1
|
Paulus MC, Drent M, Kouw IWK, Balvers MGJ, Bast A, van Zanten ARH. Vitamin K: a potential missing link in critical illness-a scoping review. Crit Care 2024; 28:212. [PMID: 38956732 PMCID: PMC11218309 DOI: 10.1186/s13054-024-05001-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Vitamin K is essential for numerous physiological processes, including coagulation, bone metabolism, tissue calcification, and antioxidant activity. Deficiency, prevalent in critically ill ICU patients, impacts coagulation and increases the risk of bleeding and other complications. This review aims to elucidate the metabolism of vitamin K in the context of critical illness and identify a potential therapeutic approach. METHODS In December 2023, a scoping review was conducted using the PRISMA Extension for Scoping Reviews. Literature was searched in PubMed, Embase, and Cochrane databases without restrictions. Inclusion criteria were studies on adult ICU patients discussing vitamin K deficiency and/or supplementation. RESULTS A total of 1712 articles were screened, and 13 met the inclusion criteria. Vitamin K deficiency in ICU patients is linked to malnutrition, impaired absorption, antibiotic use, increased turnover, and genetic factors. Observational studies show higher PIVKA-II levels in ICU patients, indicating reduced vitamin K status. Risk factors include inadequate intake, disrupted absorption, and increased physiological demands. Supplementation studies suggest vitamin K can improve status but not normalize it completely. Vitamin K deficiency may correlate with prolonged ICU stays, mechanical ventilation, and increased mortality. Factors such as genetic polymorphisms and disrupted microbiomes also contribute to deficiency, underscoring the need for individualized nutritional strategies and further research on optimal supplementation dosages and administration routes. CONCLUSIONS Addressing vitamin K deficiency in ICU patients is crucial for mitigating risks associated with critical illness, yet optimal management strategies require further investigation. IMPACT RESEARCH To the best of our knowledge, this review is the first to address the prevalence and progression of vitamin K deficiency in critically ill patients. It guides clinicians in diagnosing and managing vitamin K deficiency in intensive care and suggests practical strategies for supplementing vitamin K in critically ill patients. This review provides a comprehensive overview of the existing literature, and serves as a valuable resource for clinicians, researchers, and policymakers in critical care medicine.
Collapse
Affiliation(s)
- Michelle Carmen Paulus
- Department of Intensive Care Medicine & Research, Gelderse Vallei Hospital, Willy Brandtlaan 10, 6716 RP, Ede, The Netherlands
- Division of Human Nutrition and Health, Nutritional Biology, Wageningen University & Research, HELIX (Building 124), Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Marjolein Drent
- Department of Pharmacology and Toxicology, Faculty of Health, Medicine, and Life Science, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, The Netherlands
- Interstitial Lung Diseases (ILD) Center of Excellence, St. Antonius Hospital, Nieuwegein, Koekoekslaan 1, 3435 CM, Nieuwegein, The Netherlands
- ILD Care Foundation Research Team, Heideoordlaan 8, 6711NR, Ede, The Netherlands
| | - Imre Willemijn Kehinde Kouw
- Department of Intensive Care Medicine & Research, Gelderse Vallei Hospital, Willy Brandtlaan 10, 6716 RP, Ede, The Netherlands
- Division of Human Nutrition and Health, Nutritional Biology, Wageningen University & Research, HELIX (Building 124), Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Michiel Gerard Juliaan Balvers
- Division of Human Nutrition and Health, Nutritional Biology, Wageningen University & Research, HELIX (Building 124), Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Aalt Bast
- Department of Pharmacology and Toxicology, Faculty of Health, Medicine, and Life Science, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, The Netherlands
- ILD Care Foundation Research Team, Heideoordlaan 8, 6711NR, Ede, The Netherlands
| | - Arthur Raymond Hubert van Zanten
- Department of Intensive Care Medicine & Research, Gelderse Vallei Hospital, Willy Brandtlaan 10, 6716 RP, Ede, The Netherlands.
- Division of Human Nutrition and Health, Nutritional Biology, Wageningen University & Research, HELIX (Building 124), Stippeneng 4, 6708 WE, Wageningen, The Netherlands.
| |
Collapse
|
2
|
Hu Y, Huang Y, Zong L, Lin J, Liu X, Ning S. Emerging roles of ferroptosis in pulmonary fibrosis: current perspectives, opportunities and challenges. Cell Death Discov 2024; 10:301. [PMID: 38914560 PMCID: PMC11196712 DOI: 10.1038/s41420-024-02078-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/26/2024] Open
Abstract
Pulmonary fibrosis (PF) is a chronic interstitial lung disorder characterized by abnormal myofibroblast activation, accumulation of extracellular matrix (ECM), and thickening of fibrotic alveolar walls, resulting in deteriorated lung function. PF is initiated by dysregulated wound healing processes triggered by factors such as excessive inflammation, oxidative stress, and coronavirus disease (COVID-19). Despite advancements in understanding the disease's pathogenesis, effective preventive and therapeutic interventions are currently lacking. Ferroptosis, an iron-dependent regulated cell death (RCD) mechanism involving lipid peroxidation and glutathione (GSH) depletion, exhibits unique features distinct from other RCD forms (e.g., apoptosis, necrosis, and pyroptosis). Imbalance between reactive oxygen species (ROS) production and detoxification leads to ferroptosis, causing cellular dysfunction through lipid peroxidation, protein modifications, and DNA damage. Emerging evidence points to the crucial role of ferroptosis in PF progression, driving macrophage polarization, fibroblast proliferation, and ECM deposition, ultimately contributing to alveolar cell death and lung tissue scarring. This review provides a comprehensive overview of the latest findings on the involvement and signaling mechanisms of ferroptosis in PF pathogenesis, emphasizing potential novel anti-fibrotic therapeutic approaches targeting ferroptosis for PF management.
Collapse
Affiliation(s)
- Yixiang Hu
- Department of Clinical Pharmacy, The Affiliated Xiangtan Center Hospital of Hunan University, Xiangtan, 411100, China
| | - Ying Huang
- Zhongshan Hospital of Traditional Chinese Medicine Afflilated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, China
| | - Lijuan Zong
- Department of Rehabilitation Medicine, Zhongda Hospital of Southeast University, Nanjing, 210096, China
| | - Jiaxin Lin
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China
| | - Xiang Liu
- Department of Clinical Pharmacy, The Affiliated Xiangtan Center Hospital of Hunan University, Xiangtan, 411100, China.
| | - Shipeng Ning
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China.
| |
Collapse
|
3
|
Silva FDD, Galiciolli MEDA, Irioda AC, Oliveira CS, Piccoli BC, Prestes ADS, Borin BC, Schuch AP, Ochoa-Rodríguez E, Nuñez-Figueredo Y, Rocha JBTD. Investigation of the cytotoxicity, genotoxicity and antioxidant prospects of JM-20 on human blood cells: A multi-target compound with potential therapeutic applications. Blood Cells Mol Dis 2024; 106:102827. [PMID: 38301450 DOI: 10.1016/j.bcmd.2024.102827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 01/10/2024] [Accepted: 01/16/2024] [Indexed: 02/03/2024]
Abstract
JM-20 is a 1,5-benzodiazepine compound fused to a dihydropyridine fraction with different pharmacological properties. However, its potential toxic effects on blood cells have not yet been reported. Thus, the present study aimed to investigate, for the first time, the possible cytotoxicity of JM-20 through cell viability, cell cycle, morphology changes, reactive species (RS) to DCFH-DA, and lipid peroxidation in human leukocytes, its hemolytic effect on human erythrocytes, and its potential DNA genotoxicity using plasmid DNA in vitro. Furthermore, the compound's ability to reduce the DPPH radical was also measured. Human blood was obtained from healthy volunteers (30 ± 10 years old), and the leukocytes or erythrocytes were immediately isolated and treated with different concentrations of JM-20. A cytoprotective effect was exhibited by 10 μM JM-20 against 1 mM tert-butyl hydroperoxide (t-but-OOH) in the leukocytes. However, the highest tested concentrations of the compound (20 and 50 μM) changed the morphology and caused a significant decrease in the cell viability of leukocytes (p < 0.05, in comparison with Control). All tested concentrations of JM-20 also resulted in a significant increase in intracellular RS as measured by DCFH-DA in these cells (p < 0.05, in comparison with Control). On the other hand, the results point out a potent antioxidant effect of JM-20, which was similar to the classical antioxidant α-tocopherol. The IC50 value of JM-20 against the lipid peroxidation induced by (FeII) was 1.051 μM ± 0.21, while the IC50 value of α-tocopherol in this parameter was 1.065 μM ± 0.34. Additionally, 50 and 100 μM JM-20 reduced the DPPH radical in a statistically similar way to the 100 μM α-tocopherol (p < 0.05, in comparison with the control). No significant hemolysis in erythrocytes, no cell cycle changes in leukocytes, and no genotoxic effects in plasmid DNA were induced by JM-20 at any tested concentration. The in silico pharmacokinetic and toxicological properties of JM-20, derivatives, and nifedipine were also studied. Here, our findings demonstrate that JM-20 and its putative metabolites exhibit similar characteristics to nifedipine, and the in vitro and in silico data support the low toxicity of JM-20 to mammals.
Collapse
Affiliation(s)
- Fernanda D'Avila da Silva
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | - Maria Eduarda de Andrade Galiciolli
- Programa de Pós-Graduação Stricto Sensu em Biotecnologia Aplicada a Saúde da Criança e do Adolescente, Instituto de Pesquisa Pelé Pequeno Príncipe, Rua Silva Jardim, 1632 Curitiba, Paraná, Brazil; Faculdade Pequeno Príncipe, Avenida Iguaçu, 333 Curitiba, Paraná, Brazil
| | - Ana Carolina Irioda
- Programa de Pós-Graduação Stricto Sensu em Biotecnologia Aplicada a Saúde da Criança e do Adolescente, Instituto de Pesquisa Pelé Pequeno Príncipe, Rua Silva Jardim, 1632 Curitiba, Paraná, Brazil; Faculdade Pequeno Príncipe, Avenida Iguaçu, 333 Curitiba, Paraná, Brazil
| | - Cláudia Sirlene Oliveira
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil; Programa de Pós-Graduação Stricto Sensu em Biotecnologia Aplicada a Saúde da Criança e do Adolescente, Instituto de Pesquisa Pelé Pequeno Príncipe, Rua Silva Jardim, 1632 Curitiba, Paraná, Brazil; Faculdade Pequeno Príncipe, Avenida Iguaçu, 333 Curitiba, Paraná, Brazil
| | - Bruna Candia Piccoli
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | - Alessandro de Souza Prestes
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | - Bruna Cogo Borin
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | - Andre Passaglia Schuch
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | - Estael Ochoa-Rodríguez
- Centro de Investigación y Desarrollo de Medicamentos, Ave 26, N° 1605,e /Boyeros y Puentes Grandes, CP10600 La Habana, Cuba
| | - Yanier Nuñez-Figueredo
- Centro de Investigación y Desarrollo de Medicamentos, Ave 26, N° 1605,e /Boyeros y Puentes Grandes, CP10600 La Habana, Cuba
| | - João Batista Teixeira da Rocha
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil.
| |
Collapse
|
4
|
Kim NH, Kim HY, Lee JH, Chang I, Heo SH, Kim J, Kim JH, Kang JH, Lee SW. Superoxide dismutase secreting Bacillus amyloliquefaciens spores attenuate pulmonary fibrosis. Biomed Pharmacother 2023; 168:115647. [PMID: 37826939 DOI: 10.1016/j.biopha.2023.115647] [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: 07/15/2023] [Revised: 09/18/2023] [Accepted: 10/03/2023] [Indexed: 10/14/2023] Open
Abstract
Superoxide dismutase (SOD) can convert active oxygen to oxygen or hydrogen peroxide, and recent research has suggested that it can protect against lung damage and fibrosis. Clinical applications based on SOD remain limited however due to costs and low stability. We here investigated a potential new therapeutic delivery system for this enzyme in the form of SOD-overexpressing Bacillus amyloliquefaciens spores which we introduced into a bleomycin-induced pulmonary fibrosis mouse model. This treatment significantly alleviated the disease, as quantified using a hydroxyproline assay, at 107 colony forming unit (CFU) of Bacillus spores per day. Exposure of the mice to the spores was further found to decrease the lung mRNA levels of CTGF, Col1a1, α-SMA, TGF-β, TNF-α, and IL-6, and the protein levels of TGF-β, Smad2/3, αSMA and Col1a1, all major indicators of pulmonary fibrosis. Survival benefits, and reduced byproducts of lipid peroxidase such as malondialdehyde and 4-hydroxynen, were also noted in the treated animals. The beneficial effects of these Bacillus spores on pulmonary fibrosis were further found to be greater than the equivalent free SOD concentration. Immunofluorescence staining of primary pulmonary fibroblasts extracted from the bleomycin-induced model showed decreased αSMA expression following the in vivo treatment with SOD-overexpressing Bacillus. Our treatment approach SOD through Bacillus spores shows beneficial effects against pulmonary fibrosis, combined with the suppression of the SMAD/TGF-β pathway, suggesting that it is an effective novel delivery route for antioxidants.
