruction and restriction by lung function, and subsequent airway remodeling. Exposure to DA or IAV alone failed to result in significant pathology, whereas mice exposed to DA + IAV showed increased α-smooth muscle actin (αSMA) and epithelial cells coexpressing the basal cell marker keratin 5 (KRT5) with the club cell marker SCGB1A1. To test whether DA exposure impairs epithelial repair after IAV infection, mice were infected first with IAV and then exposed to DA during airway epithelial repair. Mice exposed to IAV + DA developed similar airway remodeling with increased subepithelial αSMA and epithelial cells coexpressing KRT5 and SCGB1A1. Our findings reveal an underappreciated concept that common environmental insults while seemingly harmless by themselves can have catastrophic implications on lung function and long-term respiratory health when combined.
Collapse
Affiliation(s)
- Matthew D McGraw
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York
| | - Min Yee
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - So-Young Kim
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - Andrew M Dylag
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - B Paige Lawrence
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York
| | - Michael A O'Reilly
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
6
|
Tang B, Ling Q, Yang Q, Li M, Shi W, Wu Q. How to survive a periviable birth baby with birth weight of 450g: A case report. Medicine (Baltimore) 2022; 101:e31356. [PMID: 36281089 PMCID: PMC9592289 DOI: 10.1097/md.0000000000031356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
RATIONALE An increasing number of periviable birth newborns (PVBs) have emerged with concurrent growing high-risk pregnancy. To date, postnatal management of PVBs remains one of the most challenging issues and limited studies have been reported. PATIENT CONCERNS A female baby born at 230/7 weeks of gestation with birth weight of 450g. DIAGNOSIS PVB baby, respiratory distress syndrome (RDS), ventilator associated pneumonia (VAP), intraventricular hemorrhage (IVH), metabolic bone disease of prematurity (MBDP), transient hypothyroxinemia of prematurity (THOP), bronchopulmonary dysplasia (BPD) and retinopathy of prematurity (ROP). INTERVENTIONS Individualized treatment and intensive care, including neonatal resuscitation, effective respiratory and circulatory support, venous access and nutrition, prevention and treatment of infection, management of endocrine and metabolic problems, individualized nursing such as developmental supportive care, integrated oral motor interventions, skin care, family-integrated-care, etc were performed according to existing literature. OUTCOMES The baby was discharged home after 138 days of hospitalization with body weight of 2700 g, a full oral feed achieved, and without any requirement of respiratory support or oxygen supply. Now she is 38-month-old, with no significant long-term adverse sequelae. LESSONS Our case expands the experience and knowledges of individualized and intensive management of PVB babies in their early life days, which increase PVBs' survival and improves their prognosis.
Collapse
Affiliation(s)
- Binzhi Tang
- Department of Pediatrics, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, Chengdu, China
- Department of Pediatrics, Clinical College of University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Qiying Ling
- Department of Pediatrics, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, Chengdu, China
- Department of Pediatrics, Clinical College of University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Qian Yang
- Department of Pediatrics, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, Chengdu, China
- Department of Pediatrics, Clinical College of University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Maojun Li
- Department of Pediatrics, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, Chengdu, China
- Department of Pediatrics, Clinical College of University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Wei Shi
- Department of Pediatrics, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, Chengdu, China
- Department of Pediatrics, Clinical College of University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Qing Wu
- Department of Pediatrics, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, Chengdu, China
- Department of Pediatrics, Clinical College of University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
- *Correspondence: Qing Wu, Department of Pediatrics, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, No. 32 West Second Section First Ring Road, Chengdu 610072, Sichuan Province, China (e-mail: )
| |
Collapse
|
7
|
Fonseca FV, Raffay TM, Xiao K, McLaughlin PJ, Qian Z, Grimmett ZW, Adachi N, Wang B, Hausladen A, Cobb BA, Zhang R, Hess DT, Gaston B, Lambert NA, Reynolds JD, Premont RT, Stamler JS. S-nitrosylation is required for β 2AR desensitization and experimental asthma. Mol Cell 2022; 82:3089-3102.e7. [PMID: 35931084 PMCID: PMC9391322 DOI: 10.1016/j.molcel.2022.06.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/18/2022] [Accepted: 06/28/2022] [Indexed: 12/22/2022]
Abstract
The β2-adrenergic receptor (β2AR), a prototypic G-protein-coupled receptor (GPCR), is a powerful driver of bronchorelaxation, but the effectiveness of β-agonist drugs in asthma is limited by desensitization and tachyphylaxis. We find that during activation, the β2AR is modified by S-nitrosylation, which is essential for both classic desensitization by PKA as well as desensitization of NO-based signaling that mediates bronchorelaxation. Strikingly, S-nitrosylation alone can drive β2AR internalization in the absence of traditional agonist. Mutant β2AR refractory to S-nitrosylation (Cys265Ser) exhibits reduced desensitization and internalization, thereby amplifying NO-based signaling, and mice with Cys265Ser mutation are resistant to bronchoconstriction, inflammation, and the development of asthma. S-nitrosylation is thus a central mechanism in β2AR signaling that may be operative widely among GPCRs and targeted for therapeutic gain.