Collapse
Affiliation(s)
- Na Hyun Kim
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hee Young Kim
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; BiomLogic, Inc., Seoul, Republic of Korea
| | - Jang Ho Lee
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Inik Chang
- BiomLogic, Inc., Seoul, Republic of Korea
| | - Sun-Hee Heo
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jiseon Kim
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Department of Pharmacology and Regnerative Medicine, University of Illinois College of Medicine, Chicago, USA
| | | | | | - Sei Won Lee
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
5
|
Wallis TJM, Minnion M, Freeman A, Bates A, Otto JM, Wootton SA, Fletcher SV, Grocott MPW, Feelisch M, Jones MG, Jack S. Individualised Exercise Training Enhances Antioxidant Buffering Capacity in Idiopathic Pulmonary Fibrosis. Antioxidants (Basel) 2023; 12:1645. [PMID: 37627640 PMCID: PMC10451244 DOI: 10.3390/antiox12081645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/13/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Exercise training is recommended for patients with idiopathic pulmonary fibrosis (IPF); however, the mechanism(s) underlying its physiological benefits remain unclear. We investigated the effects of an individualised aerobic interval training programme on exercise capacity and redox status in IPF patients. IPF patients were recruited prospectively to an 8-week, twice-weekly cardiopulmonary exercise test (CPET)-derived structured responsive exercise training programme (SRETP). Systemic redox status was assessed pre- and post-CPET at baseline and following SRETP completion. An age- and sex-matched non-IPF control cohort was recruited for baseline comparison only. At baseline, IPF patients (n = 15) had evidence of increased oxidative stress compared with the controls as judged by; the plasma reduced/oxidised glutathione ratio (median, control 1856 vs. IPF 736 p = 0.046). Eleven IPF patients completed the SRETP (median adherence 88%). Following SRETP completion, there was a significant improvement in exercise capacity assessed via the constant work-rate endurance time (+82%, p = 0.003). This was accompanied by an improvement in post-exercise redox status (in favour of antioxidants) assessed via serum total free thiols (median increase, +0.26 μmol/g protein p = 0.005) and total glutathione concentration (+0.73 μM p = 0.03), as well as a decrease in post-exercise lipid peroxidation products (-1.20 μM p = 0.02). Following SRETP completion, post-exercise circulating nitrite concentrations were significantly lower compared with baseline (-0.39 μM p = 0.04), suggestive of exercise-induced nitrite utilisation. The SRETP increased both endurance time and systemic antioxidant capacity in IPF patients. The observed reduction in nitrite concentrations provides a mechanistic rationale to investigate nitrite/nitrate supplementation in IPF patients.
Collapse
Affiliation(s)
- Tim J. M. Wallis
- NIHR Southampton Biomedical Research Centre, Respiratory and Critical Care, University Hospital Southampton, Southampton SO16 6YD, UK; (M.M.); (A.F.); (A.B.); (J.M.O.); (S.V.F.); (M.P.W.G.); (M.F.); (M.G.J.); (S.J.)
- Academic School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK;
| | - Magdalena Minnion
- NIHR Southampton Biomedical Research Centre, Respiratory and Critical Care, University Hospital Southampton, Southampton SO16 6YD, UK; (M.M.); (A.F.); (A.B.); (J.M.O.); (S.V.F.); (M.P.W.G.); (M.F.); (M.G.J.); (S.J.)
- Academic School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK;
| | - Anna Freeman
- NIHR Southampton Biomedical Research Centre, Respiratory and Critical Care, University Hospital Southampton, Southampton SO16 6YD, UK; (M.M.); (A.F.); (A.B.); (J.M.O.); (S.V.F.); (M.P.W.G.); (M.F.); (M.G.J.); (S.J.)
- Academic School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK;
| | - Andrew Bates
- NIHR Southampton Biomedical Research Centre, Respiratory and Critical Care, University Hospital Southampton, Southampton SO16 6YD, UK; (M.M.); (A.F.); (A.B.); (J.M.O.); (S.V.F.); (M.P.W.G.); (M.F.); (M.G.J.); (S.J.)
- Academic School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK;
- Department of Critical Care and Anaesthesia, University Hospital Southampton, Southampton SO16 6YD, UK
| | - James M. Otto
- NIHR Southampton Biomedical Research Centre, Respiratory and Critical Care, University Hospital Southampton, Southampton SO16 6YD, UK; (M.M.); (A.F.); (A.B.); (J.M.O.); (S.V.F.); (M.P.W.G.); (M.F.); (M.G.J.); (S.J.)
- Academic School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK;
- Department of Critical Care and Anaesthesia, University Hospital Southampton, Southampton SO16 6YD, UK
| | - Stephen A. Wootton
- Academic School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK;
- NIHR Southampton Biomedical Research Centre, Nutrition and Metabolism, University Hospital Southampton, Southampton SO16 6YD, UK
| | - Sophie V. Fletcher
- NIHR Southampton Biomedical Research Centre, Respiratory and Critical Care, University Hospital Southampton, Southampton SO16 6YD, UK; (M.M.); (A.F.); (A.B.); (J.M.O.); (S.V.F.); (M.P.W.G.); (M.F.); (M.G.J.); (S.J.)
- Academic School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK;
| | - Michael P. W. Grocott
- NIHR Southampton Biomedical Research Centre, Respiratory and Critical Care, University Hospital Southampton, Southampton SO16 6YD, UK; (M.M.); (A.F.); (A.B.); (J.M.O.); (S.V.F.); (M.P.W.G.); (M.F.); (M.G.J.); (S.J.)
- Academic School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK;
- Department of Critical Care and Anaesthesia, University Hospital Southampton, Southampton SO16 6YD, UK
| | - Martin Feelisch
- NIHR Southampton Biomedical Research Centre, Respiratory and Critical Care, University Hospital Southampton, Southampton SO16 6YD, UK; (M.M.); (A.F.); (A.B.); (J.M.O.); (S.V.F.); (M.P.W.G.); (M.F.); (M.G.J.); (S.J.)
- Academic School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK;
| | - Mark G. Jones
- NIHR Southampton Biomedical Research Centre, Respiratory and Critical Care, University Hospital Southampton, Southampton SO16 6YD, UK; (M.M.); (A.F.); (A.B.); (J.M.O.); (S.V.F.); (M.P.W.G.); (M.F.); (M.G.J.); (S.J.)
- Academic School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK;
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Sandy Jack
- NIHR Southampton Biomedical Research Centre, Respiratory and Critical Care, University Hospital Southampton, Southampton SO16 6YD, UK; (M.M.); (A.F.); (A.B.); (J.M.O.); (S.V.F.); (M.P.W.G.); (M.F.); (M.G.J.); (S.J.)
- Academic School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK;
- Department of Critical Care and Anaesthesia, University Hospital Southampton, Southampton SO16 6YD, UK
| |
Collapse
|
6
|
Douglas D, Keating L, Strykowski R, Lee CT, Garcia N, Selvan K, Kaushik N, Bauer Ventura I, Jablonski R, Vij R, Chung JH, Bellam S, Strek ME, Adegunsoye A. Tobacco smoking is associated with combined pulmonary fibrosis and emphysema and worse outcomes in interstitial lung disease. Am J Physiol Lung Cell Mol Physiol 2023; 325:L233-L243. [PMID: 37366539 PMCID: PMC10396279 DOI: 10.1152/ajplung.00083.2023] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 06/28/2023] Open
Abstract
Tobacco smoking is an established cause of pulmonary disease whose contribution to interstitial lung disease (ILD) is incompletely characterized. We hypothesized that compared with nonsmokers, subjects who smoked tobacco would differ in their clinical phenotype and have greater mortality. We performed a retrospective cohort study of tobacco smoking in ILD. We evaluated demographic and clinical characteristics, time to clinically meaningful lung function decline (LFD), and mortality in patients stratified by tobacco smoking status (ever vs. never) within a tertiary center ILD registry (2006-2021) and replicated mortality outcomes across four nontertiary medical centers. Data were analyzed by two-sided t tests, Poisson generalized linear models, and Cox proportional hazard models adjusted for age, sex, forced vital capacity (FVC), diffusion capacity of the lung for carbon monoxide (DLCO), ILD subtype, antifibrotic therapy, and hospital center. Of 1,163 study participants, 651 were tobacco smokers. Smokers were more likely to be older, male, have idiopathic pulmonary fibrosis (IPF), coronary artery disease, CT honeycombing and emphysema, higher FVC, and lower DLCO than nonsmokers (P < 0.01). Time to LFD in smokers was shorter (19.7 ± 20 mo vs. 24.8 ± 29 mo; P = 0.038) and survival time was decreased [10.75 (10.08-11.50) yr vs. 20 (18.67-21.25) yr; adjusted mortality HR = 1.50, 95%CI 1.17-1.92; P < 0.0001] compared with nonsmokers. Smokers had 12% greater odds of death for every additional 10 pack yr of smoking (P < 0.0001). Mortality outcomes remained consistent in the nontertiary cohort (HR = 1.51, 95%CI = 1.03-2.23; P = 0.036). Tobacco smokers with ILD have a distinct clinical phenotype strongly associated with the syndrome of combined PF and emphysema, shorter time to LFD, and decreased survival. Smoking prevention may improve ILD outcomes.NEW & NOTEWORTHY Smoking in ILD is associated with combined pulmonary fibrosis and emphysema and worse clinical outcomes.
Collapse
Affiliation(s)
- Dylan Douglas
- Section of Pulmonary and Critical Care, Department of Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Layne Keating
- Section of Pulmonary and Critical Care, Department of Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Rachel Strykowski
- Section of Pulmonary and Critical Care, Department of Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Cathryn T Lee
- Section of Pulmonary and Critical Care, Department of Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Nicole Garcia
- Section of Pulmonary and Critical Care, Department of Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Kavitha Selvan
- Section of Pulmonary and Critical Care, Department of Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Neha Kaushik
- Division of Pulmonary and Critical Care, Department of Medicine, NorthShore University HealthSystem, Evanston, Illinois, United States
| | - Iazsmin Bauer Ventura
- Section of Pulmonary and Critical Care, Department of Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Renea Jablonski
- Section of Pulmonary and Critical Care, Department of Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Rekha Vij
- Section of Pulmonary and Critical Care, Department of Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Jonathan H Chung
- Department of Radiology, The University of Chicago, Chicago, Illinois, United States
| | - Shashi Bellam
- Division of Pulmonary and Critical Care, Department of Medicine, NorthShore University HealthSystem, Evanston, Illinois, United States
| | - Mary E Strek
- Section of Pulmonary and Critical Care, Department of Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Ayodeji Adegunsoye
- Section of Pulmonary and Critical Care, Department of Medicine, The University of Chicago, Chicago, Illinois, United States
- Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, Chicago, Illinois, United States
| |
Collapse
|
7
|
Rajesh R, Atallah R, Bärnthaler T. Dysregulation of metabolic pathways in pulmonary fibrosis. Pharmacol Ther 2023; 246:108436. [PMID: 37150402 DOI: 10.1016/j.pharmthera.2023.108436] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/28/2023] [Accepted: 05/03/2023] [Indexed: 05/09/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic progressive disorder of unknown origin and the most common interstitial lung disease. It progresses with the recruitment of fibroblasts and myofibroblasts that contribute to the accumulation of extracellular matrix (ECM) proteins, leading to the loss of compliance and alveolar integrity, compromising the gas exchange capacity of the lung. Moreover, while there are therapeutics available, they do not offer a cure. Thus, there is a pressing need to identify better therapeutic targets. With the advent of transcriptomics, proteomics, and metabolomics, the cellular mechanisms underlying disease progression are better understood. Metabolic homeostasis is one such factor and its dysregulation has been shown to impact the outcome of IPF. Several metabolic pathways involved in the metabolism of lipids, protein and carbohydrates have been implicated in IPF. While metabolites are crucial for the generation of energy, it is now appreciated that metabolites have several non-metabolic roles in regulating cellular processes such as proliferation, signaling, and death among several other functions. Through this review, we succinctly elucidate the role of several metabolic pathways in IPF. Moreover, we also discuss potential therapeutics which target metabolism or metabolic pathways.
Collapse
Affiliation(s)
- Rishi Rajesh
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Reham Atallah
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Thomas Bärnthaler
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria.
| |
Collapse
|
8
|
Wang H, Nie J, Li P, Zhang X, Wang Y, Zhang W, Zhang W, Tang B. Exploring Idiopathic Pulmonary Fibrosis Biomarker by Simultaneous Two-Photon Fluorescence Imaging of Cysteine and Peroxynitrite. Anal Chem 2022; 94:11272-11281. [PMID: 35924865 DOI: 10.1021/acs.analchem.2c01866] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) has been characterized as a chronic inflammatory disease that leads to irreversible damage to pulmonary function. However, there is no specific IPF biomarker that can be used to distinguish IPF and not pneumonia. Endoplasmic reticulum (ER) stress is prominent in IPF. To search for a specific biomarker of IPF, we developed two ER-targeting two-photon (TP) fluorescent probes, TPER-ONOO and TPER-Cys, for peroxynitrite (ONOO-) and cysteine (Cys) imaging, respectively. A significant increase in Cys levels in the lungs was discovered only in mice with IPF, which implied that Cys might be an IPF biomarker candidate. Furthermore, we uncovered the mechanism of glutathione (GSH) deficiency in IPF, which was not due to Cys shortage but instead was attributable to impaired glutamate cysteine ligase and glutathione synthetase activities via ONOO--induced post-transcriptional modification. This work has potential to provide a new method for IPF early diagnosis and drug efficacy evaluation.
Collapse
Affiliation(s)
- Hui Wang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University, Jinan 250014, People's Republic of China
| | - Junwei Nie
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University, Jinan 250014, People's Republic of China
| | - Ping Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University, Jinan 250014, People's Republic of China
| | - Xiaoting Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University, Jinan 250014, People's Republic of China
| | - Yu Wang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University, Jinan 250014, People's Republic of China
| | - Wei Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University, Jinan 250014, People's Republic of China
| | - Wen Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University, Jinan 250014, People's Republic of China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University, Jinan 250014, People's Republic of China
| |
Collapse
|
9
|
Ritzenthaler JD, Torres-Gonzalez E, Zheng Y, Zelko IN, van Berkel V, Nunley DR, Kidane B, Halayko AJ, Summer R, Watson WH, Roman J. The profibrotic and senescence phenotype of old lung fibroblasts is reversed or ameliorated by genetic and pharmacological manipulation of Slc7a11 expression. Am J Physiol Lung Cell Mol Physiol 2022; 322:L449-L461. [PMID: 34984918 PMCID: PMC8917919 DOI: 10.1152/ajplung.00593.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Increased senescence and expression of profibrotic genes in old lung fibroblasts contribute to disrepair responses. We reported that primary lung fibroblasts from old mice have lower expression and activity of the cystine transporter Slc7a11/xCT than cells from young mice, resulting in changes in both the intracellular and extracellular redox environments. This study examines the hypothesis that low Slc7a11 expression in old lung fibroblasts promotes senescence and profibrotic gene expression. The levels of mRNA and protein of Slc7a11, senescence markers, and profibrotic genes were measured in primary fibroblasts from the lungs of old (24 mo) and young (3 mo) mice. In addition, the effects of genetic and pharmacological manipulation of Slc7a11 were investigated. We found that decreased expression of Slc7a11 in old cells was associated with elevated markers of senescence (p21, p16, p53, and β-galactosidase) and increased expression of profibrotic genes (Tgfb1, Smad3, Acta2, Fn1, Col1a1, and Col5a1). Silencing of Slc7a11 in young cells replicated the aging phenotype, whereas overexpression of Slc7a11 in old cells decreased expression of senescence and profibrotic genes. Young cells were induced to express the senescence and profibrotic phenotype by sulfasalazine, a Slc7a11 inhibitor, whereas treatment of old cells with sulforaphane, a Slc7a11 inducer, decreased senescence without affecting profibrotic genes. Like aging cells, idiopathic pulmonary fibrosis fibroblasts show decreased Slc7a11 expression and increased profibrotic markers. In short, old lung fibroblasts manifest a profibrotic and senescence phenotype that is modulated by genetic or pharmacological manipulation of Slc7a11.
Collapse
Affiliation(s)
- Jeffrey D. Ritzenthaler
- 1Division of Pulmonary, Allergy & Critical Care, Department of
Medicine, Center for Translational Medicine, The Jane & Leonard Korman Respiratory Institute, Philadelphia, Pennsylvania
| | - Edilson Torres-Gonzalez
- 1Division of Pulmonary, Allergy & Critical Care, Department of
Medicine, Center for Translational Medicine, The Jane & Leonard Korman Respiratory Institute, Philadelphia, Pennsylvania
| | - Yuxuan Zheng
- 2Department of Pharmacology & Toxicology, University of Louisville, Louisville, Kentucky
| | - Igor N. Zelko
- 3Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Victor van Berkel
- 4Department of Thoracic Surgery, Lung Transplantation Program, University of Louisville, Louisville, Kentucky
| | - David R. Nunley
- 5Department of Medicine, Lung Transplantation Program, Ohio State University, Columbus, Ohio
| | - Biniam Kidane
- 6Section of Thoracic Surgery, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Andrew J. Halayko
- 7Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ross Summer
- 1Division of Pulmonary, Allergy & Critical Care, Department of
Medicine, Center for Translational Medicine, The Jane & Leonard Korman Respiratory Institute, Philadelphia, Pennsylvania
| | - Walter H. Watson
- 2Department of Pharmacology & Toxicology, University of Louisville, Louisville, Kentucky,8Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Jesse Roman
- 1Division of Pulmonary, Allergy & Critical Care, Department of
Medicine, Center for Translational Medicine, The Jane & Leonard Korman Respiratory Institute, Philadelphia, Pennsylvania
| |
Collapse
|
10
|
Estornut C, Milara J, Bayarri MA, Belhadj N, Cortijo J. Targeting Oxidative Stress as a Therapeutic Approach for Idiopathic Pulmonary Fibrosis. Front Pharmacol 2022; 12:794997. [PMID: 35126133 PMCID: PMC8815729 DOI: 10.3389/fphar.2021.794997] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/10/2021] [Indexed: 01/19/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic interstitial lung disease characterized by an abnormal reepithelialisation, an excessive tissue remodelling and a progressive fibrosis within the alveolar wall that are not due to infection or cancer. Oxidative stress has been proposed as a key molecular process in pulmonary fibrosis development and different components of the redox system are altered in the cellular actors participating in lung fibrosis. To this respect, several activators of the antioxidant machinery and inhibitors of the oxidant species and pathways have been assayed in preclinical in vitro and in vivo models and in different clinical trials. This review discusses the role of oxidative stress in the development and progression of IPF and its underlying mechanisms as well as the evidence of oxidative stress in human IPF. Finally, we analyze the mechanism of action, the efficacy and the current status of different drugs developed to inhibit the oxidative stress as anti-fibrotic therapy in IPF.
Collapse
Affiliation(s)
- Cristina Estornut
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
- *Correspondence: Cristina Estornut, ; Javier Milara,
| | - Javier Milara
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
- Pharmacy Unit, University General Hospital Consortium, Valencia, Spain
- CIBERES, Health Institute Carlos III, Valencia, Spain
- *Correspondence: Cristina Estornut, ; Javier Milara,
| | - María Amparo Bayarri
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Nada Belhadj
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Julio Cortijo
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
- Pharmacy Unit, University General Hospital Consortium, Valencia, Spain
- CIBERES, Health Institute Carlos III, Valencia, Spain
- Research and Teaching Unit, University General Hospital Consortium, Valencia, Spain
| |
Collapse
|
11
|
Yu S, Jia J, Zheng J, Zhou Y, Jia D, Wang J. Recent Progress of Ferroptosis in Lung Diseases. Front Cell Dev Biol 2021; 9:789517. [PMID: 34869391 PMCID: PMC8635032 DOI: 10.3389/fcell.2021.789517] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 10/31/2021] [Indexed: 01/08/2023] Open
Abstract
Ferroptosis is a new form of programmed cell death due to iron-dependent excess accumulation of lipid peroxides and differs from other programmed cell deaths in morphological and biochemical characteristics. The process of ferroptosis is precisely regulated by iron metabolism, lipid metabolism, amino acid metabolism, and numerous signaling pathways, and plays a complex role in many pathophysiological processes. Recent studies have found that ferroptosis is closely associated with the development and progression of many lung diseases, including acute lung injury, pulmonary ischemia-reperfusion injury, lung cancer, chronic obstructive pulmonary disease, and pulmonary fibrosis. Here, we present a review of the main regulatory mechanisms of ferroptosis and its research progress in the pathogenesis and treatment of lung diseases, with the aim of providing new ideas for basic and clinical research of lung-related diseases.
Collapse
Affiliation(s)
- Shangjiang Yu
- Department of Clinical Medicine, Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jinqiu Jia
- Department of Pediatrics, Taizhou Women and Children's Hospital of Wenzhou Medical University, Taizhou, China
| | - Jinyu Zheng
- Department of Clinical Medicine, Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yiyang Zhou
- Department of Clinical Medicine, Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Danyun Jia
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Junlu Wang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
12
|
Audousset C, McGovern T, Martin JG. Role of Nrf2 in Disease: Novel Molecular Mechanisms and Therapeutic Approaches - Pulmonary Disease/Asthma. Front Physiol 2021; 12:727806. [PMID: 34658913 PMCID: PMC8511424 DOI: 10.3389/fphys.2021.727806] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 09/02/2021] [Indexed: 12/14/2022] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a major transcription factor involved in redox homeostasis and in the response induced by oxidative injury. Nrf2 is present in an inactive state in the cytoplasm of cells. Its activation by internal or external stimuli, such as infections or pollution, leads to the transcription of more than 500 elements through its binding to the antioxidant response element. The lungs are particularly susceptible to factors that generate oxidative stress such as infections, allergens and hyperoxia. Nrf2 has a crucial protective role against these ROS. Oxidative stress and subsequent activation of Nrf2 have been demonstrated in many human respiratory diseases affecting the airways, including asthma and chronic obstructive pulmonary disease (COPD), or the pulmonary parenchyma such as acute respiratory distress syndrome (ARDS) and pulmonary fibrosis. Several compounds, both naturally occurring and synthetic, have been identified as Nrf2 inducers and enhance the activation of Nrf2 and expression of Nrf2-dependent genes. These inducers have proven particularly effective at reducing the severity of the oxidative stress-driven lung injury in various animal models. In humans, these compounds offer promise as potential therapeutic strategies for the management of respiratory pathologies associated with oxidative stress but there is thus far little evidence of efficacy through human trials. The purpose of this review is to summarize the involvement of Nrf2 and its inducers in ARDS, COPD, asthma and lung fibrosis in both human and in experimental models.
Collapse
Affiliation(s)
- Camille Audousset
- Meakins-Christie Laboratories, McGill University, Montréal, QC, Canada
| | - Toby McGovern
- Meakins-Christie Laboratories, McGill University, Montréal, QC, Canada
| | - James G Martin
- Meakins-Christie Laboratories, McGill University, Montréal, QC, Canada
| |
Collapse
|
13
|
Taurine and vitamin E protect against pulmonary toxicity in rats exposed to cigarette smoke. SCIENTIFIC AFRICAN 2021. [DOI: 10.1016/j.sciaf.2021.e00923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
14
|
Kellogg DL, Kellogg DL, Musi N, Nambiar AM. Cellular Senescence in Idiopathic Pulmonary Fibrosis. CURRENT MOLECULAR BIOLOGY REPORTS 2021; 7:31-40. [PMID: 34401216 PMCID: PMC8358258 DOI: 10.1007/s40610-021-00145-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 07/07/2021] [Indexed: 12/28/2022]
Abstract
Cellular senescence (CS) is increasingly implicated in the etiology of age-related diseases. While CS can facilitate physiological processes such as tissue repair and wound healing, senescent cells also contribute to pathophysiological processes involving macromolecular damage and metabolic dysregulation that characterize multiple morbid and prevalent diseases, including Alzheimer's disease, osteoarthritis, atherosclerotic vascular disease, diabetes mellitus, and idiopathic pulmonary fibrosis (IPF). Preclinical studies targeting senescent cells and the senescence-associated secretory phenotype (SASP) with "senotherapeutics" have demonstrated improvement in age-related morbidity associated with these disease states. Despite promising results from these preclinical trials, few human clinical trials have been conducted. A first-in-human, open-label, pilot study of the senolytic combination of dasatinib and quercetin (DQ) in patients with IPF showed improved physical function and mobility. In this review, we will discuss our current understanding of cellular senescence, its role in age-associated diseases, with a specific focus on IPF, and potential for senotherapeutics in the treatment of fibrotic lung diseases.
Collapse
Affiliation(s)
- D L Kellogg
- University of Texas Health San Antonio, San Antonio, USA
| | - D L Kellogg
- University of Texas Health San Antonio, San Antonio, USA
- South Texas Veterans Health Care System, San Antonio, TX USA
| | - N Musi
- University of Texas Health San Antonio, San Antonio, USA
- South Texas Veterans Health Care System, San Antonio, TX USA
| | - A M Nambiar
- University of Texas Health San Antonio, San Antonio, USA
- South Texas Veterans Health Care System, San Antonio, TX USA
| |
Collapse
|
15
|
S100A9/CD163 expression profiles in classical monocytes as biomarkers to discriminate idiopathic pulmonary fibrosis from idiopathic nonspecific interstitial pneumonia. Sci Rep 2021; 11:12135. [PMID: 34108546 PMCID: PMC8190107 DOI: 10.1038/s41598-021-91407-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 05/04/2021] [Indexed: 12/22/2022] Open
Abstract
Circulating monocytes have pathogenic relevance in idiopathic pulmonary fibrosis (IPF). Here, we determined whether the cell surface levels of two markers, pro-inflammatory-related S100A9 and anti-inflammatory-related CD163, expressed on CD14strongCD16− classical monocytes by flow cytometry could discriminate IPF from idiopathic nonspecific interstitial pneumonia (iNSIP). Twenty-five patients with IPF, 25 with iNSIP, and 20 healthy volunteers were prospectively enrolled in this study. The S100A9+CD163− cell percentages in classical monocytes showed a pronounced decrease on monocytes in iNSIP compared to that in IPF. In contrast, the percentages of S100A9−CD163+ cells were significantly higher in iNSIP patients than in IPF patients and healthy volunteers. In IPF patients, there was a trend toward a correlation between the percentage of S100A9+CD163− monocytes and the surfactant protein-D (SP-D) serum levels (r = 0.4158, [95% confidence interval (CI) − 0.02042–0.7191], p = 0.051). The individual percentages of S100A9+CD163− and S100A9−CD163+ cells were also independently associated with IPF through multivariate regression analysis. The unadjusted area under the receiver operating characteristic curve (ROC-AUC) to discriminate IPF from iNSIP was (ROC-AUC 0.802, 95% CI [0.687–0.928]), suggesting that these are better biomarkers than serum SP-D (p < 0.05). This preliminary study reports the first comparative characterization of monocyte phenotypes between IPF and iNSIP.