Collapse
Affiliation(s)
- Fabio V Fonseca
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Thomas M Raffay
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Kunhong Xiao
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Precious J McLaughlin
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Zhaoxia Qian
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Zachary W Grimmett
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Naoko Adachi
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Benlian Wang
- Center for Proteomics and Bioinformatics, Department of Nutrition, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Alfred Hausladen
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Brian A Cobb
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Rongli Zhang
- Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Douglas T Hess
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Benjamin Gaston
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Nevin A Lambert
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - James D Reynolds
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Richard T Premont
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Jonathan S Stamler
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA.
| |
Collapse
|
8
|
Beqiraj-Zeqiraj Q, Thaçi Q, Sahiti F, Kovač Z, Raffay TM, Sopi RB. Rho-kinase inhibitors protect against neonatal hyperoxia-induced airway hyperreactivity in a rat pup model: Role of prostaglandin F 2α. Pediatr Pulmonol 2022; 57:1229-1237. [PMID: 35088947 DOI: 10.1002/ppul.25848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 01/06/2022] [Accepted: 01/26/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Oxygen therapy in preterm neonates is associated with airway hyperreactivity. The role of Rho/Rho-kinase smooth muscle signaling in hyperoxia-induced airway hyperreactivity remains understudied. We hypothesized that inhibition of Rho-kinase will attenuate airway hyperreactivity induced by neonatal hyperoxia. METHODS Newborn rats were raised in hyperoxia (>95% O2 ) or ambient air (AA) for 7 days. Subgroups were injected with a Rho-kinase inhibitor: Y-27632 (10 mg·kg-1 ·day-1 ) or fasudil (10 mg·kg-1 ·day-1 ), or a FP receptor antagonist - AS604872 (30 mg·kg-1 ·day-1 ). After exposures, tracheal cylinders were prepared for in vitro wire myography. Contraction to methacholine or PGF2α was measured in the presence or absence of tissue-bath Y-27632, fasudil, or AS604872. Lung PGF2α levels, Rho-kinase protein level and Rho-kinase 1 activity were measured by ELISA. RESULTS Tracheal smooth muscle contraction was significantly greater in hyperoxic compared to AA groups. Both, Y-27632 and fasudil significantly decreased contractility to MCh or PGF2α in hyperoxic groups versus hyperoxic controls (p < 0.001), but did not alter AA group responses. Inhibition of FP receptors attenuated responses to PGF2α . Hyperoxia significantly increased lung PGF2α compared to AA (p < 0.01), but Rho-kinase inhibition did not influence PGF2α level. Rho-kinase protein level (p < 0.001) and activity (p < 0.01), were increased by hyperoxia, but blockade of FP receptor reduced the Rho-kinase 1 activity (p < 0.05) under hyperoxic condition. CONCLUSIONS This study demonstrates an active role of Rho/Rho-kinase signaling on hyperoxia-induced airway hyperreactivity. These findings suggest that Rho-kinase inhibitors might serve as an effective therapy for hyperoxia-induced airway hyperreactivity.
Collapse
Affiliation(s)
- Qendresa Beqiraj-Zeqiraj
- Department of Pathophysiology, Faculty of Medicine, University of Prishtina, Prishtina, Kosovo.,Pediatric Clinic, University Clinical Centre of Kosovo, Prishtina, Kosovo
| | - Qëndrim Thaçi
- Department of Premedical Courses, Faculty of Medicine, University of Prishtina, Prishtina, Kosovo
| | - Floran Sahiti
- Department of Premedical Courses, Faculty of Medicine, University of Prishtina, Prishtina, Kosovo.,Comprehensive Heart Failure Center, University and University Hospital Würzburg, Würzburg, Germany
| | - Zdenko Kovač
- Department of Pathophysiology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Thomas M Raffay
- Department of Pediatrics, Rainbow Babies and Children's Hospital, Case Western Reserve University, Cleveland, Ohio, USA
| | - Ramadan B Sopi
- Department of Premedical Courses, Faculty of Medicine, University of Prishtina, Prishtina, Kosovo
| |
Collapse
|
9
|
Xi Y, Wang Y. Insight Into the Roles of Non-coding RNA in Bronchopulmonary Dysplasia. Front Med (Lausanne) 2021; 8:761724. [PMID: 34805228 PMCID: PMC8602187 DOI: 10.3389/fmed.2021.761724] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/13/2021] [Indexed: 02/05/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease most commonly occurring in premature infants, and its pathological manifestations are alveolar hypoplasia and dysregulation of pulmonary vasculature development. The effective treatment for BPD has not yet been established. Non-coding RNAs, including microRNAs and long non-coding RNAs do not encode proteins, but can perform its biological functions at the RNA level. Non-coding RNAs play an important role in the incidence and development of BPD by regulating the expression of genes related to proliferation, apoptosis, angiogenesis, inflammation and other cell activities of alveolar epithelial cells and vascular endothelial cells. Here we summarize the role of non-coding RNAs in BPD, which provides possible molecular marker and therapeutic target for the diagnosis and treatment of BPD.