Collapse
|
16
|
Mgbemena N, Jones A, Leicht AS. Relationship between handgrip strength and lung function in adults: a systematic review. Physiother Theory Pract 2021; 38:1908-1927. [PMID: 33870831 DOI: 10.1080/09593985.2021.1901323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Handgrip strength (HGS) is a functional test that has been directly associated with lung function in some healthy populations; however, inconsistent findings have been reported for populations with chronic diseases. The aim of this study was to identify the relationship between HGS and lung function in both healthy and unhealthy adults. A systematic search was conducted using six databases from their earliest inception to February 29, 2020. Two authors reviewed and assessed methodological quality of eligible studies using the Crowe Critical Appraisal Tool (CCAT). Twenty-five studies met the inclusion criteria with 8 and 17 studies examining healthy and unhealthy populations, respectively. Reported average methodological quality of all included studies using the CCAT was 38-85% with most rated as Good to Excellent. Despite the use of heterogeneous equipment and protocols during HGS and lung function assessments, significant positive and moderate correlations and/or regression coefficients were reported for healthy populations consistently. Conversely, the reported relationships between HGS and lung function for unhealthy counterparts were variable. Handgrip strength was significantly associated with lung function in most healthy adults. Future robust studies are needed to confirm the suitability of HGS to assess lung function for healthy and unhealthy adults.
Collapse
Affiliation(s)
- Nnamdi Mgbemena
- Department of Physiotherapy, James Cook University, Townsville, Queensland, Australia.,Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Queensland, Australia
| | - Anne Jones
- Department of Physiotherapy, James Cook University, Townsville, Queensland, Australia.,Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Queensland, Australia
| | - Anthony S Leicht
- Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Queensland, Australia.,Department of Sport and Exercise Science, James Cook University, Townsville, Queensland, Australia
| |
Collapse
|
17
|
Atabai K, Yang CD, Podolsky MJ. You Say You Want a Resolution (of Fibrosis). Am J Respir Cell Mol Biol 2020; 63:424-435. [PMID: 32640171 DOI: 10.1165/rcmb.2020-0182tr] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In pathological fibrosis, aberrant tissue remodeling with excess extracellular matrix leads to organ dysfunction and eventual morbidity. Diseases of fibrosis create significant global health and economic burdens and are often deadly. Although fibrosis has traditionally been thought of as an irreversible process, a growing body of evidence demonstrates that organ fibrosis can reverse in certain circumstances, especially if an underlying cause of injury can be removed. This body of evidence has uncovered more and more contributors to persistent and nonresolving tissue fibrosis. Here, we review the present knowledge on resolution of organ fibrosis and restoration of near-normal tissue architecture. We emphasize three critical areas of tissue homeostasis that are necessary for fibrosis resolution, namely, the elimination of matrix-producing cells, the clearance of excess matrix, and the regeneration of normal tissue constituents. In so doing, we also highlight how profibrotic pathways interact with one another and where there may be therapeutic opportunities to intervene and remediate pathological persistent fibrosis.
Collapse
Affiliation(s)
- Kamran Atabai
- Cardiovascular Research Institute.,Lung Biology Center, and.,Department of Medicine, University of California, San Francisco, San Francisco, California
| | | | - Michael J Podolsky
- Cardiovascular Research Institute.,Lung Biology Center, and.,Department of Medicine, University of California, San Francisco, San Francisco, California
| |
Collapse
|
18
|
Qu Y, Hao C, Zhai R, Yao W. Folate and macrophage folate receptor-β in idiopathic pulmonary fibrosis disease: the potential therapeutic target? Biomed Pharmacother 2020; 131:110711. [DOI: 10.1016/j.biopha.2020.110711] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/10/2020] [Accepted: 08/28/2020] [Indexed: 02/07/2023] Open
|
19
|
Effects of Pirfenidone and Nintedanib on Markers of Systemic Oxidative Stress and Inflammation in Patients with Idiopathic Pulmonary Fibrosis: A Preliminary Report. Antioxidants (Basel) 2020; 9:antiox9111064. [PMID: 33143144 PMCID: PMC7692317 DOI: 10.3390/antiox9111064] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/18/2020] [Accepted: 10/28/2020] [Indexed: 12/25/2022] Open
Abstract
Introduction: In vitro evidence suggests that pirfenidone and nintedanib, approved agents for the treatment of idiopathic pulmonary fibrosis (IPF), exert anti-inflammatory and anti-oxidant effects. We aimed to investigate such effects in vivo in IPF patients. Methods: Systemic circulating markers of oxidative stress [nuclear factor erythroid 2–related factor 2 (Nrf2), thiobarbituric acid- reactive substances (TBARS), homocysteine (Hcy), cysteine (Cys), asymmetric dimethylarginine (ADMA) and ADMA/Arginine ratio, glutathione (GSH), plasma protein –SH (PSH), and taurine (Tau)] and inflammation [Kynurenine (Kyn), Tryptophan (Trp) and Kyn/Trp ratio] were measured at baseline and after 24-week treatment in 18 IPF patients (10 treated with pirfenidone and 8 with nintedanib) and in 18 age- and sex-matched healthy controls. Results: Compared to controls, IPF patients had significantly lower concentrations of reduced blood GSH (457 ± 73 µmol/L vs 880 ± 212 µmol/L, p < 0.001) and plasma PSH (4.24 ± 0.95 µmol/g prot vs 5.28 ± 1.35 µmol/g prot, p = 0.012). Pirfenidone treatment significantly decreased the Kyn/Trp ratio (0.030 ± 0.011 baseline vs 0.025 ± 0.010 post-treatment, p = 0.048) whilst nintedanib treatment significantly increased blood GSH (486 ± 70 μmol/L vs 723 ± 194 μmol/L, p = 0.006) and reduced ADMA concentrations (0.501 ± 0.094 vs. 0.468 ± 0.071 μmol/L, p = 0.024). Conclusion: pirfenidone and nintedanib exert beneficial effects on specific markers of oxidative stress and inflammation in IPF patients.
Collapse
|
20
|
Louzada RA, Corre R, Ameziane El Hassani R, Meziani L, Jaillet M, Cazes A, Crestani B, Deutsch E, Dupuy C. NADPH oxidase DUOX1 sustains TGF-β1 signalling and promotes lung fibrosis. Eur Respir J 2020; 57:13993003.01949-2019. [DOI: 10.1183/13993003.01949-2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 07/22/2020] [Indexed: 01/20/2023]
Abstract
Interstitial lung fibroblast activation coupled with extracellular matrix production is a pathological signature of pulmonary fibrosis, and is governed by transforming growth factor (TGF)-β1/Smad signalling. TGF-β1 and oxidative stress cooperate to drive fibrosis. Cells can produce reactive oxygen species through activation and/or induction of NADPH oxidases, such as dual oxidase (DUOX1/2). Since DUOX enzymes, as extracellular hydrogen peroxide (H2O2)-generating systems, are involved in extracellular matrix formation and in wound healing in different experimental models, we hypothesised that DUOX-based NADPH oxidase plays a role in the pathophysiology of pulmonary fibrosis.Our in vivo data (idiopathic pulmonary fibrosis patients and mouse models of lung fibrosis) showed that the NADPH oxidase DUOX1 is induced in response to lung injury. DUOX1-deficient mice (DUOX1+/− and DUOX1−/−) had an attenuated fibrotic phenotype. In addition to being highly expressed at the epithelial surface of airways, DUOX1 appears to be well expressed in the fibroblastic foci of remodelled lungs. By using primary human and mouse lung fibroblasts, we showed that TGF-β1 upregulates DUOX1 and its maturation factor DUOXA1 and that DUOX1-derived H2O2 promoted the duration of TGF-β1-activated Smad3 phosphorylation by preventing phospho-Smad3 degradation. Analysis of the mechanism revealed that DUOX1 inhibited the interaction between phospho-Smad3 and the ubiquitin ligase NEDD4L, preventing NEDD4L-mediated ubiquitination of phospho-Smad3 and its targeting for degradation.These findings highlight a role for DUOX1-derived H2O2 in a positive feedback that amplifies the signalling output of the TGF-β1 pathway and identify DUOX1 as a new therapeutic target in pulmonary fibrosis.
Collapse
|
21
|
Korfei M, MacKenzie B, Meiners S. The ageing lung under stress. Eur Respir Rev 2020; 29:29/156/200126. [DOI: 10.1183/16000617.0126-2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/22/2020] [Indexed: 01/10/2023] Open
Abstract
Healthy ageing of the lung involves structural changes but also numerous cell-intrinsic and cell-extrinsic alterations. Among them are the age-related decline in central cellular quality control mechanisms such as redox and protein homeostasis. In this review, we would like to provide a conceptual framework of how impaired stress responses in the ageing lung, as exemplified by dysfunctional redox and protein homeostasis, may contribute to onset and progression of COPD and idiopathic pulmonary fibrosis (IPF). We propose that age-related imbalanced redox and protein homeostasis acts, amongst others (e.g.cellular senescence), as a “first hit” that challenges the adaptive stress-response pathways of the cell, increases the level of oxidative stress and renders the lung susceptible to subsequent injury and disease. In both COPD and IPF, additional environmental insults such as smoking, air pollution and/or infections then serve as “second hits” which contribute to persistently elevated oxidative stress that overwhelms the already weakened adaptive defence and repair pathways in the elderly towards non-adaptive, irremediable stress thereby promoting development and progression of respiratory diseases. COPD and IPF are thus distinct horns of the same devil, “lung ageing”.
Collapse
|
22
|
Galati D, Zanotta S, Polistina GE, Coppola A, Capitelli L, Bocchino M. Circulating dendritic cells are severely decreased in idiopathic pulmonary fibrosis with a potential value for prognosis prediction. Clin Immunol 2020; 215:108454. [DOI: 10.1016/j.clim.2020.108454] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 04/15/2020] [Accepted: 04/29/2020] [Indexed: 01/13/2023]
|
23
|
Otoupalova E, Smith S, Cheng G, Thannickal VJ. Oxidative Stress in Pulmonary Fibrosis. Compr Physiol 2020; 10:509-547. [PMID: 32163196 DOI: 10.1002/cphy.c190017] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Oxidative stress has been linked to various disease states as well as physiological aging. The lungs are uniquely exposed to a highly oxidizing environment and have evolved several mechanisms to attenuate oxidative stress. Idiopathic pulmonary fibrosis (IPF) is a progressive age-related disorder that leads to architectural remodeling, impaired gas exchange, respiratory failure, and death. In this article, we discuss cellular sources of oxidant production, and antioxidant defenses, both enzymatic and nonenzymatic. We outline the current understanding of the pathogenesis of IPF and how oxidative stress contributes to fibrosis. Further, we link oxidative stress to the biology of aging that involves DNA damage responses, loss of proteostasis, and mitochondrial dysfunction. We discuss the recent findings on the role of reactive oxygen species (ROS) in specific fibrotic processes such as macrophage polarization and immunosenescence, alveolar epithelial cell apoptosis and senescence, myofibroblast differentiation and senescence, and alterations in the acellular extracellular matrix. Finally, we provide an overview of the current preclinical studies and clinical trials targeting oxidative stress in fibrosis and potential new strategies for future therapeutic interventions. © 2020 American Physiological Society. Compr Physiol 10:509-547, 2020.
Collapse
Affiliation(s)
- Eva Otoupalova
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sam Smith
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Guangjie Cheng
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Victor J Thannickal
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
24
|
Veith C, Boots AW, Idris M, van Schooten FJ, van der Vliet A. Redox Imbalance in Idiopathic Pulmonary Fibrosis: A Role for Oxidant Cross-Talk Between NADPH Oxidase Enzymes and Mitochondria. Antioxid Redox Signal 2019; 31:1092-1115. [PMID: 30793932 PMCID: PMC6767863 DOI: 10.1089/ars.2019.7742] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Significance: Idiopathic pulmonary fibrosis (IPF) is a progressive age-related lung disease with a median survival of only 3 years after diagnosis. The pathogenic mechanisms behind IPF are not clearly understood, and current therapeutic approaches have not been successful in improving disease outcomes. Recent Advances: IPF is characterized by increased production of reactive oxygen species (ROS), primarily by NADPH oxidases (NOXes) and mitochondria, as well as altered antioxidant defenses. Recent studies have identified the NOX isoform NOX4 as a key player in various important aspects of IPF pathology. In addition, mitochondrial dysfunction is thought to enhance pathological features of IPF, in part by increasing mitochondrial ROS (mtROS) production and altering cellular metabolism. Recent findings indicate reciprocal interactions between NOX enzymes and mitochondria, which affect regulation of NOX activity as well as mitochondrial function and mtROS production, and collectively promote epithelial injury and profibrotic signaling. Critical Issues and Future Directions: The precise molecular mechanisms by which ROS from NOX or mitochondria contribute to IPF pathology are not known. This review summarizes the current knowledge with respect to the various aspects of ROS imbalance in the context of IPF and its proposed roles in disease development, with specific emphasis on the importance of inappropriate NOX activation, mitochondrial dysfunction, and the emerging evidence of NOX-mitochondria cross-talk as important drivers in IPF pathobiology.
Collapse
Affiliation(s)
- Carmen Veith
- Department of Pharmacology and Toxicology, Faculty of Health, Medicine and Life Sciences, NUTRIM School of Nutrition, Translational Research and Metabolism, University of Maastricht, Maastricht, the Netherlands
| | - Agnes W. Boots
- Department of Pharmacology and Toxicology, Faculty of Health, Medicine and Life Sciences, NUTRIM School of Nutrition, Translational Research and Metabolism, University of Maastricht, Maastricht, the Netherlands
| | - Musa Idris
- Department of Pharmacology and Toxicology, Faculty of Health, Medicine and Life Sciences, NUTRIM School of Nutrition, Translational Research and Metabolism, University of Maastricht, Maastricht, the Netherlands
| | - Frederik-Jan van Schooten
- Department of Pharmacology and Toxicology, Faculty of Health, Medicine and Life Sciences, NUTRIM School of Nutrition, Translational Research and Metabolism, University of Maastricht, Maastricht, the Netherlands
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
- Address correspondence to: Dr. Albert van der Vliet, Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, HSRF 216, 149 Beaumont Avenue, Burlington, VT 05405
| |
Collapse
|
25
|
Current advances in idiopathic pulmonary fibrosis: the pathogenesis, therapeutic strategies and candidate molecules. Future Med Chem 2019; 11:2595-2620. [PMID: 31633402 DOI: 10.4155/fmc-2019-0111] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a type of chronic, progressive lung disease with unknown cause, which is characterized by increasing dyspnea and destruction of lung function with a high mortality rate. Evolving evidence demonstrated that the pathogenesis of IPF involved multiple signaling pathways such as inflammation, oxidative stress and fibrosis. However, drug discovery to prevent or revert IPF has been insufficient to cope with the development. Drug discovery targeting multiple links should be considered. In this review, we will brief the pathogenesis of IPF and discuss several small chemical entities toward the pathogenesis for IPF studied in animal models and clinical trials. The field of novel anti-IPF agents and the future directions for the prevention and treatment of IPF are detailed thoroughly discussed.