Collapse
Affiliation(s)
- Yufeng Xi
- Department of Neonatology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yujia Wang
- Department of Neonatology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.,Department of Dermatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Raffay TM, Bonilla-Fernandez K, Jafri A, Sopi RB, Smith LA, Cui F, O'Reilly M, Zhang R, Hodges CA, MacFarlane PM, Deutsch G, Martin RJ, Gaston B. Bronchopulmonary Dysplasia and Pulmonary Hypertension. The Role of Smooth Muscle adh5. Am J Respir Cell Mol Biol 2021; 65:70-80. [PMID: 33780653 DOI: 10.1165/rcmb.2020-0289oc] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is characterized by alveolar simplification, airway hyperreactivity, and pulmonary hypertension. In our BPD model, we have investigated the metabolism of the bronchodilator and pulmonary vasodilator GSNO (S-nitrosoglutathione). We have shown the GSNO catabolic enzyme encoded by adh5 (alcohol dehydrogenase-5), GSNO reductase, is epigenetically upregulated in hyperoxia. Here, we investigated the distribution of GSNO reductase expression in human BPD and created an animal model that recapitulates the human data. Blinded comparisons of GSNO reductase protein expression were performed in human lung tissues from infants and children with and without BPD. BPD phenotypes were evaluated in global (adh5-/-) and conditional smooth muscle (smooth muscle/adh5-/-) adh5 knockout mice. GSNO reductase was prominently expressed in the airways and vessels of human BPD subjects. Compared with controls, expression was greater in BPD smooth muscle, particularly in vascular smooth muscle (2.4-fold; P = 0.003). The BPD mouse model of neonatal hyperoxia caused significant alveolar simplification, airway hyperreactivity, and right ventricular and vessel hypertrophy. Global adh5-/- mice were protected from all three aspects of BPD, whereas smooth muscle/adh5-/- mice were only protected from pulmonary hypertensive changes. These data suggest adh5 is required for the development of BPD. Expression in the pulmonary vasculature is relevant to the pathophysiology of BPD-associated pulmonary hypertension. GSNO-mimetic agents or GSNO reductase inhibitors, both of which are currently in clinical trials for other conditions, could be considered for further study in BPD.
Collapse
Affiliation(s)
| | - Koby Bonilla-Fernandez
- Division of Pediatric Pulmonology, Department of Pediatrics, University Hospitals Rainbow Babies & Children's Hospital and Case Western Reserve University School of Medicine, Case Western Reserve University, Cleveland, Ohio.,San Juan Bautista School of Medicine, Caguas, Puerto Rico
| | - Anjum Jafri
- Division of Pediatric Pulmonology, Department of Pediatrics, University Hospitals Rainbow Babies & Children's Hospital and Case Western Reserve University School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | | | - Laura A Smith
- Department of Pediatrics, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana
| | - Feifei Cui
- Division of Pediatric Pulmonology, Department of Pediatrics, University Hospitals Rainbow Babies & Children's Hospital and Case Western Reserve University School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | | | - Rongli Zhang
- Department of Medicine, Institute for Transformative Molecular Medicine, Cardiovascular Research Institute, University Hospitals Cleveland Medical Center, and
| | - Craig A Hodges
- Department of Genetics, Case Western Reserve University, Cleveland, Ohio; and
| | | | - Gail Deutsch
- Department of Pathology, Seattle Children's Hospital & University of Washington, Seattle, Washington
| | | | - Benjamin Gaston
- Division of Pediatric Pulmonology, Department of Pediatrics, University Hospitals Rainbow Babies & Children's Hospital and Case Western Reserve University School of Medicine, Case Western Reserve University, Cleveland, Ohio.,Department of Pediatrics, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana
| |
Collapse
|
11
|
Sex and Gender Differences in Lung Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:227-258. [PMID: 34019273 DOI: 10.1007/978-3-030-68748-9_14] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Sex differences in the anatomy and physiology of the respiratory system have been widely reported. These intrinsic sex differences have also been shown to modulate the pathophysiology, incidence, morbidity, and mortality of several lung diseases across the life span. In this chapter, we describe the epidemiology of sex differences in respiratory diseases including neonatal lung disease (respiratory distress syndrome, bronchopulmonary dysplasia) and pediatric and adult disease (including asthma, cystic fibrosis, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, lung cancer, lymphangioleiomyomatosis, obstructive sleep apnea, pulmonary arterial hypertension, and respiratory viral infections such as respiratory syncytial virus, influenza, and SARS-CoV-2). We also discuss the current state of research on the mechanisms underlying the observed sex differences in lung disease susceptibility and severity and the importance of considering both sex and gender variables in research studies' design and analysis.
Collapse
|
12
|
Bhatia V, Elnagary L, Dakshinamurti S. Tracing the path of inhaled nitric oxide: Biological consequences of protein nitrosylation. Pediatr Pulmonol 2021; 56:525-538. [PMID: 33289321 DOI: 10.1002/ppul.25201] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/28/2020] [Accepted: 11/29/2020] [Indexed: 12/12/2022]
Abstract
Nitric oxide (NO) is a comprehensive regulator of vascular and airway tone. Endogenous NO produced by nitric oxide synthases regulates multiple signaling cascades, including activation of soluble guanylate cyclase to generate cGMP, relaxing smooth muscle cells. Inhaled NO is an established therapy for pulmonary hypertension in neonates, and has been recently proposed for the treatment of hypoxic respiratory failure and acute respiratory distress syndrome due to COVID-19. In this review, we summarize the effects of endogenous and exogenous NO on protein S-nitrosylation, which is the selective and reversible covalent attachment of a nitrogen monoxide group to the thiol side chain of cysteine. This posttranslational modification targets specific cysteines based on the acid/base sequence of surrounding residues, with significant impacts on protein interactions and function. S-nitrosothiol (SNO) formation is tightly compartmentalized and enzymatically controlled, but also propagated by nonenzymatic transnitrosylation of downstream protein targets. Redox-based nitrosylation and denitrosylation pathways dynamically regulate the equilibrium of SNO-proteins. We review the physiological roles of SNO proteins, including nitrosohemoglobin and autoregulation of blood flow through hypoxic vasodilation, and pathological effects of nitrosylation including inhibition of critical vasodilator enzymes; and discuss the intersection of NO source and dose with redox environment, in determining the effects of protein nitrosylation.
Collapse
Affiliation(s)
- Vikram Bhatia
- Biology of Breathing Group, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada
| | - Lara Elnagary
- Biology of Breathing Group, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada
| | - Shyamala Dakshinamurti
- Biology of Breathing Group, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada.,Section of Neonatology, Departments of Pediatrics and Physiology, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
13
|
Dylag AM, Haak J, Yee M, O’Reilly MA. Pulmonary mechanics and structural lung development after neonatal hyperoxia in mice. Pediatr Res 2020; 87:1201-1210. [PMID: 31835269 PMCID: PMC7255955 DOI: 10.1038/s41390-019-0723-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 10/22/2019] [Accepted: 12/03/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Supplemental oxygen exposure administered to premature infants is associated with chronic lung disease and abnormal pulmonary function. This study used mild (40%), moderate (60%), and severe (80%) oxygen to determine how hyperoxia-induced changes in lung structure impact pulmonary mechanics in mice. METHODS C57BL/6J mice were exposed to room air or hyperoxia from birth through postnatal day 8. Baseline pulmonary function and methacholine challenge was assessed at 4 and 8 weeks of age, accompanied by immunohistochemical assessments of both airway (smooth muscle, tethering) and alveolar (simplification, elastin deposition) structure. RESULTS Mild/moderate hyperoxia increased baseline airway resistance (40% only) and airway hyperreactivity (40 and 60%) at 4 weeks accompanied by increased airway smooth muscle deposition, which resolved at 8 weeks. Severe hyperoxia increased baseline compliance, baseline resistance, and total elastin/surface area ratio without increasing airway hyperreactivity, and was accompanied by increased alveolar simplification, decreased airway tethering, and changes in elastin distribution at both time points. CONCLUSIONS Mild to moderate hyperoxia causes changes in airway function and airway hyperreactivity with minimal parenchymal response. Severe hyperoxia drives its functional changes through alveolar simplification, airway tethering, and elastin redistribution. These differential responses can be leveraged to further develop hyperoxia mouse models.