Collapse
|
26
|
Tsubouchi K, Araya J, Yoshida M, Sakamoto T, Koumura T, Minagawa S, Hara H, Hosaka Y, Ichikawa A, Saito N, Kadota T, Kurita Y, Kobayashi K, Ito S, Fujita Y, Utsumi H, Hashimoto M, Wakui H, Numata T, Kaneko Y, Mori S, Asano H, Matsudaira H, Ohtsuka T, Nakayama K, Nakanishi Y, Imai H, Kuwano K. Involvement of GPx4-Regulated Lipid Peroxidation in Idiopathic Pulmonary Fibrosis Pathogenesis. THE JOURNAL OF IMMUNOLOGY 2019; 203:2076-2087. [DOI: 10.4049/jimmunol.1801232] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 08/08/2019] [Indexed: 12/22/2022]
|
27
|
Bromodomain and Extraterminal (BET) Protein Inhibition Restores Redox Balance and Inhibits Myofibroblast Activation. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1484736. [PMID: 31119153 PMCID: PMC6500679 DOI: 10.1155/2019/1484736] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 03/04/2019] [Accepted: 04/01/2019] [Indexed: 12/28/2022]
Abstract
Background and Objective Progressive pulmonary fibrosis is the main cause of death in patients with systemic sclerosis (SSc) with interstitial lung disease (ILD) and in those with idiopathic pulmonary fibrosis (IPF). Transforming growth factor-β (TGF-β) and NADPH oxidase- (NOX-) derived reactive oxygen species (ROS) are drivers of lung fibrosis. We aimed to determine the role of the epigenetic readers, bromodomain and extraterminal (BET) proteins in the regulation of redox balance in activated myofibroblasts. Methods In TGF-β-stimulated fibroblasts, we investigated the effect of the BET inhibitor JQ1 on the mRNA expression of the prooxidant gene NOX4 and the antioxidant gene superoxide dismutase (SOD2) by quantitative RT-PCR, the antioxidant transcription factor NF-E2-related factor 2 (Nrf2) activity by a reporter assay, and intracellular ROS levels by dichlorofluorescein staining. Myofibroblast activation was determined by α-smooth muscle actin immunocytochemistry. The role of specific BET protein isoforms in NOX4 gene regulation was studied by siRNA silencing and chromatin-immunoprecipitation. Results and Conclusions Affymetrix gene array analysis revealed increased NOX4 and reduced SOD2 expression in SSc and IPF fibroblasts. SOD2 silencing in non-ILD control fibroblasts induced a profibrotic phenotype. TGF-β increased NOX4 and inhibited SOD2 expression, while increasing ROS production and myofibroblast differentiation. JQ1 reversed the TGF-β-mediated NOX4/SOD2 imbalance and Nrf2 inactivation and attenuated ROS production and myofibroblast differentiation. The BET proteins Brd3 and Brd4 were shown to bind to the NOX4 promoter and drive TGF-β-induced NOX4 expression. Our data indicate a critical role of BET proteins in promoting redox imbalance and pulmonary myofibroblast activation and support BET bromodomain inhibitors as a potential therapy for fibrotic lung disease.
Collapse
|
28
|
Guler SA, Hur SA, Lear SA, Camp PG, Ryerson CJ. Body composition, muscle function, and physical performance in fibrotic interstitial lung disease: a prospective cohort study. Respir Res 2019; 20:56. [PMID: 30866948 PMCID: PMC6417197 DOI: 10.1186/s12931-019-1019-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 02/27/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Patients with fibrotic interstitial lung disease (ILD) are frequently physically inactive and many ILD subtypes are characterized by risk factors for myopathy; however, the importance of body composition, muscle strength, and physical performance in this population is largely unknown. METHODS Patients were prospectively recruited from a specialized ILD clinic, baseline characteristics were collected from the clinical record, pulmonary function tests were performed per established protocols, and dyspnea was measured using the University of California San Diego Shortness of Breath Questionnaire. Dual-energy X-ray absorptiometry (DXA) was used to assess body composition; handgrip strength to determine muscle strength, and 4-m gait speed to measure physical performance. RESULTS One hundred and fifteen patients with fibrotic ILD including 40 patients with idiopathic pulmonary fibrosis were recruited. The mean age was 69+/- 10 years in men (62% of the cohort), and 66+/- 9 years in women, with mild and moderate reduction in FVC and DLCO, respectively, for both sexes. ILD severity (measured by FVC %-predicted, DLCO %-predicted, or the Composite Physiologic Index in separate models) significantly predicted muscle mass and percent body fat including with adjustment for age, sex, and weight. ILD severity was associated with grip strength and gait speed independent from body composition. CONCLUSIONS ILD severity has an important impact on body composition, particularly in men. Future studies are needed to confirm and further explore the possibility of additional pathways through which ILD directly impacts limb muscle function and physical performance.
Collapse
Affiliation(s)
- Sabina A. Guler
- Department of Medicine, University of British Columbia, Vancouver, Canada
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada
- Department of Pulmonary Medicine, University Hospital and University of Bern, Bern, Switzerland
| | - Seo Am Hur
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada
| | - Scott A. Lear
- Department of Medicine, University of British Columbia, Vancouver, Canada
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Pat G. Camp
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada
- Department of Physical Therapy, University of British Columbia, Vancouver, Canada
| | - Christopher J. Ryerson
- Department of Medicine, University of British Columbia, Vancouver, Canada
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada
- St. Paul’s Hospital, 1081 Burrard St, Ward 8B, Vancouver, BC V6Z 1Y6 Canada
| |
Collapse
|
29
|
Okuda R, Aoshiba K, Matsushima H, Ogura T, Okudela K, Ohashi K. Cellular senescence and senescence-associated secretory phenotype: comparison of idiopathic pulmonary fibrosis, connective tissue disease-associated interstitial lung disease, and chronic obstructive pulmonary disease. J Thorac Dis 2019; 11:857-864. [PMID: 31019774 DOI: 10.21037/jtd.2019.02.11] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background The senescence-associated secretory phenotype (SASP) develops due to cellular senescence during conditions such as chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). However, studies comparing the degree of cellular senescence and SASP between COPD and IPF are limited. Furthermore, to the best of our knowledge, no study has examined cellular senescence and/or SASP in connective tissue disease-associated interstitial lung disease (CTD-ILD). Methods To compare the degree of cellular senescence among COPD, IPF, and CTD-ILD, tissue samples from surgical lung biopsies or noncancerous tissue from lobectomy specimens of patients with lung cancer were subjected to immunostaining for p16 and p21. Double-staining for p16 and phosphorylated NF-κB was performed to verify the relationship between cellular senescence and SASP. Results There was a greater degree of enhancement of p16 and p21 expression in patients with IPF than in those with COPD and controls. Immunostaining for p16 revealed an enhanced expression of this marker in patients with COPD compared with that in controls. No significant differences were observed in the phosphorylated NF-κB expression rate of p16-positive and p16-negative cells among patients with IPF, CTD-ILD, and COPD. Conclusions Epithelial cells in patients with IPF express higher levels of both cellular senescence and SASP than those in patients with COPD or controls.
Collapse
Affiliation(s)
- Ryo Okuda
- Department of Respiratory Medicine, Kanagawa Cardiovascular and Respiratory Center, Yokohama, Japan
| | - Kazutetsu Aoshiba
- Department of Respiratory Medicine, Tokyo Medical University Ibaraki Medical Center, Inashiki, Japan
| | - Hidekazu Matsushima
- Department of Respiratory Medicine, Saitama Red Cross Hospital, Saitama, Japan
| | - Takashi Ogura
- Department of Respiratory Medicine, Kanagawa Cardiovascular and Respiratory Center, Yokohama, Japan
| | - Koji Okudela
- Department of Pathology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kenichi Ohashi
- Department of Pathology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
30
|
Cysteine/Glutathione Deficiency: A Significant and Treatable Corollary of Disease. THE THERAPEUTIC USE OF N-ACETYLCYSTEINE (NAC) IN MEDICINE 2019. [PMCID: PMC7120747 DOI: 10.1007/978-981-10-5311-5_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Glutathione (GSH) deficiency may play a pivotal role in a variety of apparently unrelated clinical conditions and diseases. Orally administered N-acetylcysteine (NAC), which replenishes the cysteine required for GSH synthesis, has been tested in a large number of randomized placebo-controlled trials involving these diseases and conditions. This chapter focused on developing a base of evidence suggesting that NAC administration improves disease by increasing cysteine and/or GSH in a variety of diseases, thereby implying a significant role for GSH deficiency in the clinical basis of many diseases. To develop this base of evidence, we systematically selected studies which considered the hypothesis that the therapeutic efficacy for NAC is an indication that cysteine and/or GSH deficiency is a pathophysiological part of the diseases studied. In this manner we focus this chapter on explaining the biological mechanisms of NAC therapy in a wide variety of disorders and demonstrate its ubiquitous role in improving disease that involves disrupted GSH and/or cysteine metabolism.
Collapse
|
31
|
Paliogiannis P, Fois AG, Collu C, Bandinu A, Zinellu E, Carru C, Pirina P, Mangoni AA, Zinellu A. Oxidative stress-linked biomarkers in idiopathic pulmonary fibrosis: a systematic review and meta-analysis. Biomark Med 2018; 12:1175-1184. [PMID: 30191745 DOI: 10.2217/bmm-2018-0108] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AIM The aim of this meta-analysis was to investigate associations between idiopathic pulmonary fibrosis (IPF) and markers of oxidative stress (OS) measured in different biological samples. METHODS A systematic search of publications listed in PubMed, Web of Science, Scopus and Google Scholar from inception to December 2017 was conducted. RESULTS Significant differences between IPF patients and controls were observed for all biomarkers (thiobarbituric acid reactive substances, hydroperoxides and glutathione), barring systemic isoprostanes. CONCLUSION This meta-analysis showed a consistent increase in the concentrations of OS markers or a reduction in antioxidant markers, in patients with IPF, independent of the type of biological sample. Pending the results of further studies, OS biomarkers might be useful for the diagnosis and monitoring of IPF.
Collapse
Affiliation(s)
- Panagiotis Paliogiannis
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
| | - Alessandro G Fois
- Department of Clinical, Surgical & Experimental Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
| | - Claudia Collu
- Department of Clinical, Surgical & Experimental Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
| | - Antonietta Bandinu
- Department of Clinical, Surgical & Experimental Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
| | - Elisabetta Zinellu
- Department of Clinical, Surgical & Experimental Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
| | - Pietro Pirina
- Department of Clinical, Surgical & Experimental Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
| | - Arduino A Mangoni
- Department of Clinical Pharmacology, College of Medicine & Public Health, Flinders University, Adelaide, Australia
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
| |
Collapse
|
32
|
Anathy V, Lahue KG, Chapman DG, Chia SB, Casey DT, Aboushousha R, van der Velden JLJ, Elko E, Hoffman SM, McMillan DH, Jones JT, Nolin JD, Abdalla S, Schneider R, Seward DJ, Roberson EC, Liptak MD, Cousins ME, Butnor KJ, Taatjes DJ, Budd RC, Irvin CG, Ho YS, Hakem R, Brown KK, Matsui R, Bachschmid MM, Gomez JL, Kaminski N, van der Vliet A, Janssen-Heininger YMW. Reducing protein oxidation reverses lung fibrosis. Nat Med 2018; 24:1128-1135. [PMID: 29988126 PMCID: PMC6204256 DOI: 10.1038/s41591-018-0090-y] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 05/14/2018] [Indexed: 12/21/2022]
Abstract
Idiopathic pulmonary fibrosis is characterized by excessive deposition of collagen in the lung, leading to chronically impaired gas exchange and death1-3. Oxidative stress is believed to be critical in this disease pathogenesis4-6, although the exact mechanisms remain enigmatic. Protein S-glutathionylation (PSSG) is a post-translational modification of proteins that can be reversed by glutaredoxin-1 (GLRX)7. It remains unknown whether GLRX and PSSG play a role in lung fibrosis. Here, we explored the impact of GLRX and PSSG status on the pathogenesis of pulmonary fibrosis, using lung tissues from subjects with idiopathic pulmonary fibrosis, transgenic mouse models and direct administration of recombinant Glrx to airways of mice with existing fibrosis. We demonstrate that GLRX enzymatic activity was strongly decreased in fibrotic lungs, in accordance with increases in PSSG. Mice lacking Glrx were far more susceptible to bleomycin- or adenovirus encoding active transforming growth factor beta-1 (AdTGFB1)-induced pulmonary fibrosis, whereas transgenic overexpression of Glrx in the lung epithelium attenuated fibrosis. We furthermore show that endogenous GLRX was inactivated through an oxidative mechanism and that direct administration of the Glrx protein into airways augmented Glrx activity and reversed increases in collagen in mice with TGFB1- or bleomycin-induced fibrosis, even when administered to fibrotic, aged animals. Collectively, these findings suggest the therapeutic potential of exogenous GLRX in treating lung fibrosis.