Collapse
Affiliation(s)
- Andrew M. Dylag
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Jeannie Haak
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Min Yee
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Michael A. O’Reilly
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
14
|
Owens L, Laing IA, Murdzoska J, Zhang G, Turner SW, Le Souëf PN. Glutathione S-Transferase Genotype Protects against In Utero Tobacco-linked Lung Function Deficits. Am J Respir Crit Care Med 2020; 200:462-470. [PMID: 30726102 DOI: 10.1164/rccm.201807-1332oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Rationale: In utero tobacco exposure is associated with reduced lung function from infancy. Antioxidant enzymes from the glutathione S-transferase (GST) family may protect against these lung function deficits.Objectives: To assess the long-term effect of in utero smoke exposure on lung function into adulthood, and to assess whether GSTT1 and GSTM1 active genotypes have long-term protective effects on lung function.Methods: In this longitudinal study based on a general population (n = 253), lung function was measured during infancy and at 6, 11, 18, and 24 years. GSTM1 and GSTT1 genotype was analyzed in a subgroup (n = 179). Lung function was assessed longitudinally from 6 to 24 years (n = 199).Measurements and Main Results: Exposure to maternal in utero tobacco was associated with lower FEV1 and FVC longitudinally from 6 to 24 years (mean difference, -3.87% predicted, P = 0.021; -3.35% predicted, P = 0.035, respectively). Among those homozygous for the GSTM1-null genotype, in utero tobacco exposure was associated with lower FEV1 and FVC compared with those with no in utero tobacco exposure (mean difference, -6.2% predicted, P = 0.01; -4.7% predicted, P = 0.043, respectively). For those with GSTM1 active genotype, there was no difference in lung function whether exposed to maternal in utero tobacco or not. In utero tobacco exposure was associated with deficits in lung function among those with both GSTT1-null and GSTT1-active genotypes.Conclusions: Certain GST genotypes may have protective effects against the long-term deficits in lung function associated with in utero tobacco exposure. This offers potential preventative targets in antioxidant pathways for at-risk infants of smoking mothers.
Collapse
Affiliation(s)
- Louisa Owens
- 1School of Medicine, University of Western Australia, Perth, Western Australia, Australia.,2School of Women's and Children's Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Ingrid A Laing
- 1School of Medicine, University of Western Australia, Perth, Western Australia, Australia.,3Telethon Kids Institute, Subiaco, Western Australia, Australia
| | | | - Guicheng Zhang
- 4School of Public Health, Curtin University, Bentley, Western Australia, Australia.,5Centre for Genetic Origins of Health and Disease, University of Western Australia and Curtin University, Western Australia, Australia; and
| | - Steve W Turner
- 6Child Health, University of Aberdeen, Aberdeen, United Kingdom
| | - Peter N Le Souëf
- 1School of Medicine, University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
15
|
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
|
16
|
Dylag AM, Raffay TM. Rodent models of respiratory control and respiratory system development-Clinical significance. Respir Physiol Neurobiol 2019; 268:103249. [PMID: 31315068 DOI: 10.1016/j.resp.2019.06.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/24/2019] [Accepted: 06/29/2019] [Indexed: 01/13/2023]
Abstract
The newborn infant's respiratory system must rapidly adapt to extra-uterine life. Neonatal rat and mouse models have been used to investigate early development of respiratory control and reactivity in both health and disease. This review highlights several rodent models of control of breathing and respiratory system development (including pulmonary function), discusses their translational strengths and limitations, and underscores the importance of creating clinically relevant models applicable to the human infant.
Collapse
Affiliation(s)
- Andrew M Dylag
- Division of Neonatology, Golisano Children's Hospital, Department of Pediatrics, University of Rochester, Rochester, NY, United States
| | - Thomas M Raffay
- Division of Neonatology, Rainbow Babies & Children's Hospital, Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United States.
| |
Collapse
|
17
|
Stomberski CT, Hess DT, Stamler JS. Protein S-Nitrosylation: Determinants of Specificity and Enzymatic Regulation of S-Nitrosothiol-Based Signaling. Antioxid Redox Signal 2019; 30:1331-1351. [PMID: 29130312 PMCID: PMC6391618 DOI: 10.1089/ars.2017.7403] [Citation(s) in RCA: 198] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
SIGNIFICANCE Protein S-nitrosylation, the oxidative modification of cysteine by nitric oxide (NO) to form protein S-nitrosothiols (SNOs), mediates redox-based signaling that conveys, in large part, the ubiquitous influence of NO on cellular function. S-nitrosylation regulates protein activity, stability, localization, and protein-protein interactions across myriad physiological processes, and aberrant S-nitrosylation is associated with diverse pathophysiologies. Recent Advances: It is recently recognized that S-nitrosylation endows S-nitroso-protein (SNO-proteins) with S-nitrosylase activity, that is, the potential to trans-S-nitrosylate additional proteins, thereby propagating SNO-based signals, analogous to kinase-mediated signaling cascades. In addition, it is increasingly appreciated that cellular S-nitrosylation is governed by dynamically coupled equilibria between SNO-proteins and low-molecular-weight SNOs, which are controlled by a growing set of enzymatic denitrosylases comprising two main classes (high and low molecular weight). S-nitrosylases and denitrosylases, which together control steady-state SNO levels, may be identified with distinct physiology and pathophysiology ranging from cardiovascular and respiratory disorders to neurodegeneration and cancer. CRITICAL ISSUES The target specificity of protein S-nitrosylation and the stability and reactivity of protein SNOs are determined substantially by enzymatic machinery comprising highly conserved transnitrosylases and denitrosylases. Understanding the differential functionality of SNO-regulatory enzymes is essential, and is amenable to genetic and pharmacological analyses, read out as perturbation of specific equilibria within the SNO circuitry. FUTURE DIRECTIONS The emerging picture of NO biology entails equilibria among potentially thousands of different SNOs, governed by denitrosylases and nitrosylases. Thus, to elucidate the operation and consequences of S-nitrosylation in cellular contexts, studies should consider the roles of SNO-proteins as both targets and transducers of S-nitrosylation, functioning according to enzymatically governed equilibria.