Collapse
Affiliation(s)
- Vikas Anathy
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Karolyn G Lahue
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - David G Chapman
- Department of Medicine, University of Vermont, Burlington, VT, USA
| | - Shi B Chia
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Dylan T Casey
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Reem Aboushousha
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Jos L J van der Velden
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Evan Elko
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Sidra M Hoffman
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - David H McMillan
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Jane T Jones
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - James D Nolin
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Sarah Abdalla
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Robert Schneider
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - David J Seward
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | | | - Matthew D Liptak
- Department of Chemistry, University of Vermont, Burlington, VT, USA
| | - Morgan E Cousins
- Department of Chemistry, University of Vermont, Burlington, VT, USA
| | - Kelly J Butnor
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Douglas J Taatjes
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Ralph C Budd
- Department of Medicine, University of Vermont, Burlington, VT, USA
| | - Charles G Irvin
- Department of Medicine, University of Vermont, Burlington, VT, USA
| | - Ye-Shih Ho
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, USA
| | - Razq Hakem
- Department of Medical Biophysics and Immunology, University of Toronto, and the Ontario Cancer Institute/University Health Network, Toronto, Ontario, Canada
| | - Kevin K Brown
- Department of Medicine, Pulmonary, Critical Care and Sleep Section, National Jewish Health and the University of Colorado, Denver, CO, USA
| | - Reiko Matsui
- Department of Medicine, Boston University, Boston, MA, USA
| | | | - Jose L Gomez
- Department of Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Naftali Kaminski
- Department of Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | | |
Collapse
|
33
|
Strzelak A, Ratajczak A, Adamiec A, Feleszko W. Tobacco Smoke Induces and Alters Immune Responses in the Lung Triggering Inflammation, Allergy, Asthma and Other Lung Diseases: A Mechanistic Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 15:E1033. [PMID: 29883409 PMCID: PMC5982072 DOI: 10.3390/ijerph15051033] [Citation(s) in RCA: 353] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/08/2018] [Accepted: 05/09/2018] [Indexed: 02/06/2023]
Abstract
Many studies have been undertaken to reveal how tobacco smoke skews immune responses contributing to the development of chronic obstructive pulmonary disease (COPD) and other lung diseases. Recently, environmental tobacco smoke (ETS) has been linked with asthma and allergic diseases in children. This review presents the most actual knowledge on exact molecular mechanisms responsible for the skewed inflammatory profile that aggravates inflammation, promotes infections, induces tissue damage, and may promote the development of allergy in individuals exposed to ETS. We demonstrate how the imbalance between oxidants and antioxidants resulting from exposure to tobacco smoke leads to oxidative stress, increased mucosal inflammation, and increased expression of inflammatory cytokines (such as interleukin (IL)-8, IL-6 and tumor necrosis factor α ([TNF]-α). Direct cellular effects of ETS on epithelial cells results in increased permeability, mucus overproduction, impaired mucociliary clearance, increased release of proinflammatory cytokines and chemokines, enhanced recruitment of macrophages and neutrophils and disturbed lymphocyte balance towards Th2. The plethora of presented phenomena fully justifies a restrictive policy aiming at limiting the domestic and public exposure to ETS.
Collapse
Affiliation(s)
- Agnieszka Strzelak
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warszawa, Poland.
| | - Aleksandra Ratajczak
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warszawa, Poland.
| | - Aleksander Adamiec
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warszawa, Poland.
| | - Wojciech Feleszko
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warszawa, Poland.
| |
Collapse
|
34
|
Fois AG, Paliogiannis P, Sotgia S, Mangoni AA, Zinellu E, Pirina P, Carru C, Zinellu A. Evaluation of oxidative stress biomarkers in idiopathic pulmonary fibrosis and therapeutic applications: a systematic review. Respir Res 2018; 19:51. [PMID: 29587761 PMCID: PMC5872514 DOI: 10.1186/s12931-018-0754-7] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 03/19/2018] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Idiopathic pulmonary fibrosis (IPF), a fatal lung disease of unknown origin, is characterized by chronic and progressive fibrosing interstitial pneumonia which progressively impairs lung function. Oxidative stress is one of the main pathogenic pathways in IPF. The aim of this systematic review was to describe the type of markers of oxidative stress identified in different biological specimens and the effects of antioxidant therapies in patients with IPF. METHODS We conducted a systematic search of publications listed in electronic databases (Pubmed, Web of Science, Scopus and Google Scholar) from inception to October 2017. Two investigators independently reviewed all identified articles to determine eligibility. RESULTS After a substantial proportion of the initially identified articles (n = 554) was excluded because they were duplicates, abstracts, irrelevant, or did not meet the selection criteria, we identified 30 studies. In each study, we critically appraised the type, site (systemic vs. local, e.g. breath, sputum, expired breath condensate, epithelial lining fluid, bronchoalveolar lavage, and lung tissue specimens), and method used for measuring the identified oxidative stress biomarkers. Furthermore, the current knowledge on antioxidant therapies in IPF was summarized. CONCLUSIONS A number of markers of oxidative stress, with individual advantages and limitations, have been described in patients with IPF. Nevertheless, trials of antioxidant treatments have been unable to demonstrate consistent benefits, barring recent pharmacogenomics data suggesting different results in specific genotype subgroups of patients with IPF.
Collapse
Affiliation(s)
- Alessandro G Fois
- Department of Clinical and Experimental Medicine, University of Sassari, Sassari, Italy. .,Department of Respiratory Diseases, University Hospital Sassari (AOU), Sassari, Italy.
| | | | - Salvatore Sotgia
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Arduino A Mangoni
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Elisabetta Zinellu
- Department of Respiratory Diseases, University Hospital Sassari (AOU), Sassari, Italy
| | - Pietro Pirina
- Department of Clinical and Experimental Medicine, University of Sassari, Sassari, Italy.,Department of Respiratory Diseases, University Hospital Sassari (AOU), Sassari, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
35
|
Wei Z, Caty J, Whitson J, Zhang AD, Srinivasagan R, Kavanagh TJ, Yan H, Fan X. Reduced Glutathione Level Promotes Epithelial-Mesenchymal Transition in Lens Epithelial Cells via a Wnt/β-Catenin-Mediated Pathway: Relevance for Cataract Therapy. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2399-2412. [PMID: 28827139 DOI: 10.1016/j.ajpath.2017.07.018] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/19/2017] [Accepted: 07/26/2017] [Indexed: 12/11/2022]
Abstract
The epithelial-mesenchymal transition (EMT) process plays a pivotal role in the pathogenesis of posterior capsular opacification because of remnant lens epithelial cell proliferation, migration, and transformation after cataract surgery. The latter, we hypothesize, may result in posterior capsule wrinkling and opacification because of a profound change in the lens growth environment via a 1000-fold reduction of extracellular glutathione (GSH) levels. To test this hypothesis, we investigated the EMT process in cell culture and GSH biosynthesis deficiency mouse models. Our data indicate a dramatic increase of pro-EMT markers, such as type I collagen, α-smooth muscle actin, vimentin, and fibronectin, under conditions of lens GSH depletion. Further study suggests that decreased GSH triggers the Wnt/β-catenin signal transduction pathway, independent of transforming growth factor-β. Equally important, the antioxidants N-acetyl cysteine and GSH ethyl ester could significantly attenuate the EMT signaling stimulated by decreased GSH levels. These findings were further confirmed by mock cataract surgery in both gamma glutamyl-cysteine ligase, catalytic subunit, and gamma glutamyl-cysteine ligase, modifier subunit, knockout mouse models. Remarkably, increased EMT marker expression, β-catenin activation, and translocation into the nucleus were found in both knockout mice compared with the wild type, and such increased expression could be significantly attenuated by N-acetyl cysteine or GSH ethyl ester treatment. This study, for the first time we believe, links oxidative stress to lens fibrosis and posterior capsular opacification formation via EMT-mediated mechanisms.
Collapse
Affiliation(s)
- Zongbo Wei
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Jane Caty
- Department of Ophthalmology, University Hospitals of Cleveland Medical Center, Cleveland, Ohio
| | - Jeremy Whitson
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Amy D Zhang
- Department of Ophthalmology, University Hospitals of Cleveland Medical Center, Cleveland, Ohio
| | | | - Terrance J Kavanagh
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Hong Yan
- Department of Ophthalmology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China; Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xingjun Fan
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio.
| |
Collapse
|
36
|
Chu H, Shi Y, Jiang S, Zhong Q, Zhao Y, Liu Q, Ma Y, Shi X, Ding W, Zhou X, Cui J, Jin L, Guo G, Wang J. Treatment effects of the traditional Chinese medicine Shenks in bleomycin-induced lung fibrosis through regulation of TGF-beta/Smad3 signaling and oxidative stress. Sci Rep 2017; 7:2252. [PMID: 28533545 PMCID: PMC5440393 DOI: 10.1038/s41598-017-02293-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 04/05/2017] [Indexed: 12/20/2022] Open
Abstract
Pulmonary fibrosis is a kind of devastating interstitial lung disease due to the limited therapeutic strategies. Traditional Chinese medicine (TCM) practices have put forth Shenks as a promising treatment approach. Here, we performed in vivo study and in vitro study to delineate the anti-fibrotic mechanisms behind Shenks treatment for pulmonary fibrosis. We found that regardless of the prophylactic or therapeutic treatment, Shenks was able to attenuate BLM-induced-fibrosis in mice, down regulate extracellular matrix genes expression, and reduce collagen production. The aberrantly high Smad3 phosphorylation levels and SBE activity in TGF-β-induced fibroblasts were dramatically decreased as a result of Shenks treatment. At the same time, Shenks was able to increase the expression of antioxidant-related genes, including Gclc and Ec-sod, while reduce the transcription levels of oxidative-related genes, such as Rac1 and Nox4 demonstrated by both in vivo and in vitro studies. Further investigations found that Shenks could decrease the oxidative productions of protein (3-nitrotyrosine) and lipid (malondialdehyde) and increase GSH content both in bleomycin treated mouse lungs and TGF-β stimulated fibroblasts, as well as inhibit the production of ROS stimulated by TGF-β to fight against oxidative stress. Overall, Shenks inhibited fibrosis by blocking TGF-β pathway and modulating the oxidant/antioxidant balance.
Collapse
Affiliation(s)
- Haiyan Chu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200438, P. R. China
| | - Ying Shi
- Department of Rheumatology and Immunology, Yiling Affiliated Hospital of Hebei Medical University, Shijiazhuang, 050091, China
- Department of Traditional Chinese Medicine, Geriatric Hospital of Hebei Province, Shijiazhuang, 050011, China
| | - Shuai Jiang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200438, P. R. China
| | - Qicheng Zhong
- Department of Rheumatology and Immunology, Yiling Affiliated Hospital of Hebei Medical University, Shijiazhuang, 050091, China
| | - Yongqiang Zhao
- Department of Rheumatology and Immunology, Yiling Affiliated Hospital of Hebei Medical University, Shijiazhuang, 050091, China
| | - Qingmei Liu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200438, P. R. China
| | - Yanyun Ma
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200438, P. R. China
| | - Xiangguang Shi
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200438, P. R. China
| | - Weifeng Ding
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200438, P. R. China
| | - Xiaodong Zhou
- University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, Texas, 77030, USA
| | - Jimin Cui
- Department of Rheumatology and Immunology, Yiling Affiliated Hospital of Hebei Medical University, Shijiazhuang, 050091, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200438, P. R. China
| | - Gang Guo
- Department of Rheumatology and Immunology, Yiling Affiliated Hospital of Hebei Medical University, Shijiazhuang, 050091, China.
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200438, P. R. China.
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, 200040, P. R. China.
| |
Collapse
|
37
|
Layton AM, Armstrong HF, Kim HP, Meza KS, D'Ovidio F, Arcasoy SM. Cardiopulmonary exercise factors predict survival in patients with advanced interstitial lung disease referred for lung transplantation. Respir Med 2017; 126:59-67. [PMID: 28427551 DOI: 10.1016/j.rmed.2017.03.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 02/06/2017] [Accepted: 03/24/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND The purpose of this work was to determine if parameters assessed during Cardiopulmonary Exercise Testing (CPET) while using supplemental oxygen can independently predict one-year transplant-free survival in patients with Interstitial Lung Disease (ILD) referred for lung transplant evaluation. METHODS We performed a chart review of patients with ILD who completed CPET with 30% FiO2 and gathered spirometry, pulmonary hemodynamic, six-minute walk, and CPET data. The primary end-point was death or lung transplantation within one-year of CPET. RESULTS The final data set included 192 patients. 79 patients died/underwent transplant, 113 survived transplant-free. Multivariable Cox regression revealed peak workload % predicted, nadir CPET SpO2, and FVC% predicted as independent predictors of one-year transplant-free survival. Of the independent predictors of survival, receiver operating characteristics analysis revealed peak workload %predicted cutoff of 35% to be highly discriminatory, more so than nadir CPET SpO2 or FVC % predicted in identifying patients at risk for one-year mortality or transplant (peak workload % predicted < 35% HR = 4.71, 95% CI = 2.64-8.38 and area under the curve (AUC) = 0.740, nadir CPET SpO2 < 86% HR = 2.27, 95%CI = 1.41-3.68, AUC = 0.645, FVC %predicted <45% HR = 1.82, 95% CI = 1.15-2.87, AUC = 0.624). CONCLUSION Peak workload % predicted, nadir CPET SpO2, and FVC% predicted in ILD patients referred for lung transplant evaluation are independently predictive of one-year mortality or need for transplant.
Collapse
Affiliation(s)
- Aimee M Layton
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Columbia University Medical Center, New York, NY, USA.
| | - Hilary F Armstrong
- Department of Rehabilitation and Regenerative Medicine, Columbia University Medical Center, New York, NY, USA; Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | | | | | - Frank D'Ovidio
- Columbia Lung Transplant Program, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Selim M Arcasoy
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Columbia University Medical Center, New York, NY, USA; Columbia Lung Transplant Program, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
38
|
Poh TY, Mac Aogáin M, Chan AKW, Yii ACA, Yong VFL, Tiew PY, Koh MS, Chotirmall SH. Understanding COPD-overlap syndromes. Expert Rev Respir Med 2017; 11:285-298. [PMID: 28282995 DOI: 10.1080/17476348.2017.1305895] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Chronic obstructive pulmonary disease accounts for a large burden of lung disease. It can 'overlap' with other respiratory diseases including bronchiectasis, fibrosis and obstructive sleep apnea (OSA). While COPD alone confers morbidity and mortality, common features with contrasting clinical outcomes can occur in COPD 'overlap syndromes'. Areas covered: Given the large degree of heterogeneity in COPD, individual variation to treatment is adopted based on its observed phenotype, which in turn overlaps with features of other respiratory disease states such as asthma. This is coined asthma-COPD overlap syndrome ('ACOS'). Other examples of such overlapping clinical states include bronchiectasis-COPD ('BCOS'), fibrosis-COPD ('FCOS') and OSA-COPD ('OCOS'). The objective of this review is to highlight similarities and differences between the COPD-overlap syndromes in terms of risk factors, pathophysiology, diagnosis and potential treatment differences. Expert commentary: As a consequence of COPD overlap syndromes, a transition from the traditional 'one size fits all' treatment approach is necessary. Greater treatment stratification according to clinical phenotype using a precision medicine approach is now required. In this light, it is important to recognize and differentiate COPD overlap syndromes as distinct disease states compared to individual diseases such as asthma, COPD, fibrosis or bronchiectasis.
Collapse
Affiliation(s)
- Tuang Yeow Poh
- a Lee Kong Chian School of Medicine, Translational Respiratory Research Laboratory , Nanyang Technological University , Singapore , Singapore
| | - Micheál Mac Aogáin
- a Lee Kong Chian School of Medicine, Translational Respiratory Research Laboratory , Nanyang Technological University , Singapore , Singapore
| | - Adrian Kwok Wai Chan
- b Department of Respiratory & Critical Care Medicine , Singapore General Hospital , Singapore , Singapore
| | - Anthony Chau Ang Yii
- b Department of Respiratory & Critical Care Medicine , Singapore General Hospital , Singapore , Singapore
| | - Valerie Fei Lee Yong
- a Lee Kong Chian School of Medicine, Translational Respiratory Research Laboratory , Nanyang Technological University , Singapore , Singapore
| | - Pei Yee Tiew
- b Department of Respiratory & Critical Care Medicine , Singapore General Hospital , Singapore , Singapore
| | - Mariko Siyue Koh
- b Department of Respiratory & Critical Care Medicine , Singapore General Hospital , Singapore , Singapore
| | - Sanjay Haresh Chotirmall
- a Lee Kong Chian School of Medicine, Translational Respiratory Research Laboratory , Nanyang Technological University , Singapore , Singapore
| |
Collapse
|
39
|
Glutathione peroxidase 3 localizes to the epithelial lining fluid and the extracellular matrix in interstitial lung disease. Sci Rep 2016; 6:29952. [PMID: 27435875 PMCID: PMC4951690 DOI: 10.1038/srep29952] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 06/27/2016] [Indexed: 12/20/2022] Open
Abstract
Aberrant antioxidant activity and excessive deposition of extracellular matrix (ECM) are hallmarks of interstitial lung diseases (ILD). It is known that oxidative stress alters the ECM, but extracellular antioxidant defence mechanisms in ILD are incompletely understood. Here, we extracted abundance and detergent solubility of extracellular antioxidant enzymes from a proteomic dataset of bleomycin-induced lung fibrosis in mice and assessed regulation and distribution of glutathione peroxidase 3 (GPX3) in murine and human lung fibrosis. Superoxide dismutase 3 (Sod3), Gpx3, and Gpx activity were increased in mouse BALF during bleomycin-induced lung fibrosis. In lung tissue homogenates, Gpx3, but not Sod3, was upregulated and detergent solubility profiling indicated that Gpx3 associated with ECM proteins. Immunofluorescence analysis showed that Gpx3 was expressed by bronchial epithelial cells and interstitial fibroblasts and localized to the basement membrane and interstitial ECM in lung tissue. As to human ILD samples, BALF of some patients contained high levels of GPX3, and GPX3 was upregulated in lung homogenates from IPF patients. GPX3 expression in primary human bronchial epithelial cells and lung fibroblasts was downregulated by TNF-α, but more variably regulated by TGF-β1 and menadione. In conclusion, the antioxidant enzyme GPX3 localizes to lung ECM and is variably upregulated in ILD.
Collapse
|
40
|
Di Paola R, Impellizzeri D, Fusco R, Cordaro M, Siracusa R, Crupi R, Esposito E, Cuzzocrea S. Ultramicronized palmitoylethanolamide (PEA-um(®)) in the treatment of idiopathic pulmonary fibrosis. Pharmacol Res 2016; 111:405-412. [PMID: 27402190 DOI: 10.1016/j.phrs.2016.07.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 07/06/2016] [Accepted: 07/07/2016] [Indexed: 12/15/2022]
Abstract
Pulmonary fibrosis is a chronic condition characterized by progressive scarring of lung parenchyma. The aim of this study was to examine the effects of an ultramicronized preparation of palmitoylethanolamide (PEA-um(®)), an endogenous fatty acid amide, in mice subjected to idiopathic pulmonary fibrosis. Idiopathic pulmonary fibrosis was induced in male mice by a single intratracheal administration of saline with bleomycin sulphate (1mg/kg body weight) in a volume of 100μL. PEA-um(®) was injected intraperitoneally at 1, 3 or 10mg/kg 1h after bleomycin instillation and daily thereafter. Animals were sacrificed after 7 and 21days by pentobarbitone overdose. One cohort of mice was sacrificed after seven days of bleomycin administration, followed by bronchoalveloar lavage and determination of myeloperoxidase activity, lung edema and histopathology features. In the 21-day cohort, mortality was assessed daily, and surviving mice were sacrificed followed by the above analyses together with immunohistochemical localization of CD8, tumor necrosis factor-α, CD4, interleukin-1β, transforming growth factor-β, inducible nitric oxide synthase and basic fibroblast growth factor. Compared to bleomycin-treated mice, animals that received also PEA-um(®) (3 or 10mg/kg) had significantly decreased weight loss, mortality, inflammation, lung damage at the histological level, and lung fibrosis at 7 and 21days. PEA-um(®) (1mg/kg) did not significantly inhibit the inflammation response and lung fibrosis. This study demonstrates that PEA-um(®) (3 and 10mg/kg) reduces the extent of lung inflammation in a mouse model of idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Rosanna Di Paola
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Italy
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Italy
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Italy
| | - Marika Cordaro
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Italy
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Italy
| | - Rosalia Crupi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Italy; Department of Pharmacological and Physiological Science, Saint Louis University, Saint Louis, MO, USA.
| |
Collapse
|
41
|
Kurundkar A, Thannickal VJ. Redox mechanisms in age-related lung fibrosis. Redox Biol 2016; 9:67-76. [PMID: 27394680 PMCID: PMC4943089 DOI: 10.1016/j.redox.2016.06.005] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 06/24/2016] [Accepted: 06/24/2016] [Indexed: 12/12/2022] Open
Abstract
Redox signaling and oxidative stress are associated with tissue fibrosis and aging. Aging is recognized as a major risk factor for fibrotic diseases involving multiple organ systems, including that of the lung. A number of oxidant generating enzymes are upregulated while antioxidant defenses are deficient with aging and cellular senescence, leading to redox imbalance and oxidative stress. However, the precise mechanisms by which redox signaling and oxidative stress contribute to the pathogenesis of lung fibrosis are not well understood. Tissue repair is a highly regulated process that involves the interactions of several cell types, including epithelial cells, fibroblasts and inflammatory cells. Fibrosis may develop when these interactions are dysregulated with the acquisition of pro-fibrotic cellular phenotypes. In this review, we explore the roles of redox mechanisms that promote and perpetuate fibrosis in the context of cellular senescence and aging.
Collapse
Affiliation(s)
- Ashish Kurundkar
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Victor J Thannickal
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
42
|
Zhou X, Gao T, Jiang XG, Xie ML. Protective effect of apigenin on bleomycin-induced pulmonary fibrosis in mice by increments of lung antioxidant ability and PPARγ expression. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.04.039] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
43
|
Papaioannou AI, Kostikas K, Manali ED, Papadaki G, Roussou A, Kolilekas L, Borie R, Bouros D, Papiris SA. Combined pulmonary fibrosis and emphysema: The many aspects of a cohabitation contract. Respir Med 2016; 117:14-26. [PMID: 27492509 DOI: 10.1016/j.rmed.2016.05.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Revised: 03/20/2016] [Accepted: 05/05/2016] [Indexed: 12/12/2022]
Abstract
Combined pulmonary fibrosis and emphysema (CPFE) is a clinical entity characterized by the coexistence of upper lobe emphysema and lower lobe fibrosis. Patients with this condition experience severe dyspnea and impaired gas exchange with preserved lung volumes. The diagnosis of the CPFE syndrome is based on HRCT imaging, showing the coexistence of emphysema and pulmonary fibrosis both in varying extent and locations within the lung parenchyma. Individual genetic background seem to predispose to the development of the disease. The risk of the development of pulmonary hypertension in patients with CPFE is high and related to poor prognosis. CPFE patients also present a high risk of lung cancer. Mortality is significant in patients with CPFE and median survival is reported between 2.1 and 8.5 years. Currently, no specific recommendations are available regarding the management of patients with CPFE. In this review we provide information on the existing knowledge on CPFE regarding the pathophysiology, clinical manifestations, imaging, complications, possible therapeutic interventions and prognosis of the disease.
Collapse
Affiliation(s)
- Andriana I Papaioannou
- 2nd Respiratory Medicine Department, "Attikon" University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Greece.
| | - Konstantinos Kostikas
- 2nd Respiratory Medicine Department, "Attikon" University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Greece.
| | - Effrosyni D Manali
- 2nd Respiratory Medicine Department, "Attikon" University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Greece.
| | - Georgia Papadaki
- 2nd Respiratory Medicine Department, "Attikon" University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Greece.
| | - Aneza Roussou
- 2nd Respiratory Medicine Department, "Attikon" University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Greece.
| | - Likurgos Kolilekas
- 7th Department of Pneumonology, "Sotiria" Chest Diseases Hospital, Athens, Greece.
| | - Raphaël Borie
- APHP, Hôpital Bichat, DHU FIRE Service de Pneumologie A, Centre de compétence des maladies pulmonaires rares, INSERM, Unité 1152, Université Paris Diderot, Paris, France.
| | - Demosthenis Bouros
- 1st Respiratory Medicine Department, "Sotiria" Chest Diseases Hospital, Athens, Medical School, National and Kapodistrian University of Athens, Greece.
| | - Spyridon A Papiris
- 2nd Respiratory Medicine Department, "Attikon" University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Greece.
| |
Collapse
|
44
|
Divya T, Dineshbabu V, Soumyakrishnan S, Sureshkumar A, Sudhandiran G. Celastrol enhances Nrf2 mediated antioxidant enzymes and exhibits anti-fibrotic effect through regulation of collagen production against bleomycin-induced pulmonary fibrosis. Chem Biol Interact 2016; 246:52-62. [PMID: 26768587 DOI: 10.1016/j.cbi.2016.01.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 12/01/2015] [Accepted: 01/04/2016] [Indexed: 01/14/2023]
Abstract
Pulmonary fibrosis (PF) is characterized by excessive accumulation of extracellular matrix components in the alveolar region which distorts the normal lung architecture and impairs the respiratory function. The aim of this study is to evaluate the anti-fibrotic effect of celastrol, a quinine-methide tri-terpenoid mainly found in Thunder God Vine root extracts against bleomycin (BLM)-induced PF through the enhancement of antioxidant defense system. A single intratracheal instillation of BLM (3 U/kg.bw) was administered in rats to induce PF. Celastrol (5 mg/kg) was given intraperitoneally, twice a week for a period of 28 days. BLM-induced rats exhibits declined activities of enzymatic and non-enzymatic antioxidants which were restored upon treatment with celastrol. BLM-induced rats show increased total and differential cell counts as compared to control and celastrol treated rats. Histopathological analysis shows increased inflammation and alveolar damage; while assay of hydroxyproline and Masson's trichrome staining shows an increased collagen deposition in BLM-challenged rats that were decreased upon celastrol treatment. Celastrol also reduces inflammation in BLM-induced rats as evidenced by decrease in the expressions of mast cells, Tumor necrosis factor-alpha (TNF- α) and matrix metalloproteinases (MMPs) 2 and 9. Further, Western blot analysis shows that celastrol is a potent inducer of NF-E2-related factor 2 (Nrf2) and it restores the activities of Phase II enzymes such as hemoxygenase-1 (HO-1), glutathione-S-transferase (GSTs) and NADP(H): quinine oxidoreductase (NQO1) which were declined upon BLM administration. The results of this study show evidence on the protective effect of celastrol against BLM-induced PF through its antioxidant and anti-fibrotic effects.