Collapse
Affiliation(s)
- Colin T Stomberski
- 1 Institute for Transformative Molecular Medicine, Case Western Reserve University, Cleveland, Ohio.,2 Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio
| | - Douglas T Hess
- 1 Institute for Transformative Molecular Medicine, Case Western Reserve University, Cleveland, Ohio.,3 Department of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Jonathan S Stamler
- 2 Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio.,3 Department of Medicine, Case Western Reserve University, Cleveland, Ohio.,4 Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| |
Collapse
|
18
|
Di Fiore JM, Dylag AM, Honomichl RD, Hibbs AM, Martin RJ, Tatsuoka C, Raffay TM. Early inspired oxygen and intermittent hypoxemic events in extremely premature infants are associated with asthma medication use at 2 years of age. J Perinatol 2019; 39:203-211. [PMID: 30367103 PMCID: PMC6351157 DOI: 10.1038/s41372-018-0264-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/07/2018] [Accepted: 10/08/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Extremely premature infants are at risk for childhood wheezing. Early respiratory support and intermittent hypoxemia (IH) events may be associated with adverse breathing outcomes. STUDY DESIGN A single-center retrospective cohort study of 137 premature infants <28 weeks gestational age characterized the associations of cumulative oxygen, cumulative mean airway pressure, IH, and oxygen saturation (SpO2) on the primary outcome of prescription asthma medication use at 2-year follow-up. Relative risk was calculated by generalized estimating equations. RESULTS Reported asthma medication use was 46%. At 1-3 days of age, elevated cumulative oxygen exposure, increased daily IH, and lower mean SpO2 (adjusted for gestational age and sex) and increased cumulative mean airway pressure exposure (unadjusted) were associated with asthma medication use. CONCLUSION Increased oxygen and frequent IH events during just the first 3 days of age may help identify extremely premature newborns at risk for symptomatic childhood wheezing requiring prescription asthma medications.
Collapse
Affiliation(s)
- Juliann M. Di Fiore
- Division of Neonatology, Rainbow Babies & Children’s Hospital, Case Western Reserve University, Cleveland, OH
| | - Andrew M. Dylag
- Division of Neonatology, Golisano Children’s Hospital, University of Rochester, Rochester, NY
| | - Ryan D. Honomichl
- Division of Neurology, University Hospitals of Cleveland, Case Western Reserve University, Cleveland, OH
| | - Anna Maria Hibbs
- Division of Neonatology, Rainbow Babies & Children’s Hospital, Case Western Reserve University, Cleveland, OH
| | - Richard J. Martin
- Division of Neonatology, Rainbow Babies & Children’s Hospital, Case Western Reserve University, Cleveland, OH
| | - Curtis Tatsuoka
- Division of Neurology, University Hospitals of Cleveland, Case Western Reserve University, Cleveland, OH
| | - Thomas M. Raffay
- Division of Neonatology, Rainbow Babies & Children’s Hospital, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
19
|
Turan I, Sayan Ozacmak H, Ozacmak VH, Barut F, Ozacmak ID. The effects of S-nitrosoglutathione on intestinal ischemia reperfusion injury and acute lung injury in rats: Roles of oxidative stress and NF-κB. Tissue Cell 2018; 52:35-41. [PMID: 29857826 DOI: 10.1016/j.tice.2018.03.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 03/16/2018] [Accepted: 03/24/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Intestinal ischemia and reperfusion (I/R) induces oxidative stress, inflammatory response, and acute lung injury. S-nitrosoglutathione (GSNO), a nitric oxide donor, has been documented to have protective effects on experimental ischemia models. AIM The aim of this study was to examine the effect of GSNO on I/R-induced intestine and lung damage and detect the potential mechanisms emphasizing the protective role of GSNO. METHODS Intestinal I/R was induced by occluding the superior mesenteric artery for 30 min followed by reperfusion for 180 min. GSNO was administered intravenously before reperfusion period (0.25 mg/kg). The levels of lipid peroxidation, reduced glutathione, and myeloperoxidase (MPO), histopathological evaluation and immunohistochemical expressions of both nuclear factor KappaB (NF-κB) and inducible nitric oxide (iNOS) in intestine and lung tissues were assessed. RESULTS Histolopathologic evaluation demonstrated that intestinal I/R induced severe damages in the intestine and the lung tissues. Histopathological scores decreased with GSNO treatment. GSNO treatment reduced lipid peroxidation and MPO levels and inhibited expression of NF-κB and iNOS in the intestine. CONCLUSION Our results suggest that GSNO treatment may ameliorate the intestinal and lung injury in rats, at least in part, by inhibiting inflammatory response and oxidative stress.
Collapse
Affiliation(s)
- Inci Turan
- Department of Physiology, Bulent Ecevit University Faculty of Medicine, Turkey.
| | - Hale Sayan Ozacmak
- Department of Physiology, Bulent Ecevit University Faculty of Medicine, Turkey
| | - V Haktan Ozacmak
- Department of Physiology, Bulent Ecevit University Faculty of Medicine, Turkey
| | - Figen Barut
- Department of Pathology, Bulent Ecevit University Faculty of Medicine, Turkey
| | - I Diler Ozacmak
- Or-Ahayim Private Balat Hospital, Department of General surgery, Bulent Ecevit University Faculty of Medicine, Turkey
| |
Collapse
|
20
|
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: 16] [Impact Index Per Article: 2.0] [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
|
21
|
Surate Solaligue DE, Rodríguez-Castillo JA, Ahlbrecht K, Morty RE. Recent advances in our understanding of the mechanisms of late lung development and bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2017; 313:L1101-L1153. [PMID: 28971976 DOI: 10.1152/ajplung.00343.2017] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/21/2017] [Accepted: 09/23/2017] [Indexed: 02/08/2023] Open
Abstract
The objective of lung development is to generate an organ of gas exchange that provides both a thin gas diffusion barrier and a large gas diffusion surface area, which concomitantly generates a steep gas diffusion concentration gradient. As such, the lung is perfectly structured to undertake the function of gas exchange: a large number of small alveoli provide extensive surface area within the limited volume of the lung, and a delicate alveolo-capillary barrier brings circulating blood into close proximity to the inspired air. Efficient movement of inspired air and circulating blood through the conducting airways and conducting vessels, respectively, generates steep oxygen and carbon dioxide concentration gradients across the alveolo-capillary barrier, providing ideal conditions for effective diffusion of both gases during breathing. The development of the gas exchange apparatus of the lung occurs during the second phase of lung development-namely, late lung development-which includes the canalicular, saccular, and alveolar stages of lung development. It is during these stages of lung development that preterm-born infants are delivered, when the lung is not yet competent for effective gas exchange. These infants may develop bronchopulmonary dysplasia (BPD), a syndrome complicated by disturbances to the development of the alveoli and the pulmonary vasculature. It is the objective of this review to update the reader about recent developments that further our understanding of the mechanisms of lung alveolarization and vascularization and the pathogenesis of BPD and other neonatal lung diseases that feature lung hypoplasia.
Collapse
Affiliation(s)
- David E Surate Solaligue
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| | - José Alberto Rodríguez-Castillo
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| | - Katrin Ahlbrecht
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| | - Rory E Morty
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and .,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| |
Collapse
|
22
|
Bodas M, Silverberg D, Walworth K, Brucia K, Vij N. Augmentation of S-Nitrosoglutathione Controls Cigarette Smoke-Induced Inflammatory-Oxidative Stress and Chronic Obstructive Pulmonary Disease-Emphysema Pathogenesis by Restoring Cystic Fibrosis Transmembrane Conductance Regulator Function. Antioxid Redox Signal 2017; 27:433-451. [PMID: 28006950 PMCID: PMC5564030 DOI: 10.1089/ars.2016.6895] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
AIMS Cigarette smoke (CS)-mediated acquired cystic fibrosis transmembrane conductance regulator (CFTR)-dysfunction, autophagy-impairment, and resulting inflammatory-oxidative/nitrosative stress leads to chronic obstructive pulmonary disease (COPD)-emphysema pathogenesis. Moreover, nitric oxide (NO) signaling regulates lung function decline, and low serum NO levels that correlates with COPD severity. Hence, we aim to evaluate here the effects and mechanism(s) of S-nitrosoglutathione (GSNO) augmentation in regulating inflammatory-oxidative stress and COPD-emphysema pathogenesis. RESULTS Our data shows that cystic fibrosis transmembrane conductance regulator (CFTR) colocalizes with aggresome bodies in the lungs of COPD subjects with increasing emphysema severity (Global Initiative for Chronic Obstructive Lung Disease [GOLD] I - IV) compared to nonemphysema controls (GOLD 0). We further demonstrate that treatment with GSNO or S-nitrosoglutathione reductase (GSNOR)-inhibitor (N6022) significantly inhibits cigarette smoke extract (CSE; 5%)-induced decrease in membrane CFTR expression by rescuing it from ubiquitin (Ub)-positive aggresome