Collapse
Affiliation(s)
- Thomas Divya
- Cell Biology Laboratory, Department of Biochemistry, University of Madras, Guindy Campus, Chennai 600 025, India
| | - Vadivel Dineshbabu
- Cell Biology Laboratory, Department of Biochemistry, University of Madras, Guindy Campus, Chennai 600 025, India
| | - Syamala Soumyakrishnan
- Cell Biology Laboratory, Department of Biochemistry, University of Madras, Guindy Campus, Chennai 600 025, India
| | | | - Ganapasam Sudhandiran
- Cell Biology Laboratory, Department of Biochemistry, University of Madras, Guindy Campus, Chennai 600 025, India.
| |
Collapse
|
45
|
Lung extracellular matrix and redox regulation. Redox Biol 2016; 8:305-15. [PMID: 26938939 PMCID: PMC4777985 DOI: 10.1016/j.redox.2016.02.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 02/15/2016] [Accepted: 02/17/2016] [Indexed: 12/28/2022] Open
Abstract
Pulmonary fibrosis affects millions worldwide and, even though there has been a significant investment in understanding the processes involved in wound healing and maladaptive repair, a complete understanding of the mechanisms responsible for lung fibrogenesis eludes us, and interventions capable of reversing or halting disease progression are not available. Pulmonary fibrosis is characterized by the excessive expression and uncontrolled deposition of extracellular matrix (ECM) proteins resulting in erosion of the tissue structure. Initially considered an 'end-stage' process elicited after injury, these events are now considered pathogenic and are believed to contribute to the course of the disease. By interacting with integrins capable of signal transduction and by influencing tissue mechanics, ECM proteins modulate processes ranging from cell adhesion and migration to differentiation and growth factor expression. In doing so, ECM proteins help orchestrate complex developmental processes and maintain tissue homeostasis. However, poorly controlled deposition of ECM proteins promotes inflammation, fibroproliferation, and aberrant differentiation of cells, and has been implicated in the pathogenesis of pulmonary fibrosis, atherosclerosis and cancer. Considering their vital functions, ECM proteins are the target of investigation, and oxidation-reduction (redox) reactions have emerged as important regulators of the ECM. Oxidative stress invariably accompanies lung disease and promotes ECM expression directly or through the overproduction of pro-fibrotic growth factors, while affecting integrin binding and activation. In vitro and in vivo investigations point to redox reactions as targets for intervention in pulmonary fibrosis and related disorders, but studies in humans have been disappointing probably due to the narrow impact of the interventions tested, and our poor understanding of the factors that regulate these complex reactions. This review is not meant to provide a comprehensive review of this field, but rather to highlight what has been learned and to raise interest in this area in need of much attention.
Collapse
|
46
|
Panagiotou M, Polychronopoulos V, Strange C. Respiratory and lower limb muscle function in interstitial lung disease. Chron Respir Dis 2016; 13:162-72. [PMID: 26768011 DOI: 10.1177/1479972315626014] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Growing evidence suggests that respiratory and limb muscle function may be impaired in patients with interstitial lung disease (ILD). Importantly, muscle dysfunction could promote dyspnoea, fatigue and functional limitation all of which are cardinal features of ILD. This article examines the risk factors for skeletal muscle dysfunction in ILD, reviews the current evidence on overall respiratory and limb muscle function and focuses on the occurrence and implications of skeletal muscle dysfunction in ILD. Research limitations and pathways to address the current knowledge gaps are highlighted.
Collapse
Affiliation(s)
- Marios Panagiotou
- Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, UK
| | | | - Charlie Strange
- Division of Pulmonary and Critical Care Medicine, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
47
|
Liu RM, Desai LP. Reciprocal regulation of TGF-β and reactive oxygen species: A perverse cycle for fibrosis. Redox Biol 2015; 6:565-577. [PMID: 26496488 PMCID: PMC4625010 DOI: 10.1016/j.redox.2015.09.009] [Citation(s) in RCA: 456] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 09/17/2015] [Accepted: 09/20/2015] [Indexed: 12/21/2022] Open
Abstract
Transforming growth factor beta (TGF-β) is the most potent pro-fibrogenic cytokine and its expression is increased in almost all of fibrotic diseases. Although signaling through Smad pathway is believed to play a central role in TGF-β's fibrogenesis, emerging evidence indicates that reactive oxygen species (ROS) modulate TGF-β's signaling through different pathways including Smad pathway. TGF-β1 increases ROS production and suppresses antioxidant enzymes, leading to a redox imbalance. ROS, in turn, induce/activate TGF-β1 and mediate many of TGF-β's fibrogenic effects, forming a vicious cycle (see graphic flow chart on the right). Here, we review the current knowledge on the feed-forward mechanisms between TGF-β1 and ROS in the development of fibrosis. Therapeutics targeting TGF-β-induced and ROS-dependent cellular signaling represents a novel approach in the treatment of fibrotic disorders. TGF-β1 is the most potent ubiquitous profibrogenic cytokine. TGF- β 1 induces redox imbalance by ↑ ROS production and ↓ anti-oxidant defense system Redox imbalance, in turn, activates latent TGF-β1 and induces TGF-β1 expression. Redox imbalance also mediates many of TGF-β1’s profibrogenic effects
Collapse
Affiliation(s)
- Rui-Ming Liu
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmi ngham, Birmingham, AL, USA.
| | - Leena P Desai
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmi ngham, Birmingham, AL, USA
| |
Collapse
|
48
|
Osborn-Heaford HL, Murthy S, Gu L, Larson-Casey JL, Ryan AJ, Shi L, Glogauer M, Neighbors JD, Hohl R, Carter AB. Targeting the isoprenoid pathway to abrogate progression of pulmonary fibrosis. Free Radic Biol Med 2015; 86:47-56. [PMID: 25958207 PMCID: PMC4554879 DOI: 10.1016/j.freeradbiomed.2015.04.031] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 04/07/2015] [Accepted: 04/27/2015] [Indexed: 11/22/2022]
Abstract
Fibrotic remodeling in lung injury is a major cause of morbidity. The mechanism that mediates the ongoing fibrosis is unclear, and there is no available treatment to abate the aberrant repair. Reactive oxygen species (ROS) have a critical role in inducing fibrosis by modulating extracellular matrix deposition. Specifically, mitochondrial hydrogen peroxide (H2O2) production by alveolar macrophages is directly linked to pulmonary fibrosis as inhibition of mitochondrial H2O2 attenuates the fibrotic response in mice. Prior studies indicate that the small GTP-binding protein, Rac1, directly mediates H2O2 generation in the mitochondrial intermembrane space. Geranylgeranylation of the C-terminal cysteine residue (Cys(189)) is required for Rac1 activation and mitochondrial import. We hypothesized that impairment of geranylgeranylation would limit mitochondrial oxidative stress and, thus, abrogate progression of pulmonary fibrosis. By targeting the isoprenoid pathway with a novel agent, digeranyl bisphosphonate (DGBP), which impairs geranylgeranylation, we demonstrate that Rac1 mitochondrial import, mitochondrial oxidative stress, and progression of the fibrotic response to lung injury are significantly attenuated. These observations reveal that targeting the isoprenoid pathway to alter Rac1 geranylgeranylation halts the progression of pulmonary fibrosis after lung injury.
Collapse
Affiliation(s)
| | | | - Linlin Gu
- Deparment of Medicine, University of Alabama at Birmingham, AL
| | - Jennifer L Larson-Casey
- Free Radical and Radiation Biology Program, University of Iowa
- Deparment of Medicine, University of Alabama at Birmingham, AL
| | - Alan J Ryan
- Department of Internal Medicine, University of Iowa
| | - Lei Shi
- Human Toxicology Program, University of Iowa
| | - Michael Glogauer
- Canadian Institutes of Health Research Group in Matrix Dynamics, University of Toronto, Toronto, Ontario, Canada
| | | | - Raymond Hohl
- Department of Internal Medicine, University of Iowa
- Department of Pharmacology, University of Iowa
| | - A Brent Carter
- Department of Internal Medicine, University of Iowa
- Free Radical and Radiation Biology Program, University of Iowa
- Human Toxicology Program, University of Iowa
- Deparment of Medicine, University of Alabama at Birmingham, AL
- Iowa City VA Healthcare System, Iowa City, IA
- Birmingham VAMC, Birmingham, AL
| |
Collapse
|
49
|
Murray MA, Chotirmall SH. The Impact of Immunosenescence on Pulmonary Disease. Mediators Inflamm 2015; 2015:692546. [PMID: 26199462 PMCID: PMC4495178 DOI: 10.1155/2015/692546] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 06/09/2015] [Indexed: 01/09/2023] Open
Abstract
The global population is aging with significant gains in life expectancy particularly in the developed world. Consequently, greater focus on understanding the processes that underlie physiological aging has occurred. Key facets of advancing age include genomic instability, telomere shortening, epigenetic changes, and declines in immune function termed immunosenescence. Immunosenescence and its associated chronic low grade systemic "inflamm-aging" contribute to the development and progression of pulmonary disease in older individuals. These physiological processes predispose to pulmonary infection and confer specific and unique clinical phenotypes observed in chronic respiratory disease including late-onset asthma, chronic obstructive pulmonary disease, and pulmonary fibrosis. Emerging concepts of the gut and airway microbiome further complicate the interrelationship between host and microorganism particularly from an immunological perspective and especially so in the setting of immunosenescence. This review focuses on our current understanding of the aging process, immunosenescence, and how it can potentially impact on various pulmonary diseases and the human microbiome.
Collapse
Affiliation(s)
- Michelle A. Murray
- Department of Respiratory Medicine, Mater Misericordiae Hospital, Eccles Street, Dublin 7, Ireland
| | - Sanjay H. Chotirmall
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232
| |
Collapse
|
50
|
Matsuzawa Y, Kawashima T, Kuwabara R, Hayakawa S, Irie T, Yoshida T, Rikitake H, Wakabayashi T, Okada N, Kawashima K, Suzuki Y, Shirai K. Change in serum marker of oxidative stress in the progression of idiopathic pulmonary fibrosis. Pulm Pharmacol Ther 2015; 32:1-6. [PMID: 25862941 DOI: 10.1016/j.pupt.2015.03.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 02/26/2015] [Accepted: 03/29/2015] [Indexed: 11/15/2022]
Abstract
BACKGROUND Increased oxidative stress is supposed to be involved in the etiology of idiopathic pulmonary fibrosis (IPF). It was reported that oxidative stress values measured by a spectrophotometric technique (d-ROMs test) were significantly higher in IPF patients than in controls, and were negatively correlated with Forced Vital Capacity (FVC) and Carbon Monoxide Diffusing Capacity (DLCO). However, the relationship between progression of IPF over time and change in serum oxidative stress marker remains unclarified. AIMS This study aimed to investigate the change in serum oxidative stress marker during progression of IPF. SUBJECTS AND METHODS The levels of oxidative stress in blood samples of 43 treatment-naïve IPF patients were measured by the d-ROMs test. FVC and DLCO were measured concurrently. The changes in oxidative stress and pulmonary function were evaluated in 27 untreated patients 6 months later. Oxidative stress levels of 13 patients with acute exacerbation of IPF (AE-IPF) and 30 healthy controls were also evaluated. RESULTS Oxidative stress values [median, interquartile range (IQR); Carratelli units (U.CARR)] were significantly higher in 43 IPF patients than in controls (366, 339-443 vs. 289, 257-329, p < 0.01) and were significantly increased 6 months later in 27 untreated patients (353, 311-398 at baseline to 385, 345-417 at follow-up, p < 0.01). The increase in oxidative stress values (24.0, 6.0-49.0 U.CARR/6 months) was negatively correlated with baseline DLCO (rs = -0.44, p < 0.05) and FVC changes after 6 months (rs = -0.54, p < 0.01). Oxidative stress values were significantly higher in IPF patients with acute exacerbation than in those with stable disease (587, 523-667 vs. 366, 339-443 U.CARR, respectively; p < 0.01). CONCLUSIONS Serum oxidative stress values increased with disease progression in IPF patients.
Collapse
Affiliation(s)
- Yasuo Matsuzawa
- Department of Internal Medicine, Toho University Medical Center, Sakura Hospital, Japan.
| | - Tatsuo Kawashima
- Department of Internal Medicine, Toho University Medical Center, Sakura Hospital, Japan
| | - Ryosei Kuwabara
- Department of Internal Medicine, Toho University Medical Center, Sakura Hospital, Japan
| | - Sho Hayakawa
- Department of Internal Medicine, Toho University Medical Center, Sakura Hospital, Japan
| | - Tamako Irie
- Department of Internal Medicine, Toho University Medical Center, Sakura Hospital, Japan
| | - Tadashi Yoshida
- Department of Internal Medicine, Toho University Medical Center, Sakura Hospital, Japan
| | - Hagino Rikitake
- Department of Internal Medicine, Toho University Medical Center, Sakura Hospital, Japan
| | - Toru Wakabayashi
- Department of Internal Medicine, Toho University Medical Center, Sakura Hospital, Japan
| | - Noriaki Okada
- Department of Internal Medicine, Toho University Medical Center, Sakura Hospital, Japan
| | - Kengo Kawashima
- Department of Internal Medicine, Toho University Medical Center, Sakura Hospital, Japan
| | - Yasuo Suzuki
- Department of Internal Medicine, Toho University Medical Center, Sakura Hospital, Japan
| | - Kohji Shirai
- Department of Internal Medicine, Toho University Medical Center, Sakura Hospital, Japan
| |
Collapse
|