bodies (p < 0.05). Moreover, GSNO restoration significantly (p < 0.05) decreases CSE-induced reactive oxygen species (ROS) activation and autophagy impairment (decreased accumulation of ubiquitinated proteins in the insoluble protein fractions and restoration of autophagy flux). In addition, GSNO augmentation inhibits protein misfolding as CSE-induced colocalization of ubiquitinated proteins and LC3B (in autophagy bodies) is significantly reduced by GSNO/N6022 treatment. We verified using the preclinical COPD-emphysema murine model that chronic CS (Ch-CS)-induced inflammation (interleukin [IL]-6/IL-1β levels), aggresome formation (perinuclear coexpression/colocalization of ubiquitinated proteins [Ub] and p62 [impaired autophagy marker], and CFTR), oxidative/nitrosative stress (p-Nrf2, inducible nitric oxide synthase [iNOS], and 3-nitrotyrosine expression), apoptosis (caspase-3/7 activity), and alveolar airspace enlargement (Lm) are significantly (p < 0.05) alleviated by augmenting airway GSNO levels. As a proof of concept, we demonstrate that GSNO augmentation suppresses Ch-CS-induced perinuclear CFTR protein accumulation (p < 0.05), which restores both acquired CFTR dysfunction and autophagy impairment, seen in COPD-emphysema subjects. INNOVATION GSNO augmentation alleviates CS-induced acquired CFTR dysfunction and resulting autophagy impairment. CONCLUSION Overall, we found that augmenting GSNO levels controls COPD-emphysema pathogenesis by reducing CS-induced acquired CFTR dysfunction and resulting autophagy impairment and chronic inflammatory-oxidative stress. Antioxid. Redox Signal. 27, 433-451.
Collapse
Affiliation(s)
- Manish Bodas
- 1 College of Medicine, Central Michigan University , Mt. Pleasant, Michigan
| | - David Silverberg
- 1 College of Medicine, Central Michigan University , Mt. Pleasant, Michigan
| | - Kyla Walworth
- 1 College of Medicine, Central Michigan University , Mt. Pleasant, Michigan
| | - Kathryn Brucia
- 1 College of Medicine, Central Michigan University , Mt. Pleasant, Michigan
| | - Neeraj Vij
- 1 College of Medicine, Central Michigan University , Mt. Pleasant, Michigan.,2 Department of Pediatrics and Pulmonary Medicine, The Johns Hopkins University School of Medicine , Baltimore, Maryland
| |
Collapse
|
23
|
Sun BL, Palmer L, Alam SR, Adekoya I, Brown-Steinke K, Periasamy A, Mutus B. O-Aminobenzoyl-S-nitrosoglutathione: A fluorogenic, cell permeable, pseudo-substrate for S-nitrosoglutathione reductase. Free Radic Biol Med 2017; 108:445-451. [PMID: 28419866 DOI: 10.1016/j.freeradbiomed.2017.04.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 04/07/2017] [Accepted: 04/10/2017] [Indexed: 10/19/2022]
Abstract
S-nitrosoglutathione reductase (GSNOR) is a multifunctional enzyme. It can catalyze NADH-dependent reduction of S-nitrosoglutathione (GSNO); as well as NAD+-dependent oxidation of hydroxymethylglutathione (HMGSH; an adduct formed by the spontaneous reaction between formaldehyde and glutathione). While initially recognized as the enzyme that is involved in formaldehyde detoxification, increasing amount of evidence has shown that GSNOR also plays a significant role in nitric oxide mediated signaling through its modulation of protein S-nitrosothiol signaling. In humans, GSNOR/S-nitrosothiols have been implicated in the etiology of several diseases including lung cancer, cystic fibrosis, asthma, pulmonary hypertension, and neuronal dysfunction. Currently, it is not possible to monitor the activity of GSNOR in live cells. In this article, we present a new compound, O-aminobenzoyl-S-nitrosoglutathione (OAbz-GSNO), which acts as a fluorogenic pseudo-substrate for GSNOR with an estimated Km value of 320µM. The weak OAbz-GSNO fluorescence increases by approximately 14 fold upon reduction of its S-NO moiety. In live cell imaging studies, OAbz-GSNO is readily taken up by primary pulmonary endothelial cells and localizes to the same perinuclear region as GSNOR. The perinuclear OAbz-GSNO fluorescence increases in a time dependent manner and this increase in fluorescence is abolished by siRNA knockdown of GSNOR or by treatment with GSNOR-specific inhibitors N6022 and C3. Taken together, these data demonstrate that OAbz-GSNO can be used as a tool to monitor the activity of GSNOR in live cells.
Collapse
Affiliation(s)
- Bei Lei Sun
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario, Canada
| | - Lisa Palmer
- Department of Pediatrics, University of Virginia, Charlottesville, VA, USA
| | | | - Itunuoluwa Adekoya
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario, Canada
| | | | - Ammasi Periasamy
- W. M. Keck Center for Cellular Imaging, Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Bulent Mutus
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario, Canada.
| |
Collapse
|